RESEARCH DOCUMENTS

Joan S. Brugge, Ph.D.

Brief Biography of Dr. Joan Brugge

Dr. Brugge is currently the Louise Foote Pfeiffer Professor of Cell Biology at HMS and co-director of the Ludwig Center at Harvard.  She joined the faculty of the Harvard Medical School as a professor in July 1997. A graduate of Northwestern University, she did her graduate work at the Baylor College of Medicine, completing her PhD in 1975. She then performed her postdoctoral training at the University of Colorado with Dr. Raymond Erikson. Dr. Brugge has held full professorships at the State University of New York, Stony Brook, and the University of Pennsylvania, where she was also named as an investigator at the Howard Hughes Medical Institute. From 1992-1997 Dr. Brugge was a founder and scientific director of the biotechnology company ARIAD. She then joined the faculty in 1997 as Professor of Cell Biology, and became Chair of Cell Biology in 2004 and Co-Director of the Ludwig Center at Harvard in 2014.

Joan Brugge has made significant contributions to our understanding of the mechanisms involved on tumorigenesis, initially through the first identification of the v-SRC oncoprotein and cellular processes that are regulated by it, and subsequently through critical investigations of critical steps in breast cancer initiation and progression. Her work has highlighted the how tumor cell evade natural processes that eliminate early stage tumor cells as they proliferate outside their natural niches, and the role of anti-oxidants in promoting survival of displaced tumor cells. More recently, she has brought new insights into cellular responses to cancer therapies that lead to therapy resistance and strategies to overcome resistance. Lastly, she has begun a new program focused on the diagnosis and prevention of cancer in women who carry BRCA1 and BRCA2 mutations that significantly increase the risk of breast and other cancers.

Dr. Brugge has received several awards recognizing her scientific accomplishments including American Cancer Society Medal of Honor,  the Science of Oncology Award from the Americal Society of Clinical Oncology, the Senior Career Recognition Award from the American Society of Cell Biology, and she has been elected to the American Academy of Arts and Sciences, the National Academy of Sciences and the National Academy of Medicine.

Brugge Laboratory Research Programs

Dr. Joan Brugge has had a long and distinguished research career studying the genes, proteins and molecular pathways that are involved in transforming normal cells into malignant cells. It is this type of fundamental knowledge that is now informing the newest and most innovative strategies and approaches for treating human cancers. The Brugge laboratory has produced and published data on some of the most important basic mechanisms involving the molecular changes brought about by these transforming proteins and pathways. While she and her laboratory group continues to conduct this type of research, she has also turned additional focus onto the mechanisms by which cancer cells develop resistance to what should otherwise be effective therapies in order to either initially resist or ultimately escape treatment responses. These “resistance” mechanisms can be either present at the outset of a cancer’s development, making initial treatments ineffective or alternatively, can be acquired after initial responses to therapy are observed. This phenomenon makes the resistance mechanisms crucial to the development of more effective treatment strategies. In a new and exciting area of research she is also tracking the evolution of breast tumors in women who are at high risk for tumor formation due to inherited mutations in the BRCA1 and BRCA2 genes.  This could lead to  transformative ways to diagnose and prevent breast cancer in these women. In summary, the Brugge laboratory is now among the leaders in translating this new basic knowledge into development of novel clinical treatment strategies for patients challenged with cancer.

Development of combination therapies to target breast and ovarian cancer.

The overarching goal of this project is to develop strategies to overcome the barriers that limit the efficacy of current and emerging cancer therapies and develop new therapies that escape resistance mechanisms.  While significant advances have been made in the treatment of cancer, therapy resistance, whether primary or acquired, remains a major challenge to reducing the cancer burden. The overall approach is to increase our understanding of the fundamental states of resistance and then to translate this understanding into more effective treatment strategies.

The reason that therapy resistance poses such a significant challenge is because there are many distinct mechanisms that protect tumor cells from cancer therapies. These include diverse genetic, epigenetic, and stochastic alterations within tumors, as well as a broad spectrum of factors contributed by many diverse cells that are recruited to the tumor microenvironment. This diversity of factors within tumor cells and the tumor microenvironment, as well as the plasticity of tumors which allows them to evolve rapidly, significantly complicates the rational choice of combination therapies to overcome resistance. In addition to this diversity of tumor cell-based resistance mechanisms, there are physical and chemical barriers to effective drug delivery (e.g. poor vascular integrity, interstitial pressure, acidity, and hypoxia).  Lastly, many factors interfere with effective immune cell surveillance to eliminate tumor cells.

Approach:  Our laboratory is defining the adaptive responses to treatment with cancer therapies which make tumor cells resistant to the drugs.  After defining the nature of the response, we then develop combinations therapies that abrogate these resistance mechanisms.  For example. we have found that anti-apoptotic proteins are critical mediators of resistance to chemotherapies and targeted therapies.  We have systematically and comprehensively explored the efficacy of treatment with inhibitors of the anti-apoptotic proteins in breast and ovarian cancer and have clearly demonstrated the enhanced efficacy of these drug combinations and defined markers that predict efficacy. In future studies, she is continuing with these studies and working with clinicians to  carry out key preclinical studies to move these treatments to the clinic and also working with surgeons and oncologists, to identify adaptive changes that occur in primary patient tumors follow treatment.

Muranen, T, Selfors LM, Worster DT, Iwanicki MP, Song L, Morales FC, Gao S, Mills GB and Brugge JS, Inhibition of PI3K/mTOR Leads to Adaptive Resistance in Matrix-Attached Cancer Cells. Cancer Cell, 2012 Feb 14;21(2):227-39. PMCID: PMC3297962

Muranen, T, Selfors LT, Julie Hwang, Gallegos LL, Coloff JL, Thoreen CC, Kang SA, Sabatini DM, Mills GB and Brugge JS  The Balance of ERK and p38 MAPK Activity Determines the Sensitivity to PI3K/mTOR Inhibition Through Regulation of MYC and YAP, Cancer Res 2016, 76(24):7168-7180.  PMCID: PMC5161652

Zervantonakis IK, Iavarone C, Chen H-Y, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis U, Leverson JD, Sampath D, Mills GB, Brugge JS Targeting apoptotic vulnerabilities in patient-derived ovarian cancer xenografts to enhance the efficacy of PI3K pathway inhibitors,  Nature Comm,;8(1):365. doi: 10.1038/s41467-017-00263-7 PMCID: PMC5573720

Iavarone C,  Zervantonakis IK, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis UA, Hallberg D, Velculescu VE, Leverson JD, Sampath D, Mills GB, Brugge JS. Combined MEK and BCL-2/XL inhibition is effective in high-grade serous ovarian cancer patient-derived xenograft models and BIM levels are predictive of responsiveness. Mol. Cancer Therapeutics 2019 Mar;18(3):642-655. PMID:30679390

Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers.Sun C, Fang Y, Yin J, Chen J, Ju Z, Zhang D, Chen X, Vellano CP, Jeong KJ, Ng PK, Eterovic AKB, Bhola NH, Lu Y, Westin SN, Grandis JR, Lin SY, Scott KL, Peng G, Brugge J, Mills GB. Sci Transl Med. 2017 May 31;9(392). pii: eaal5148. doi: 10.1126/scitranslmed.aal5148. PMID:28566428

Zoeller JJ, Bronson RT, Selfors LM,. Mills GB, Brugge JS. Niche-localized tumor cells are protected from HER2-targeted therapy via upregulation of an anti-apoptotic program in vivo, npj Breast Cancer 2017: 3, doi: 10.1038/s41523-017-0020-z

Zoeller JJ, Vagodny A, Taneja K, Tan BY, O’Brien N, Slamon DJ, Sampath D, Leverson JD,  Bronson RT, Dillon DA, Brugge JS, Neutralization of BCL-2/XL enhances the cytotoxicity of T-DM1 in vivo Mol. Can. Therapeutics, in press

Anti-oxidants as resistance mediators: In addition to anti-apoptotic proteins, another set of cellular proteins that protect tumors from drug treatment are proteins that neutralize reactive oxygen species – these are referred to as anti-oxidants.  While there has been a great deal of public interest in anti-oxidant dietary supplements as cancer prevention agents, we and others have strong evidence that anti-oxidants can promote tumor progression in early stages of tumorigenesis as well as therapy resistance. We have identified anti-oxidant mediators of tumor resistance and our future research is directed towards investigating how inhibitors of these mediators can enhance cancer therapy, as well as to refine our understanding of the mechanisms of action of these proteins.

Schafer ZT, Grassian AR, Song L, Jiang Z, Gerhart-Hines Z, Irie HY, Gao S, Puigserver P, Brugge JS. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature. 2009;461(7260):109-13.

Takahashi N, Chen H-Y,  Harris IS,  Stover D, Bronson RT, Takemura RT, Kitao A, Deraedt T, Cichowski K, Welm AL, Mori, Y, Mills GB, Brugge JS.  Cancer Cells Co-opt the Neuronal Redox-Sensing Channel TRPA1 to Promote Oxidative-Stress Tolerance. 2018. Cancer Cell  33: 6, p985-1003. https://doi.org/10.1016/j.ccell.2018.05.001

Isaac S. Harris, IS, Endress, JE, Coloff, JL, Selfors,LS,  Rosenbluth, JR, Takahashi N, McBrayer SK, Koduri B, Oser MG, Schauer NJ, Doherty LM, Hong AL, Younger ST, Doench JG, Hahn WC, Buhrlage SJ, Kaelin WG,  Brugge JS, Deubiquitinases maintain protein homeostasis and survival of cancer cells upon glutathione depletion. Cell Metabolism, 2019 Feb 13. pii: S1550-4131(19)30020-8. doi: 10.1016/j.cmet.2019.01.020. [Epub ahead of print]

Cho, P, Takahashi, N, Brugge, JS. Interplay between anti-oxidants NRF2 and GPX4 is critical for lung cancer cell survival., manuscript in preparation.

Tumor Heterogeneity: Cancer therapy resistance is further complicated by the heterogeneity within individual tumors, within tumors at different sites in the same patient, and between tumors from different individuals. Recent evidence has shown that the vast majority of cells in many tumors do not share a common set of mutations and that multiple branched genetic lineages exist within different regions of each tumor and at metastatic sites of the same tumor. This genetic heterogeneity generates diversity in responses to cancer therapies.  In addition to the genetic heterogeneity, tumor cells with the same genetic constitution can exist in different states that reflect their differentiated phenotype, their response to matrix and paracrine factors derived from cells within the microenvironment,

Approach: We are using multiple approaches to identify drug combinations that optimally target the diversity of cells within tumors.  The first step towards this goal is to define and understand the differential sensitivity of individual cells within tumors.  We have optimized three different single-cell technologies to profile single cells within tumors [mass cytometry (CyTOF), single cell RNA sequencing (scRNAseq), and multiplex immunofluorescence imaging )CyCIF).  To this end, we are analyzing patient samples from the clinic as well as patient-derived xenografts (PDX) models in order to profile cells within tumors that are most and least sensitive to drug. Computational analysis of the genes expressed in these cells will identify candidate pathways that are responsible for resistance and sensitivity.   These approaches will identify markers on the more resistant cells that then allow us to isolate these cells in order to test their sensitivity to drug combinations that are informed by this approach.  CyCIF analysis is in collaboration with Dr. Peter Sorger. We have not yet published on these studies but all of the techniques are optimized and working well on tumor samples.

Development of strategies to track and prevent cancer in women that inherited mutation in genes that significantly increase cancer risk

Germline mutations in BRCA1,BRCA2, and several other genes significantly increase the lifetime risk of developing breast cancer.  It has been difficult to track and prevent breast cancer in these mutation carriers because we do not yet understand how their cancers begin nor how they progress. In order to develop novel cancer prevention and early detection strategies for inherited mutation carriers, it is critical to (1) identify pre-malignant changes in breast tissue from high-risk women, (2) elucidate mechanisms involved in carcinogenesis, and (3) use this information to develop strategies to track and prevent cancer development.

Using transformative new single cell technologies (CyTOF, scRNAseq, and CyCIF), we have examined hundreds of thousands of individual cells from breast tissues of BRCA1/2 mutation carriers in order to identify the earliest changes in cells from ostensibly normal tissues, opening an unprecedented opportunity to track breast cancer development and develop strategies for cancer prevention. We have identified two specific subtypes of cells that accumulate significantly in breast tissues from BRCA1/2 carriers and are rare in breast tissues without BRCA1/2 mutations. Initial characterization of these cells provides intriguing clues as to why they accumulate, and how to prevent their accumulation. We are now carrying out studies to understand why BRCA mutations cause these cells to accumulate, and to expose vulnerabilities that could lead to new strategies for their elimination. By identifying specific markers for these cells, we will devise clinical approaches to track their accumulation in BRCA1/2 mutation carriers, in order to precisely time clinical interventions. This work could transform both our understanding of breast carcinogenesis in BRCA1/2 mutation carriers and our ability to predict and prevent it.

CURRICULUM VITAE

Name Joan Siefert Brugge

Department of Cell Biology
Harvard Medical School

Education
1971 Northwestern University, Evanston, IL
B.A. in Biology, with honors (minor in Chemistry)
1975 Baylor College of Medicine, Houston, TX
Ph.D. in Virology Adviser: Dr. Janet Butel

Professional Experience
1975-79 Postdoctoral Fellow
University of Colorado Medical Center
Denver, CO
Adviser: Dr. Raymond Erikson
7/79 – 6/84 Assistant Professor
Department of Microbiology
State University of New York at Stony Brook
Stony Brook, NY 11794
7/84 – 12/87 Associate Professor
Department of Microbiology
State University of New York at Stony Brook
Stony Brook, NY 11794
1/88 – 12/88 Professor
Department of Microbiology
State University of New York at Stony Brook
Stony Brook, NY 11794
1/89 – 5/92 Investigator
Howard Hughes Medical Institute
Professor
Department of Microbiology
University of Pennsylvania
5/92 – 9/96 Scientific Director
Senior Vice President, Research and Biology
ARIAD Pharmaceuticals, Inc.
10/96 – 7/97 Senior Vice President, Exploratory Research
ARIAD Pharmaceuticals, Inc.
7/97-present Professor
Harvard Medical School
Department of Cell Biology
7/03 – 07/04 Acting Chair
Harvard Medical School
Department of Cell Biology

7/04 – 12/14 Chair
Department of Cell Biology
Harvard Medical School

1/15- present Director Ludwig Center at Harvard

Fellowships
1977 – 1978 National Institutes of Health Postdoctoral Fellowship
1976 – 1977 American Cancer Society Postdoctoral Fellowship

Professional Awards

• American Society of Clinical Oncology (Science of Oncology Award) 2019
• American Cancer Society Medal of Honor Recipient (2016)
• Election as Inaugural American Society of Cell Biology Fellow (2016)
• The Hope Funds for Cancer Research Award of Excellence (2016)
• Memorial Sloan Kettering Medal for Outstanding Contributions to Biomedical Research (2016)
• Honorary Degree: Gerstner Sloan Kettering Graduate School of Biomedical Sciences (2016)
• Distinguished Woman Scientist Award (2016)
• Breast Cancer Research Foundation Jill Rose Memorial Award (2015)
• Brinker Award for Scientific Distinction (2014)
• Elected as Fellow of the Americal Association of Cancer Research Academy 2014)
• Elected as Fellow, Massachusetts Academy of Sciences (2013)
• AACR Distinguished Lectureship in Breast Cancer Research: (2011)
• Elected as Fellow, American Association of Science (2008)
• Honorary Ph.D., Northwestern University (2007)
• Charlotte Friend Memorial Lectureship Award, AACR (2005)
• NCI Rosalind Franklin Award (2005)
• Distinguished Alumnus Award – Baylor College of Medicine (2003)
• Faculty of Arts and Sciences Mentoring Award (2002)
• Election to the Institute of Medicine (2001)
• Election to the National Academy of Science (2001)
• Senior Career Recognition Award, ASCB Women in Cell Biology (2001)
• BBS Graduate Program Mentoring Award (2001)
• Arthur and Rochelle Belfer Foundation Award (2001)
• American Cancer Society Research Professorship (2001)
• Election to the American Academy of Arts and Sciences (2000)
• Ludwig Foundation Faculty Award (1998)
• American Cancer Society Faculty Research Award (1986)
• National Cancer Institute MERIT Award 2R37 CA 27951 (1986-1994)
• Florence M. Catacosinos Cancer Award (1982)

Honorary Lectureships

Keynote Lecture, ACS Postdoctoral Symposium
Keynote Lecture, American Society for Investigative Pathology, San Francisco
Brody Lectureship, University of Iowa School of Medicine
Staples Lectureship, University of Maine, Bangor, MA
Keynote Lecture, Brown University School of Medicine, Pathobiology Retreat, Providence, RI
Keynote Lecture, MIT Cell Decision Process Retreat
Kreb’s Lectureship, University of Washington, Seattle, WA
Department of Defense Era of Hope meeting Keynote lecturer, Philadelphia, PA
National Cancer Institute Rosalind Franklin Lectureship
University of Nebraska Eppley Cancer Center Grand Rounds
AACR Charlotte Friend Lectureship
Dean’s Distinguished Lecture, U-Colorado Health Sciences Center
Keynote Lecture: ASCB Cell Engineering Meeting, U. Washington, Seattle, WA
American Society for Investigative Pathology, Keynote lecture
University of Pennsylvania: Pharmacology Training Program Keynote Speaker
Baylor College of Medicine. Virology and Microbiology Dept Retreat Keynote Speaker
Women in Life Sciences Lectureship: University of California at San Francisco
Marine Biology Laboratory: Forkosh-Waxler Lecture in Physiology
Schmidt Distinguished Lectureship Tufts University School of Medicine: Sackler School of Graduate Biomedical Sciences Boston, MA
Kathleen Robison Huntsman Distinguished Lectureship, University of Utah, Salt Lake City, Utah
McCormick Distinguished Lectureship Stanford University, Palo Alto, CA
Massachusetts General Hospital Keynote Lecturer – Science Day
Steelman Lectureship, University of North Carolina, Chapel Hill, NC
Walter Rubin Memorial Keynote Lecturer –Discovery Day -Drexel University College of Medicine Philadelphia, PA
Keynote Lecture: FEBS workshop on Invadopodia, Podosomes and Focal Adhesion, Ciente, Italy
National Cancer Institute CCR Grand Rounds Lecture Series
University of Virginia Distinguished Lectureship 2008
Gordon Research Conference on Basement Membrane, Keynote lecture 2008
Irish Cancer Society Keynote Lectureship
Drexel University College of Medicine Walter Rubin Memorial Keynote Lecturer
Massachusetts General Hospital SAB “Celebration of Science” Keynote Lecture
IRCM Boehringer-Ingelheim Honorary Lectureship 2007, Montreal CA
U-Texas MD Anderson Cancer Center, Cancer Biology Program Retreat, Keynote Speaker
UCLA David Geffen School of Medicine Annual Lecturer 2008
Soma Weiss Student Research Days 2008, Harvard Medical School, Keynote Speaker
Molecular Targets and Cancer Therapeutice meeting, Keynote lecture, 2009
AACR Translation of the Cancer Genome Conference 2009
MD Anderson Retreat Keynote Lecturer 2009,
Damon Runyon Annual Fellows’ Retreat, Keynote speaker Cape Cod, Massachusetts
Keynote Lecture, Keystone Symposium Apoptosis, Autophagy, Necrosis, Metabolism and Cancer (X3) Joint meeting with Metabolism & Cancer
Invited Honorary Lecture Harvey Society Rockefeller University NYC, NY
Keynote Lecture Cold Spring Harbor Laboratory Mechanisms & Models of Cancer meeting
Keynote Lecture AACR/Metastasis Research Society joint meeting on the Tumor Micro environment
Invited Honorary Lecture University of Alabama School of Medicine
Keynote Lecture Gordon Conference Epithelial Differentiation and Keratinization
Keynote Lecture AACR Special Conference Advances in Breast Cancer Research
Honorary Lecture SABCS AACR Distinguished Lectureship in Breast Cancer Research, 2011
Mitchell Lectureship Queen’s College Belfast, Ireland
David. M. Kovitz Visiting professorship, University of Calgary, Canada 2012
Eminent Lecturer in Cancer Research, NIC, Bethesda MD 2012
Mary Bartlett Bunge Lectureship, Miami Miller School of Medicine, Miami Florida, 2012
Sokolow lecture University of California San Francisco School of Medicine
Postdoctoral Science Symposium: Collaboration in Science, Keynote lecturer, MD Anderson Cancer Center
Danny Brower Memorial Lectureship, University of Arizona, February 2013
Keynote speaker – Professional Advancement Session – AACR, April, 2013
Bard Lectureship, Johns Hopkins Medical Institute, May 2013
Keynote Speaker Society for Melanoma Research Keynote presentation Nov.2013
Keynote Plenary lecture Hunter Cell Biology meeting –, Australia April 2014
Robert Sinscheimer lectureship, University of California Santa Cruz May 2014
San Antonio Breast Cancer meeting – Brinker Award lecture presentation
Keystone Meeting on Metabolism: Keynote Lecture February 2015
Sterlicht Lecturer, Case Western Reserve Medical School, May, 2015
Honorary Degree Keynote commencement address: Gerstner Sloan Kettering Graduate School of Biomedical Sciences May 2016
University of Colorado dean’s distinguished lecturer February 2017
Helen Gurley Brown Symposium Presidential Initiative: Keynote lecture March 2017
Carolyn Kaelin Memorial Lecture, Aspen Cancer conference July 18th, 2017
FASEB meeting on Signaling in Cancer- Keynote Lecture, June 2018
Gordon Conference on Adhesion Signaling – Keynote Lecture, June 2018
CSHL Mechanisms and Models of Cancer, Keynote lecture, August 2018
MD Anderson Cancer Symposium, Keynote Lecture, October 2018
CSU Interdisciplinary Cancer Symposium, Keynote Lecture, November 2018
Lippard Lectureship, MIT and MGH, November 2018
SABCS, Cancer Metabolism Symposium December 2018

Professional Activities Outside Harvard Medical School

Scientific Advisory Boards:

1995-1997 Howard Hughes Medical Institute – Scientific Review Board
1995-2002 Howard Hughes Medical Institute – Medical Advisory Board
1997-1999 ARIAD Pharmaceuticals, Chairman, Scientific Advisory Board
1994-1998 Massachusetts General Hospital Cancer Center (Chairman, 1995)
1994-1997 Massachusetts Institute of Technology Cancer Center
1996-2000 National Cancer Institute, Board of Scientific Advisors
(Chairman, Subcommittee, Cancer Biology, Epidemiology, and Genetics)
1997-2000 Advisory Committee to the Director, National Cancer Institute
1998-2000 Division of Tumor Biology, Chairman, Task Force
2001-2004 Fox Chase Cancer Center Scientific Advisory Board
2001-2005 Alliance for Cellular Signaling, External Advisory Board, Chair
2001- 2013 Millennium Pharmaceuticals, Oncology Advisory Board
2002- 2005 Vertex Pharmaceuticals, Kinase Advisory Board
2003- 2004 Akceli Pharmceuticals, Scientific Advisory Board
2002-2013 Van Andel Cancer Institute Scientific Advisory Board
2001- 2006 DGAP Project Scientific Advisory Board
2004-2007 Merck Pharmaceuticals, consultant
2007-2014 Massachusetts General Hospital Scientific Advisory Committee
2009- Massachusetts Institute of Technology Visiting Committee, Biology Dept
2012- Agios Scientific Advisory Board
2012- Effector Therapeutics Scientific Advisory Board
2013-2016 Komen Scholar
2015- present Scientific Advisory Board Allen Institute of Cell Sciences

Other Professional Activities:

1994-2014 Foundation for Advanced Cancer Studies (Board of Directors)
1994-1999 Mass. Biotechnology Council member
1995-1999 Member, Board of Directors, ARIAD Pharmaceuticals, Inc.
1996-2000 American Society for Cell Biology, Education Committee and Subcommittee on Graduate Education
1997, 1998 American Society for Cell Biology, Program Committee
1997 American Society for Cell Biology, Nominating Committee for President and Council
2000-2003 American Society for Cell Biology Council Member
2001-2002 National Cancer Legislation Advisory Committee
2005-2006 AACR Annual Meeting Program Committee
2006- 2012 AACR Council of Scientific Advsiors
2007 AACR- Japan Cancer Society Joint meeting planning committee
2008 Testified before the Senate Appropriations Committee on Funding crisis
2008 Organizing Committee, San Antonio Breast Cancer Meeting
2008 Kennedy Hutchinson Cancer Bill Advisory group
2008 AACR/CTRC SABCS SCIENTIFIC PROGRAM PLANNING COMMIITTEE
2009 AACR Annual Meeting (Planning Committee)

Chairperson/Organizer:

1984 RNA Tumor Virus Meeting, Cold Spring Harbor Labs
1990 Cold Spring Harbor Centennial Symposium – “Origins of Human Cancer”
1991 VII Meeting on Oncogenes, Frederick, MD
2002- FASEB meeting on Protein Kinases (co-chair), 2001,(chairman)
2000 Chairman, Program Committee for 2001 ASCB Annual Meeting
2004 Gordon Research Conference on Adhesion Signaling, Co-chair Chair
2005 2006 Gordon Research Conference on Adhesion Signaling, Chair
2009 AACR International Conference on Frontiers in Basic Cancer Research (Conference Co-chair)
2010 AACR Annual Meeting (Tuesday Plenary Session Chairperson)
2008-2009 AACR Chair Nominating Committee
2011 AACR Annual Meeting (Program Committee Chairperson)
2013 AACR Frontiers in Cancer Organizing Committee

Editorial Boards
1989 – 1991 Senior Editor, Journal of Virology
1986 – 1992 Journal of Virology
1983 – 1989 Molecular and Cellular Biology
1983 – 1986 Virology
1986 – 1990 Oncogene Research
1986 – 1990 Book Review Advisor – Quarterly Review of Biology
1995 – present Associate Editor, Cell Adhesion and Communication
1994 – 2002 Chemistry & Biology
1994 – 1996 Current Drugs
1995 – 1999 Reviews on Cancer
1995 – 2002 Genes and Development
1995 – 2014 Genes to Cells
1993-2001 Associate Editor, Molecular Biology of Cell
2000-2010 Nature Reviews Molecular Cell Biology, Highlights editor for Cell Adhesion
2001-2010 Reviews Editor, Developmental Cell
2001-present Faculty of 1000, Editor for Cell Signaling
2002-2004 Journal Cell Biology, Editorial Board
2002- Cancer Cell Editorial Board
2003- Cell Editorial Board
2006- 2010 Mol Cell Biol Associate Editor
2012- present Cancer Discovery, a Scientific Editor

NIH/DOD Review Panels
Ad hoc Member:
1984 Molecular Biology
1986 Virology and Immunology Clinical Fellowships
1987 Ad hoc member Molecular Cytology study section
1988-1992 Permanent Member: Molecular Cytology Study section
2001 Site visit for NCI Program Project grant
2002 Review panel for Organotypic Models grant applications
2003 NIGMS Program Project grant review
2007 Chair, NIH Center for Scientific Review Special Emphasis Panel
(Cell Biology R01)
2016 BRCR Innovator Award review panel

Testimony to Senate Appropriations committee: 2007 http://www.c-spanvideo.org/program/Year2008Nat

Other Review/Award Selection Panels
1993-1994 Member, Life Sciences Research Foundation Review Board
1993-1994 Chairperson-CHIRON Corporation Biotechnology Research Award
Selection Committee
1994-1997 General Motors Cancer Research Foundation, General Assembly member
1990-1992 General Motors Cancer Research Foundation, Sloan Award Selection Committee (1991-2, Chairperson)
2001- National Academy of Science, Molecular Biology Award Selection committee
2001-2002 American Academy of Arts and Sciences, Membership Selection Committee (Chair 02)
2006 American Society for Cell Biology, Chair, Early Career Life Scientist Award Selection Committee
2011 SABCS-AACR Award Committee Chair –
2014 Komen Postdoctoral Grants review committee
2015 Komen Career Catalyst Review committee
2016 Komen Career Catalyst Review committee
2017 Komen Career Catalyst Review Committee

Other NIH/NCI/NAS Advisory Services
2003 Chair: Tumor Microenvironment Think tank
2003 NIH Study Panel Reorganization Committee
2003 Keck Foundation/NAS Future Initiatives Steering Committee
2004 Presentation to the National Cancer Advisory Board on the Tumor Microenvironment
2005 Presentation to the NCI Board of Scientific Advisors on the Tumor Microenvironment
2005-2006 Organized two workshops on Culturing Cells In 3D Basement Membrane Gels for 10 investigators from outside institutions
2007 NIH Directors Fostering Innovation Workshop
2009-2012 NIH Advisory Committee to the Director (ACD)
2018 NCI Tumor models workshop
2019 NCI Tumor microenvironment network conference

Contract Reviews
1985 Use of Molecular Probes for Tumor Diagnosis and Therapy
1986 Endogenous Human Retroviruses
1983 – 1985 New Jersey Cancer Review Study Section

Patents:

The molecular cloning of human syk DNA, compositions containing same and uses thereof are disclosed. 5981262Joan Brugge, Jay Morganstern, Lily Shiue, Lynne Zydowsky, Mark Zoller, Anthony Pawson

Regulatable elimination of gene expression, gene product function and engineered host cells. WO1996006111 A1, Joan S. Brugge and Gerald R. Crabtree.

Methods and materials for identifying inhibitors of molecular interactions mediated by sh3 domains EP0750630 A4 Richard J Rickles, Joan S Brugge, Martyn C Botfield, Mark J Zoller

Publications in Refereed Journals

1. Butel JS, Brugge JS, Noonan CA. Transformation of primate and rodent cells by temperature-sensitive mutants of SV40. Cold Spring Harb Symp Quant Biol. 1975;39 Pt 1:25-36.

2. Brugge JS, Butel JS. Role of simian virus 40 gene A function in maintenance of transformation. J Virol. 1975;15(3):619-35. PMCID: 354498.

3. Vollett J, Brugge JS, and Butel JS. Temperature-dependent microfilament alterations in cells transformed by group A mutants of SV40. J. Cell. Biol. 67:114 (1975).

4. Noonan CA, Brugge JS, Butel JS. Characterization of simian cells tranformed by temperature-sensitive mutants of simian virus 40. J Virol. 1976;18(3):1106-19. PMCID: 354810.

5. Vollet JJ, Brugge JS, Noonan CA, Butel JS. The role of SV40 gene A in the alteration of microfilaments in transformed cells. Exp Cell Res. 1977;105(1):119-26.

6. Erikson E, Brugge JS, Erikson RL. Phosphorylated and nonphosphorylated forms of avian sarcoma virus polypeptide p19. Virology. 1977;80(1):177-85.

7. Brugge JS, Erikson RL. Identification of a transformation-specific antigen induced by an avian sarcoma virus. Nature. 1977;269(5626):346-8.

8. Brugge JS, Purchio AF, Erikson RL. Virus-specific RNA species present in the cytoplasm of Rous sarcoma virus-infected chicken cells. Virology. 1977;83(1):16-26.

9. Brugge JS, Purchio AF, Erikson RL. The distribution of virus-specific RNA in Rous sarcoma virus-induced hamster tumor cells. Virology. 1977;83(1):27-33.

10. Brugge JS, Erikson E, Erikson RL. Antibody to virion structural proteins in mammals bearing avian sarcoma virus-induced tumors. Virology. 1978;84(2):429-33.

11. Purchio AF, Erikson E, Brugge JS, Erikson RL. Identification of a polypeptide encoded by the avian sarcoma virus src gene. Proc Natl Acad Sci U S A. 1978;75(3):1567-71. PMCID: 411515.

12. Brugge JS, Erikson E, Collett M, Erikson RL. Peptide analysis of the transformation-specific antigen from avian sarcoma virus-transformed cells. J. Virol. 26:773 (1978).

13. Brugge JS, Steinbaugh PJ, Erikson RL. Characterization of the avian sarcoma virus protein p60src. Virology. 1978;91(1):130-40.

14. Collett MS, Brugge JS, Erikson RL. Characterization of a normal avian cell protein related to the avian sarcoma virus transforming gene product. Cell. 1978;15(4):1363-9.

15. Brugge JS, Collett MS, Siddiqui A, Marczynska B, Deinhardt F, Erikson RL. Detection of the viral sarcoma gene product in cells infected with various strains of avian sarcoma virus and of a related protein in uninfected chicken cells. J Virol. 1979;29(3):1196-203. PMCID: 353280.

16. Collett MS, Erikson E, Purchio AF, Brugge JS, Erikson RL. A normal cell protein similar in structure and function to the avian sarcoma virus transforming gene product. Proc Natl Acad Sci U S A. 1979;76(7):3159-63. PMCID: 383783.

17. Collett MS, Brugge JS, Erikson RL, Lau AF, Krzyzek RA, Faras AJ. The src gene product of transformed and morphologically reverted ASV-infected mammalian cells. Nature. 1979;281(5728):195-8.

18. Lau AF, Krzyzek RA, Brugge JS, Erikson RL, Schollmeyer J, Faras AJ. Morphological revertants of an avian sarcoma virus-transformed mammalian cell line exhibit tumorigenicity and contain pp60src. Proc Natl Acad Sci U S A. 1979;76(8):3904-8. PMCID: 383944.

19. Erikson RI, Collett MS, Erikson E, Purchio AF, Brugge JS. Protein phosphorylation mediated by partially purified avian sarcoma virus transforming-gene product. Cold Spring Harb Symp Quant Biol. 1980;44 Pt 2:907-17.

20. Erikson RL, Purchio AF, Erikson E, Collett MS, Brugge JS. Molecular events in cells transformed by Rous Sarcoma virus. J Cell Biol. 1980;87(2 Pt 1):319-25. PMCID: 2110755.

21. Brugge JS, Erikson E, Erikson RL. The specific interaction of the Rous sarcoma virus transforming protein, pp60src, with two cellular proteins. Cell. 1981;25(2):363-72.

22. Brugge JS, Darrow D. Rous sarcoma virus-induced phosphorylation of a 50,000-molecular weight cellular protein. Nature. 1982;295(5846):250-3.

23. Lipsich LA, Cutt JR, Brugge JS. Association of the transforming proteins of Rous, Fujinami, and Y73 avian sarcoma viruses with the same two cellular proteins. Mol Cell Biol. 1982;2(7):875-80. PMCID: 369870.

24. Brugge JS, Yonemoto W and Darrow D. Interaction between the Rous sarcoma virus transforming protein and two cellular phosphoproteins: Analysis of the turnover and distribution of this complex. Mol. Cell. Biol. 3:9 (1983).

25. Lanks KW, Kasambalides EJ, Chinkers M, Brugge JS. A major cytoplasmic glucose-regulated protein is associated with the Rous sarcoma virus pp60src protein. J Biol Chem. 1982;257(15):8604-7.

26. Cotton PC, Brugge JS. Neural tissues express high levels of the cellular src gene product pp60c-src. Mol Cell Biol. 1983;3(6):1157-62. PMCID: 368645.

27. Lipsich LA, Lewis AJ, Brugge JS. Isolation of monoclonal antibodies that recognize the transforming proteins of avian sarcoma viruses. J Virol. 1983;48(2):352-60. PMCID: 255359.

28. Keane RW, Lipsich LA, Brugge JS. Differentiation and transformation of neural plate cells. Dev Biol. 1984;103(1):38-52.

29. Brugge JS, Darrow D. Analysis of the catalytic domain of phosphotransferase activity of two avian sarcoma virus-transforming proteins. J Biol Chem. 1984;259(7):4550-7.

30. Chinkers M, Brugge JS. Characterization of structural domains of the human epidermal growth factor receptor obtained by partial proteolysis. J Biol Chem. 1984;259(18):11534-42.

31. Lipsich L, Brugge JS, Boettiger D. Expression of the Rous sarcoma virus src gene in avian macrophages fails to elicit transformed cell phenotype. Mol Cell Biol. 1984;4(7):1420-4. PMCID: 368926.

32. Bolen JB, Thiele CJ, Israel MA, Yonemoto W, Lipsich LA, Brugge JS. Enhancement of cellular src gene product associated tyrosyl kinase activity following polyoma virus infection and transformation. Cell. 1984;38(3):767-77.

33. Hughes S, Mellstrom K, Kosik E, Tamanoi F, Brugge JS. Mutation of a termination codon affects src initiation. Mol. Cell. Bio. 4:1738-1746 (1984).

34. Yonemoto W, Jarvis-Morar M, Brugge JS, Bolen JB, Israel MA. Tyrosine phosphorylation within the amino-terminal domain of pp60c-src molecules associated with polyoma virus middle-sized tumor antigen. Proc Natl Acad Sci U S A. 1985;82(14):4568-72. PMCID: 390426.

35. Brugge JS, Cotton PC, Queral AE, Barrett JN, Nonner D, Keane RW. Neurones express high levels of a structurally modified, activated form of pp60c-src. Nature. 1985;316(6028):554-7.

36. Schuh SM, Brugge JS. Investigation of factors that influence phosphorylation of pp60c-src on tyrosine 527. Mol Cell Biol. 1988;8(6):2465-71. PMCID: 363446.

37. Golden A, Nemeth SP, Brugge JS. Blood platelets express high levels of the pp60c-src-specific tyrosine kinase activity. Proc Natl Acad Sci U S A. 1986;83(4):852-6. PMCID: 322968.

38. Lynch SA, Brugge JS, Levine JM. Induction of altered c-src product during neural differentiation of embryonal carcinoma cells. Science. 1986;234(4778):873-6.

39. Yonemoto W, Filson AJ, Queral-Lustig AE, Wang JY, Brugge JS. Detection of phosphotyrosine-containing proteins in polyomavirus middle tumor antigen-transformed cells after treatment with a phosphotyrosine phosphatase inhibitor. Mol Cell Biol. 1987;7(2):905-13. PMCID: 365149.

40. Brugge JS, Jarosik G, Andersen J, Queral-Lustig A, Fedor-Chaiken M, Broach JR. Expression of Rous sarcoma virus transforming protein pp60v-src in Saccharomyces cerevisiae cells. Mol Cell Biol. 1987;7(6):2180-7. PMCID: 365341.

41. Brugge JS, Cotton PM, Lustig A, Yonemoto W, Lipsich LA, Coussens PM, Barrett JN, Nonner D, Keane RW. Characterization of the altered form of the c-src gene product in neuronal cells. Genes & Dev. 1:287-296 (1987).

42. Brugge JS, Lustig A, Messer A. Changes in the pattern of expression of pp60c-src in cerebellar mutants of mice. J Neurosci Res. 1987;18(4):532-8.

43. Levy JB, Dorai T, Wang LH, Brugge JS. The structurally distinct form of pp60c-src detected in neuronal cells is encoded by a unique c-src mRNA. Mol Cell Biol. 1987;7(11):4142-5. PMCID: 368089.

44. Walaas SI, Lustig A, Greengard P, Brugge JS. Widespread distribution of the c-src gene product in nerve cells and axon terminals in the adult rat brain. Brain Res. 1988;427(3):215-22.

45. Schuh SM, Brugge JS. Investigation of factors that influence phosphorylation of pp60c-src on tyrosine 527. Mol Cell Biol. 1988;8(6):2465-71. PMCID: 363446.

46. Lundy J, Chen J, Wang P, Fromowitz F, Schuss A, Lynch S., Brugge JS, Viola MV. Phenotypic and genetic alterations in pre-cancerous cells in the colon. Anticancer Research 8:1005-1014 (1988).

47. Golden A, Brugge JS. Thrombin treatment induces rapid changes in tyrosine phosphorylation in platelets. Proc Natl Acad Sci U S A. 1989;86(3):901-5. PMCID: 286586.

48. Nemeth SP, Fox LG, DeMarco M, Brugge JS. Deletions within the amino-terminal half of the c-src gene product that alter the functional activity of the protein. Mol Cell Biol. 1989;9(3):1109-19. PMCID: 362701.

49. Levy JB, Brugge JS. Biological and biochemical properties of the c-src+ gene product overexpressed in chicken embryo fibroblasts. Mol Cell Biol. 1989;9(8):3332-41. PMCID: 362378.

50. Ferracini R, Brugge JS. Analysis of mutant forms of the c-src gene product containing a phenylalanine substitution for tyrosine 416. Oncogene Research 5:205-219 (1990).

51. Sugrue MM, Brugge JS, Marshak DR, Greengard P, Gustafson EL. Immunocytochemical localization of the neuron-specific form of the c-src gene product, pp60c-src(+), in rat brain. J Neurosci. 1990;10(8):2513-27.

52. Golden A, Brugge JS, Shattil SJ. Role of platelet membrane glycoprotein IIb-IIIa in agonist-induced tyrosine phosphorylation of platelet proteins. J Cell Biol. 1990;111(6 Pt 2):3117-27. PMCID: 2116418.

53. Filson AJ, Azarnia R, Beyer EC, Loewenstein WR, Brugge JS. Tyrosine phosphorylation of a gap junction protein correlates with inhibition of cell-to-cell communication. Cell Growth Differ. 1990;1(12):661-8.

54. Mitchell CA, Jefferson AB, Bejeck BE, Brugge JS, Deuel TF, Majerus PW. Thrombin-stimulated immunoprecipitation of phosphatidylinositol 3-kinase from human platelets. Proc Natl Acad Sci U S A. 1990;87(23):9396-400. PMCID: 55172.

55. Lord KA, Abdollahi A, Thomas SM, DeMarco M, Brugge JS, Hoffman-Liebermann B, Liebermann DA. Leukemia inhibitory factor and interleukin-6 trigger the same immediate early response, including tyrosine phosphorylation, upon induction of myeloid leukemia differentiation. Mol Cell Biol. 1991;11(9):4371-9. PMCID: 361299.

56. Thomas SM, Hayes M, D’Arcangelo G, Armstrong RC, Meyer BE, Zilberstein A, Brugge JS, Halegoua S. Induction of neurite outgrowth by v-src mimics critical aspects of nerve growth factor-induced differentiation. Mol Cell Biol. 1991;11(9):4739-50. PMCID: 361372.

57. Dorai T, Levy JB, Kang L, Brugge JS, Wang LH. Analysis of cDNAs of the proto-oncogene c-src: heterogeneity in 5′ exons and possible mechanism for the genesis of the 3′ end of v-src. Mol Cell Biol. 1991;11(8):4165-76. PMCID: 361236.

58. Huang MM, Bolen JB, Barnwell JW, Shattil SJ, Brugge JS. Membrane glycoprotein IV (CD36) is physically associated with the Fyn, Lyn, and Yes protein-tyrosine kinases in human platelets. Proc Natl Acad Sci U S A. 1991;88(17):7844-8. PMCID: 52400.

59. Kremer NE, D’Arcangelo G, Thomas SM, DeMarco M, Brugge JS, Halegoua S. Signal transduction by nerve growth factor and fibroblast growth factor in PC12 cells requires a sequence of src and ras actions. J Cell Biol. 1991;115(3):809-19. PMCID: 2289191.

60. Thomas JE, Soriano P, Brugge JS. Phosphorylation of c-Src on tyrosine 527 by another protein tyrosine kinase. Science. 1991;254(5031):568-71.

61. Schmidt JW, Brugge JS, Nelson WJ. pp60src tyrosine kinase modulates P19 embryonal carcinoma cell fate by inhibiting neuronal but not epithelial differentiation. J Cell Biol. 1992;116(4):1019-33. PMCID: 2289338.

62. Huang MM, Indik Z, Brass LF, Hoxie JA, Schreiber AD, Brugge JS. Activation of Fc gamma RII induces tyrosine phosphorylation of multiple proteins including Fc gamma RII. J Biol Chem. 1992;267(8):5467-73.

63. Cichowski K, McCormick F, Brugge JS. p21rasGAP association with Fyn, Lyn, and Yes in thrombin-activated platelets. J Biol Chem. 1992;267(8):5025-8.

64. Seidel-Dugan C, Meyer BE, Thomas SM, Brugge JS. Effects of SH2 and SH3 deletions on the functional activities of wild-type and transforming variants of c-Src. Mol Cell Biol. 1992;12(4):1835-45. PMCID: 369627.

65. Thomas SM, DeMarco M, D’Arcangelo G, Halegoua S, Brugge JS. Ras is essential for nerve growth factor- and phorbol ester-induced tyrosine phosphorylation of MAP kinases. Cell. 1992;68(6):1031-40.

66. Maroney AC, Qureshi SA, Foster DA, Brugge JS. Cloning and characterization of a thermolabile v-src gene for use in reversible transformation of mammalian cells. Oncogene. 1992;7(6):1207-14.

67. Lipfert L, Haimovich B, Schaller MD, Cobb BS, Parsons JT, Brugge JS. Integrin-dependent phosphorylation and activation of the protein tyrosine kinase pp125FAK in platelets. J Cell Biol. 1992;119(4):905-12. PMCID: 2289696.

68. Yu H, Rosen MK, Shin TB, Seidel-Dugan C, Brugge JS, Schreiber SL. Solution structure of the SH3 domain of Src and identification of its ligand-binding site. Science. 1992;258(5088):1665-8.

69. Rozakis-Adcock M, McGlade J, Mbamalu G, Pelicci G, Daly R, Li W, Batzer A, Thomas S, Brugge JS, Pelicci PG, Schlessinger J, Pawson T. Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases. Nature 360: 689-692 (1992).

70. Clark EA, Brugge JS. Redistribution of activated pp60c-src to integrin-dependent cytoskeletal complexes in thrombin-stimulated platelets. Mol Cell Biol. 1993;13(3):1863-71. PMCID: 359499.

71. Fox JE, Lipfert L, Clark EA, Reynolds CC, Austin CD, Brugge JS. On the role of the platelet membrane skeleton in mediating signal transduction. Association of GP IIb-IIIa, pp60c-src, pp62c-yes, and the p21ras GTPase-activating protein with the membrane skeleton. J Biol Chem. 1993;268(34):25973-84.

72. Haimovich B, Lipfert L, Brugge JS, Shattil SJ. Tyrosine phosphorylation and cytoskeletal reorganization in platelets are triggered by interaction of integrin receptors with their immobilized ligands. J Biol Chem. 1993;268(21):15868-77.

73. Thomas JE, Aguzzi A, Soriano P, Wagner EF, Brugge JS. Induction of tumor formation and cell transformation by polyoma middle T antigen in the absence of Src. Oncogene. 1993;8(9):2521-9.

74. Weng Z, Taylor JA, Turner CE, Brugge JS, Seidel-Dugan C. Detection of Src homology 3-binding proteins, including paxillin, in normal and v-Src-transformed Balb/c 3T3 cells. J Biol Chem. 1993;268(20):14956-63.

75. Lynch SA, Brugge JS, Fromowitz F, Glantz L, Wang P, Caruso R, Viola MV. Increased expression of the src proto-oncogene in hairy cell leukemia and a subgroup of B-cell lymphomas. Leukemia. 1993;7(9):1416-22.

76. Huang MM, Lipfert L, Cunningham M, Brugge JS, Ginsberg MH, Shattil SJ. Adhesive ligand binding to integrin alpha IIb beta 3 stimulates tyrosine phosphorylation of novel protein substrates before phosphorylation of pp125FAK. J Cell Biol. 1993;122(2):473-83. PMCID: 2119653.

77. Shattil SJ, Haimovich B, Cunningham M, Lipfert L, Parsons JT, Ginsberg MH, Brugge JS. Tyrosine phosphorylation of pp125FAK in platelets requires coordinated signaling through integrin and agonist receptors. J Biol Chem. 1994;269(20):14738-45.(1)

78. Weng Z, Thomas SM, Rickles RJ, Taylor JA, Brauer AW, Seidel-Dugan C, Michael WM, Dreyfuss G, Brugge JS. Identification of Src, Fyn, and Lyn SH3-binding proteins: implications for a function of SH3 domains. Mol Cell Biol. 1994;14(7):4509-21. PMCID: 358823.

79. Clark EA, Shattil SJ, Ginsberg MH, Bolen J, Brugge JS. Regulation of the protein tyrosine kinase pp72syk by platelet agonists and the integrin alpha IIb beta 3. J Biol Chem. 1994;269(46):28859-64.

80. Clark EA, Trikha M, Markland FS, Brugge JS. Structurally distinct disintegrins contortrostatin and multisquamatin differentially regulate platelet tyrosine phosphorylation. J Biol Chem. 1994;269(35):21940-3.

81. Rickles RJ, Botfield MC, Weng Z, Taylor JA, Green OM, Brugge JS, Zoller MJ. Identification of Src, Fyn, Lyn, PI3K and Abl SH3 domain ligands using phage display libraries. EMBO J. 1994;13(23):5598-604. PMCID: 395523.

82. Shiue L, Green J, Green OM, Karas JL, Morgenstern JP, Ram MK. Taylor MK, Zoller MJ, Zydowsky LD, Bolen JB, Brugge JS. Interaction of p72syk with the g and b subunits of the high-affinity receptor for immunoglobulin E, FcERI. Mol. Cell. Biol. 15: 272-281 (1995).

83. Rivera VM, Brugge JS. Clustering of Syk is sufficient to induce tyrosine phosphorylation and release of allergic mediators from rat basophilic leukemia cells. Mol Cell Biol. 1995;15(3):1582-90. PMCID: 230382.

84. Shiue L, Zoller MJ, Brugge JS. Syk is activated by phosphotyrosine-containing peptides representing the tyrosine-based activation motifs of the high affinity receptor for IgE. J Biol Chem. 1995;270(18):10498-502.

85. Mukhopadhyay D, Tsiokas L, Zhou XM, Foster D, Brugge JS, Sukhatme VP. Hypoxic induction of human vascular endothelial growth factor expression through c-Src activation. Nature. 1995;375(6532):577-81.

86. Weng Z, Rickles RJ, Feng S, Richard S, Shaw AS, Schreiber SL, Brugge JS. Structure-function analysis of SH3 domains: SH3 binding specificity altered by single amino acid substitutions. Mol Cell Biol. 1995;15(10):5627-34. PMCID: 230813.

87. Rickles RJ, Botfield MC, Zhou XM, Henry PA, Brugge JS, Zoller MJ. Phage display selection of ligand residues important for Src homology 3 domain binding specificity. Proc Natl Acad Sci U S A. 1995;92(24):10909-13. PMCID: 40540.

88. Rusanescu G, Qi H, Thomas SM, Brugge JS, Halegoua S. Calcium influx induces neurite growth through a Src-Ras signaling cassette. Neuron. 1995;15(6):1415-25.

89. Cichowski K, Brugge JS, Brass LF. Thrombin receptor activation and integrin engagement stimulate tyrosine phosphorylation of the proto-oncogene product, p95vav, in platelets. J Biol Chem. 1996;271(13):7544-50.

90. Zoller KE, MacNeil IA, Brugge JS. Protein tyrosine kinases Syk and ZAP-70 display distinct requirements for Src family kinases in immune response receptor signal transduction. J Immunol. 1997;158(4):1650-9.

91. King WG, Mattaliano MD, Chan TO, Tsichlis PN, Brugge JS. Phosphatidylinositol 3-kinase is required for integrin-stimulated AKT and Raf-1/mitogen-activated protein kinase pathway activation. Mol Cell Biol. 1997;17(8):4406-18. PMCID: 232295.

92. Gao J, Zoller KE, Ginsberg MH, Brugge JS, Shattil SJ. Regulation of the pp72syk protein tyrosine kinase by platelet integrin alpha IIb beta 3. EMBO J. 1997;16(21):6414-25. PMCID: 1170248.

93. Clark EA, King WG, Brugge JS, Symons M, Hynes RO. Integrin-mediated signals regulated by members of the rho family of GTPases. J Cell Biol. 1998;142(2):573-86. PMCID: 2133065.

94. Miranti CK, Leng L, Maschberger P, Brugge JS, Shattil SJ. Identification of a novel integrin signaling pathway involving the kinase Syk and the guanine nucleotide exchange factor Vav1. Curr Biol. 1998;8(24):1289-99.

95. Miranti CK, Ohno S, Brugge JS. Protein kinase C regulates integrin-induced activation of the extracellular regulated kinase pathway upstream of Shc. J Biol Chem. 1999;274(15):10571-81.

96. Muthuswamy SK, Gilman M, Brugge JS. Controlled dimerization of ErbB receptors provides evidence for differential signaling by homo- and heterodimers. Mol Cell Biol. 1999;19(10):6845-57. PMCID: 84681.

97. Rao VR, Corradetti MN, Chen J, Peng J, Yuan J, Prestwich GD, Brugge JS. Expression cloning of protein targets for 3-phosphorylated phosphoinositides. J Biol Chem. 1999;274(53):37893-900.

98. Chaudhary A, King WG, Mattaliano MD, Frost JA, Diaz B, Morrison DK, Cobb MH, Marshall MS, Brugge JS. Phosphatidylinositol 3-kinase regulates Raf1 through Pak phosphorylation of serine 338. Curr Biol. 2000;10(9):551-4.

99. Moores SL, Selfors LM, Fredericks J, Breit T, Fujikawa K, Alt FW, Brugge JS, Swat W. Vav family proteins couple to diverse cell surface receptors. Mol Cell Biol. 2000;20(17):6364-73. PMCID: 86111.

100. Muthuswamy SK, Li D, Lelievre S, Bissell MJ, Brugge JS. ErbB2, but not ErbB1, reinitiates proliferation and induces luminal repopulation in epithelial acini. Nat Cell Biol. 2001;3(9):785-92. PMCID: 2952547.

101. Woodside DG, Obergfell A, Leng L, Wilsbacher JL, Miranti CK, Brugge JS, Shattil SJ, Ginsberg MH. Activation of Syk protein tyrosine kinase through interaction with integrin beta cytoplasmic domains. Curr Biol. 2001;11(22):1799-804.

102. Chaudhary A, Brugge JS, Cooper JA. Direct phosphorylation of focal adhesion kinase by c-Src: evidence using a modified nucleotide pocket kinase and ATP analog. Biochem Biophys Res Commun. 2002;294(2):293-300.

103. Obergfell A, Eto K, Mocsai A, Buensuceso C, Moores SL, Brugge JS, Lowell CA, Shattil SJ. Coordinate interactions of Csk, Src, and Syk kinases with [alpha]IIb[beta]3 initiate integrin signaling to the cytoskeleton. J Cell Biol. 2002;157(2):265-75. PMCID: 2199242.

104. Debnath J, Mills KR, Collins NL, Reginato MJ, Muthuswamy SK, Brugge JS. The role of apoptosis in creating and maintaining luminal space within normal and oncogene-expressing mammary acini. Cell. 2002;111(1):29-40.

105. Debnath J, Muthuswamy SK, Brugge JS. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods. 2003;30(3):256-68.

106. Reginato MJ, Mills KR, Paulus JK, Lynch DK, Sgroi DC, Debnath J, Muthuswamy SK, Brugge JS. Integrins and EGFR coordinately regulate the pro-apoptotic protein Bim to prevent anoikis. Nat Cell Biol. 2003;5(8):733-40.

107. Gozani O, Karuman P, Jones DR, Ivanov D, Cha J, Lugovskoy AA, Baird CL, Zhu H, Field SJ, Lessnick SL, Villasenor J, Mehrotra B, Chen J, Rao VR, Brugge JS, Ferguson CG, Payrastre B, Myszka DG, Cantley LC, Wagner G, Divecha N, Prestwich GD, Yuan J. The PHD finger of the chromatin-associated protein ING2 functions as a nuclear phosphoinositide receptor. Cell. 2003;114(1):99-111.

108. Debnath J, Walker SJ, Brugge JS. Akt activation disrupts mammary acinar architecture and enhances proliferation in an mTOR-dependent manner. J Cell Biol. 2003;163(2):315-26. PMCID: 2173511.

109. Arias-Salgado EG, Lizano S, Sarkar S, Brugge JS, Ginsberg MH, Shattil SJ. Src kinase activation by direct interaction with the integrin beta cytoplasmic domain. Proc Natl Acad Sci U S A. 2003;100(23):13298-302. PMCID: 263791.

110. Fujikawa, K., Miletic, A.V., Alt, FW, Faccio, R, Brown, T. , Hoog. J, Fredericks, J., Nichi, S, Mildiner, S. Moores, S. Brugge, JS, Rosen, F, Swat, W. Vav1/2/3-Null Mice Define an Essential Role for Vav Family Proteins in Lymphocyte Development and Activation but a Differential Requirement in MAPK Signaling in T and B Cells. J. Exp. Med. 198: 1595-608. (2003).

111. Seton-Rogers SE, Lu Y, Hines LM, Koundinya M, LaBaer J, Muthuswamy SK, Brugge JS. Cooperation of the ErbB2 receptor and transforming growth factor beta in induction of migration and invasion in mammary epithelial cells. Proc Natl Acad Sci U S A. 2004;101(5):1257-62. PMCID: 337040.

112. Mills KR, Reginato M, Debnath J, Queenan B, Brugge JS. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is required for induction of autophagy during lumen formation in vitro. Proc Natl Acad Sci U S A. 2004;101(10):3438-43. PMCID: 373480.

113. Wrobel CN, Debnath J, Lin E, Beausoleil S, Roussel MF, Brugge JS. Autocrine CSF-1R activation promotes Src-dependent disruption of mammary epithelial architecture. J Cell Biol. 2004;165(2):263-73. PMCID: 2172030.

114. Bill HM, Knudsen B, Moores SL, Muthuswamy SK, Rao VR, Brugge JS, Miranti CK. Epidermal growth factor receptor-dependent regulation of integrin-mediated signaling and cell cycle entry in epithelial cells. Mol Cell Biol. 2004;24(19):8586-99. PMCID: 516761.

115. Martin SS, Ridgeway AG, Pinkas J, Lu Y, Reginato MJ, Koh EY, Michelman M, Daley GQ, Brugge JS, Leder P. A cytoskeleton-based functional genetic screen identifies Bcl-xL as an enhancer of metastasis, but not primary tumor growth. Oncogene. 2004;23(26):4641-5.

116. Gakidis MA, Cullere X, Olson T, Wilsbacher JL, Zhang B, Moores SL, Ley K, Swat W, Mayadas T, Brugge JS. Vav GEFs are required for beta2 integrin-dependent functions of neutrophils. J Cell Biol. 2004;166(2):273-82. PMCID: 2172310.

117. Ma X, Wang Z, Ryan PD, Isakoff SI, Barmettler A, Fuller A, Muir B, Mohapatra G, Salunga R, Tuggle JT, Tran Y, Tran D, Sollberger A, Amon P, Wang W, Wang W, Stecker K, Estepa-Sabal E, Smith B, Younger J, Balis U, Bhan A, Habin K, Baer TM, Brugge JS, Haber DA, Erlander MG, Sgroi DC. A two-gene expression ratio predicts clinical outcome in breast cancer patients treated with tamoxifen. Cancer Cell 5:607-16 (2004).

118. Reginato MJ, Mills KR, Becker EB, Lynch DK, Bonni A, Muthuswamy SK, Brugge JS. Bim regulation of lumen formation in cultured mammary epithelial acini is targeted by oncogenes. Mol Cell Biol. 2005;25(11):4591-601. PMCID: 1140636.

119. Collins NL, Reginato MJ, Paulus JK, Sgroi DC, Labaer J, Brugge JS. G1/S cell cycle arrest provides anoikis resistance through Erk-mediated Bim suppression. Mol Cell Biol. 2005;25(12):5282-91. PMCID: 1140593.

120. Cowan CW, Shao YR, Sahin M, Shamah SM, Lin MZ, Greer PL, Gao S, Griffith EC, Brugge JS, Greenberg ME. Vav family GEFs link activated Ephs to endocytosis and axon guidance. Neuron. 2005;46(2):205-17.

121. Yan SR, Joseph RR, Rosen K, Reginato MJ, Jackson A, Allaire N, Brugge JS, Jobin C, Stadnyk AW. Activation of NF-kappaB following detachment delays apoptosis in intestinal epithelial cells. Oncogene. 2005;24(43):6482-91. PMCID: 1509103.

122. Isakoff SJ, Engelman JA, Irie HY, Luo J, Brachmann SM, Pearline RV, Cantley LC, Brugge JS. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005;65(23):10992-1000.

123. Gunawardane RN, Sgroi DC, Wrobel CN, Koh E, Daley GQ, Brugge JS. Novel role for PDEF in epithelial cell migration and invasion. Cancer Res. 2005;65(24):11572-80.

124. Irie HY, Pearline RV, Grueneberg D, Hsia M, Ravichandran P, Kothari N, Natesan S, Brugge JS. Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J Cell Biol. 2005;171(6):1023-34. PMCID: 2171329.

125. Carroll DK, Carroll JS, Leong CO, Cheng F, Brown M, Mills AA, Brugge JS, Ellisen LW. p63 regulates an adhesion programme and cell survival in epithelial cells. Nat Cell Biol. 2006;8(6):551-61.

126. Hall AB, Gakidis MA, Glogauer M, Wilsbacher JL, Gao S, Swat W, Brugge JS. Requirements for Vav guanine nucleotide exchange factors and Rho GTPases in FcgammaR- and complement-mediated phagocytosis. Immunity. 2006;24(3):305-16.

127. Witt AE, Hines LM, Collins NL, Hu Y, Gunawardane RN, Moreira D, Raphael J, Jepson D, Koundinya M, Rolfs A, Taron B, Isakoff SJ, Brugge JS, LaBaer J. Functional proteomics approach to investigate the biological activities of cDNAs implicated in breast cancer. J Proteome Res. 2006;5(3):599-610. PMCID: 2522320.

128. Wilsbacher JL, Moores SL, Brugge JS. An active form of Vav1 induces migration of mammary epithelial cells by stimulating secretion of an epidermal growth factor receptor ligand. Cell Commun Signal. 2006;4:5. PMCID: 1524963.

129. Overholtzer M, Zhang J, Smolen GA, Muir B, Li W, Sgroi DC, Deng CX, Brugge JS, Haber DA. Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci U S A. 2006;103(33):12405-10. PMCID: 1533802.

130. Mailleux AA, Overholtzer M, Schmelzle T, Bouillet P, Strasser A, Brugge JS. BIM regulates apoptosis during mammary ductal morphogenesis, and its absence reveals alternative cell death mechanisms. Dev Cell. 2007;12(2):221-34. PMCID: 2698712.

131. Schmelzle T, Mailleux AA, Overholtzer M, Carroll JS, Solimini NL, Lightcap ES, Veiby OP, Brugge JS. Functional role and oncogene-regulated expression of the BH3-only factor Bmf in mammary epithelial anoikis and morphogenesis. Proc Natl Acad Sci U S A. 2007;104(10):3787-92. PMCID: 1820662.

132. Miller-Jensen K, Janes KA, Brugge JS, Lauffenburger DA. Common effector processing mediates cell-specific responses to stimuli. Nature 448 (7153): 604-8, 2007.

133. Overholtzer M, Mailleux AA, Mouneimne G, Normand G, Schnitt SJ, King RW, Cibas ES, Brugge JS. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. Cell 131 (5): 966-79, 2007.

134. Fischbach C, Chen R, Matsumoto T, Schmelzle T, Brugge JS, Polverini PJ, Mooney DJ. Engineering tumors with 3D scaffolds. Nat Methods 4 (10): 855-60, 2007.

135. Melani M, Simpson KJ, Brugge JS, Montell D. Regulation of Cell Adhesion and Collective Cell Migration by Hindsight and Its Human Homolog RREB1. Curr Biol 18 (7): 532-7, 2008.

136. Simpson KJ, Selfors LM, Bui J, Reynolds A, Leake D, Khvorova A, Brugge JS. Identification of genes that regulate epithelial cell migration using an siRNA screening approach. Nat Cell Biol. 2008;10(9):1027-38.

137. Tamimi RM, Brugge JS, Freedman ML, Miron A, Iglehart JD, Colditz GA, Hankinson SE. Circulating colony stimulating factor-1 and breast cancer risk. Cancer Res. 2008;68(1):18-21. PMCID: 2821592.

138. Konecny GE, Glas R, Dering J, Manivong K, Qi J, Finn RS, Yang GR, Hong KL, Ginther C, Winterhoff B, Gao G, Brugge J, Slamon DJ. Activity of the multikinase inhibitor dasatinib against ovarian cancer cells. Br J Cancer. 2009;101(10):1699-708. PMCID: 2778533.

139. Muller PA, Caswell PT, Doyle B, Iwanicki MP, Tan EH, Karim S, Lukashchuk N, Gillespie DA, Ludwig RL, Gosselin P, Cromer A, Brugge JS, Sansom OJ, Norman JC, Vousden KH. Mutant p53 drives invasion by promoting integrin recycling. Cell. 2009;139(7):1327-41.

140. Schafer ZT, Grassian AR, Song L, Jiang Z, Gerhart-Hines Z, Irie HY, Gao S, Puigserver P, Brugge JS. Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment. Nature. 2009;461(7260):109-13.

141. Xian W, Pappas L, Pandya D, Selfors LM, Derksen PW, de Bruin M, Gray NS, Jonkers J, Rosen JM, Brugge JS. Fibroblast growth factor receptor 1-transformed mammary epithelial cells are dependent on RSK activity for growth and survival. Cancer Res. 2009;69(6):2244-51.

142. Zhang J, Ji JY, Yu M, Overholtzer M, Smolen GA, Wang R, Brugge JS, Dyson NJ, Haber DA. YAP-dependent induction of amphiregulin identifies a non-cell-autonomous component of the Hippo pathway. Nat Cell Biol. 2009;11(12):1444-50. PMCID: 2819909.

143. Janes KA, Wang CC, Holmberg KJ, Cabral K, Brugge JS. Identifying single-cell molecular programs by stochastic profiling. Nat Methods. 2010;7(4):311-7. PMCID: 2849806.

144. Mazzone M, Selfors LM, Albeck J, Overholtzer M, Sale S, Carroll DL, Pandya D, Lu Y, Mills GB, Aster JC, Artavanis-Tsakonas S, Brugge JS. Dose-dependent induction of distinct phenotypic responses to Notch pathway activation in mammary epithelial cells. Proc Natl Acad Sci U S A. 2010;107(11):5012-7. