Network


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

Hotspot


Dive into the research topics where Anna Barker is active.

Publication


Featured researches published by Anna Barker.


Clinical Pharmacology & Therapeutics | 2009

I‐SPY 2: An Adaptive Breast Cancer Trial Design in the Setting of Neoadjuvant Chemotherapy

Anna Barker; Cc Sigman; Gary J. Kelloff; Nola M. Hylton; Da Berry; Laura Esserman

I‐SPY 2 (investigation of serial studies to predict your therapeutic response with imaging and molecular analysis 2) is a process targeting the rapid, focused clinical development of paired oncologic therapies and biomarkers. The framework is an adaptive phase II clinical trial design in the neoadjuvant setting for women with locally advanced breast cancer. I‐SPY 2 is a collaborative effort among academic investigators, the National Cancer Institute, the US Food and Drug Administration, and the pharmaceutical and biotechnology industries under the auspices of the Foundation for the National Institutes of Health Biomarkers Consortium.


Clinical Cancer Research | 2013

AACR Cancer Progress Report 2013

Charles L. Sawyers; Cory Abate-Shen; Kenneth C. Anderson; Anna Barker; José Baselga; Nathan A. Berger; Margaret Foti; Ahmedin Jemal; Theodore S. Lawrence; Christopher I. Li; Elaine R. Mardis; Peter J. Neumann; Drew M. Pardoll; George C. Prendergast; John C. Reed; George J. Weiner

### AACR Staffnn![Figure][1] nnOn April 8, 2013, in an unprecedented effort to highlight the critical importance of biomedical research, the American Association for Cancer Research (AACR) joined with more than 200 organizations representing a broad spectrum of research interests and diseases


Lancet Oncology | 2017

Future cancer research priorities in the USA: a Lancet Oncology Commission

Elizabeth M. Jaffee; Chi Van Dang; David B. Agus; Brian M. Alexander; Kenneth C. Anderson; Alan Ashworth; Anna Barker; Roshan Bastani; Sangeeta N. Bhatia; Jeffrey A. Bluestone; Otis W. Brawley; Atul J. Butte; Daniel G. Coit; Nancy E. Davidson; Mark E. Davis; Ronald A. DePinho; Robert B. Diasio; Giulio Draetta; A. Lindsay Frazier; Andrew Futreal; S. S. Gambhir; Patricia A. Ganz; Levi A. Garraway; Stanton L. Gerson; Sumit Gupta; James R. Heath; Ruth I. Hoffman; C. Hudis; Chanita Hughes-Halbert; Ramy Ibrahim

We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US


Clinical Cancer Research | 2014

Turning the Tide Against Cancer Through Sustained Medical Innovation: The Pathway to Progress

Amy P. Abernethy; Edward Abrahams; Anna Barker; Kenneth Buetow; Randy Burkholder; William S. Dalton; Margaret Foti; Felix W. Frueh; Richard B. Gaynor; Marcia A. Kean; Zeba M. Khan; Tracy Lessor; J. Leonard Lichtenfeld; John Mendelsohn; Laura J. van 't Veer

2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.


Expert Review of Molecular Diagnostics | 2015

The national biomarker development alliance: confronting the poor productivity of biomarker research and development

George Poste; Carolyn C. Compton; Anna Barker

An ever-expanding understanding of the molecular basis of the more than 200 unique diseases collectively called cancer, combined with efforts to apply these insights to clinical care, is forming the foundation of an era of personalized medicine that promises to improve cancer treatment. At the same time, these extraordinary opportunities are occurring in an environment of intense pressure to contain rising healthcare costs. This environment presents a challenge to oncology research and clinical care, because both are becoming progressively more complex and expensive, and because the current tools to measure the cost and value of advances in care (e.g., comparative effectiveness research, cost-effectiveness analysis, and health technology assessments) are not optimized for an ecosystem moving toward personalized, patient-centered care. Reconciling this tension will be essential to maintaining progress in a cost-constrained environment, especially because emerging innovations in science (e.g., increasing identification of molecular biomarkers) and in clinical process (implementation of a learning healthcare system) hold potential to dramatically improve patient care, and may ultimately help address the burden of rising costs. For example, the rapid pace of innovation taking place within oncology calls for increased capability to integrate clinical research and care to enable continuous learning, so that lessons learned from each patient treated can inform clinical decision making for the next patient. Recognizing the need to define the policies required for sustained innovation in cancer research and care in an era of cost containment, the stakeholder community must engage in an ongoing dialogue and identify areas for collaboration. This article reflects and seeks to amplify the ongoing robust discussion and diverse perspectives brought to this issue by multiple stakeholders within the cancer community, and to consider how to frame the research and regulatory policies necessary to sustain progress against cancer in an environment of constrained resources. Clin Cancer Res; 20(5); 1081–6. ©2014 AACR.


Biomarkers in Medicine | 2014

The National Biomarker Development Alliance accelerating the translation of biomarkers to the clinic

Anna Barker; Carolyn C. Compton; George Poste

Making precision (personalized) medicine a routine clinical reality will require a comprehensive inventory of validated biomarkers and molecular diagnostic tests to stratify disease subtypes and improve accuracy in diagnosis and treatment selection. Realization of this promise has been hindered by the poor productivity of biomarker identification and validation. This situation reflects deficiencies that are pervasive across the entire spectrum of biomarker R&D, from discovery to clinical validation and in the failure of regulatory and reimbursement policies to accommodate new classes of biomarkers. The launch of the National Biomarker Development Alliance is the culmination of a 2-year review and consultation process involving diverse stakeholders to advance standards, best practices and guidelines to enhance biomarker discovery and validation by adoption of systems-based approaches and trans-sector collaboration between academia, clinical medicine and relevant private and public sector stakeholders.


Clinical Cancer Research | 2017

Adaptive Global Innovative Learning Environment for Glioblastoma: GBM AGILE

Brian M. Alexander; Sujuan Ba; Mitchel S. Berger; Donald A. Berry; Webster K. Cavenee; Susan M. Chang; Timothy F. Cloughesy; Tao Jiang; Mustafa Khasraw; Wenbin Li; Robert Mittman; George Poste; Patrick Y. Wen; W. K. Alfred Yung; Anna Barker

Biomarker development is rarely successful Realizing the much vaunted promise of precision (personalized) medicine is linked inextricably to the availability of robust and clinically relevant biomarkers. Given their importance in supporting more informed treatment decisions, enabling earlier diagnosis and driving the development of molecular based therapies, biomarkers arguably represent the greatest potential value in biomedicine today. In short, biomarkers are the ‘holy grail’ of precision medicine. The BCR–ABL and HER-2 proteins are prototypic examples of the clinical utility of biomarkers in cancer that have transformed the classification of lymphocytic leukemia and HER-2 positive breast cancer, respectively, and driven more rational treatment decisions. Unfortunately, these remain rare examples. The potential of biomarkers to revolutionize the detection, diagnosis and clinical management of cancer remains largely unrealized, with fewer than 100 biomarkers approved by regulatory agencies as companion diagnostics for molecular profiling to guide treatment selection [1]. There is a staggering asymmetry between the several 100,000 publications describing candidate biomarkers and the mere fraction of 1% of these that ever progress to clinical practice [2]. The systematic failure in biomarker R&D is further illustrated by the fact that the US FDA has approved less than one protein biomarker per year since the mid 1990s [3]. The burden of these failures, which come at incalculable cost in wasted investment, intellectual resources and patient samples, are incompatible with charting a reliable intellectual foundation for molecular medicine and improved patient care.


Nature Communications | 2018

Poly-ligand profiling differentiates trastuzumab-treated breast cancer patients according to their outcomes

Valeriy Domenyuk; Zoran Gatalica; Radhika Santhanam; Xixi Wei; Adam Stark; Patrick Kennedy; Brandon Toussaint; Symon Levenberg; Jie Wang; Nianqing Xiao; Richard Greil; Gabriel Rinnerthaler; Simon Peter Gampenrieder; Amy B. Heimberger; Donald A. Berry; Anna Barker; John Quackenbush; John L. Marshall; George Poste; Jeffrey L. Vacirca; Gregory A. Vidal; Lee S. Schwartzberg; David D. Halbert; Andreas Voss; Daniel Magee; Mark Robert Miglarese; Michael Famulok; Günter Mayer; David Spetzler

Glioblastoma (GBM) is a deadly disease with few effective therapies. Although much has been learned about the molecular characteristics of the disease, this knowledge has not been translated into clinical improvements for patients. At the same time, many new therapies are being developed. Many of these therapies have potential biomarkers to identify responders. The result is an enormous amount of testable clinical questions that must be answered efficiently. The GBM Adaptive Global Innovative Learning Environment (GBM AGILE) is a novel, multi-arm, platform trial designed to address these challenges. It is the result of the collective work of over 130 oncologists, statisticians, pathologists, neurosurgeons, imagers, and translational and basic scientists from around the world. GBM AGILE is composed of two stages. The first stage is a Bayesian adaptively randomized screening stage to identify effective therapies based on impact on overall survival compared with a common control. This stage also finds the population in which the therapy shows the most promise based on clinical indication and biomarker status. Highly effective therapies transition in an inferentially seamless manner in the identified population to a second confirmatory stage. The second stage uses fixed randomization to confirm the findings from the first stage to support registration. Therapeutic arms with biomarkers may be added to the trial over time, while others complete testing. The design of GBM AGILE enables rapid clinical testing of new therapies and biomarkers to speed highly effective therapies to clinical practice. Clin Cancer Res; 24(4); 737–43. ©2017 AACR.


Cancer Research | 2017

Abstract 3594: Adaptively randomized seamless-phase multiarm platform trial: Glioblastoma Multiforme Adaptive Global Innovative Learning Environment (GBM AGILE)

Donald A. Berry; Todd Graves; Jason T. Connor; Brian M. Alexander; Timothy Cloughesy; Anna Barker; Scott M. Berry

Assessing the phenotypic diversity underlying tumour progression requires the identification of variations in the respective molecular interaction networks. Here we report proof-of-concept for a platform called poly-ligand profiling (PLP) that surveys these system states and distinguishes breast cancer patients who did or did not derive benefit from trastuzumab. We perform tissue-SELEX on breast cancer specimens to enrich single-stranded DNA (ssDNA) libraries that preferentially interact with molecular components associated with the two clinical phenotypes. Testing of independent sample sets verifies the ability of PLP to classify trastuzumab-treated patients according to their clinical outcomes with ROC-AUC of 0.78. Standard HER2 testing of the same patients gives a ROC-AUC of 0.47. Kaplan–Meier analysis reveals a median increase in benefit from trastuzumab-containing treatments of 300 days for PLP-positive compared to PLP-negative patients. If prospectively validated, PLP may increase success rates in precision oncology and clinical trials, thus improving both patient care and drug development.Patients’ selection is particularly important in cancer treatment. Here the authors present a proof-of-principle methodology that could be potentially important in assisting therapeutic decisions in the treatment of breast cancer patients.


Journal of Clinical Oncology | 2012

Adaptive Trials in the Neoadjuvant Setting: A Model to Safely Tailor Care While Accelerating Drug Development

Douglas Yee; Tufia C. Haddad; Kathy S. Albain; Anna Barker; Christopher C. Benz; Judy C. Boughey; Meredith Buxton; Amy Jo Chien; Angela DeMichele; David M. Dilts; Anthony Elias; Paul Haluska; Michael Hogarth; Alan Hu; Nola Hytlon; Henry G. Kaplan; Gary G. Kelloff; Qamar J. Khan; Julie E. Lang; Brian Leyland-Jones; Minetta C. Liu; Rita Nanda; Donald W. Northfelt; Olufunmilayo I. Olopade; John W. Park; Barbara A. Parker; David R. Parkinson; Sonia Pearson-White; Jane Perlmutter; Lajos Pusztai

Traditional phase 3 clinical trials compare an experimental arm with control. They inefficiently use patients, time, and finances. Dramatic and rapid changes in biology makes such trials untenable. We describe an alternative drug development strategy that we are using in a particular setting, the trial GBM AGILE (Glioblastoma Multiforme Adaptive Global Innovative Learning Environment). The trial’s design employs many innovations. Some aspects are similar to those of I-SPY 2 (see 4 articles in July 7, 2016 NEJM) but GBM AGILE extends I-SPY 2 in many ways. (1) It is a Bayesian platform trial that simultaneously evaluates many treatment arms (including combinations) from many companies. (2) Arms are added to the trial at any time and leave when they have been evaluated, whether positively or negatively. (3) An arm’s sample size is adaptive and based on frequent analyses of the trial results. (4) Every arm has an initial stage in which it is randomized adaptively: arms performing better in disease subtypes are assigned with higher probability to such patients. (5) An arm that performs sufficiently well in a disease subset moves seamlessly into a small (50-patient) confirmatory, registration stage in the same subset, with equal randomization against control. (6) All experimental arms are compared against a common control arm that is assigned to 20% of patients in every subtype; a bridging model takes advantage of having many arms in the trial and many comparisons among arms, and enables indirect randomization comparisons of all arms with all controls. (7) Patient subtypes are defined by line of therapy, MGMT methylation status for newly diagnosed patients, and biomarkers associated with targeted therapies, although adaptive randomization enables us to draw conclusions about off-target effects. The many possible subtypes means that there are many possible drug indications. So there are many possible “error types” and no single definition of statistical power. For example, the trial may conclude that a drug’s indication is “recurrent, biomarker-positive” disease when in truth it is “all recurrent” disease. We show how the design addresses this issue and we define “pure type I error.” GBM AGILE’s primary endpoint is overall survival (OS). To make the design more efficient we incorporate evaluations of patients’ statuses over time using a longitudinal model based on periodic MRI assessments and performance status. The longitudinal model and its components are not end points but rather provide auxiliary information that enables multiply imputing OS for surviving patients. We represent the trial’s coordinating committees that are made up of more than 150 enthusiastic and devoted disease experts and advocates from around the globe, including from Australia and China. The U.S. FDA has been enormously helpful in designing GBM AGILE, especially as regards its potential for drug and biomarker registration. Our approach provides a model for other diseases, including those outside of cancer. Citation Format: Donald A. Berry, Todd Graves, Jason Connor, Brian Alexander, Timothy Cloughesy, Anna Barker, Scott M. Berry, for the GBM AGILE Global Alliance. Adaptively randomized seamless-phase multiarm platform trial: Glioblastoma Multiforme Adaptive Global Innovative Learning Environment (GBM AGILE) [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 3594. doi:10.1158/1538-7445.AM2017-3594

Collaboration


Dive into the Anna Barker's collaboration.

Top Co-Authors

Avatar

George Poste

Arizona State University

View shared research outputs
Top Co-Authors

Avatar

Donald A. Berry

University of Texas MD Anderson Cancer 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

Brian M. Alexander

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

David Spetzler

Arizona State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy B. Heimberger

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge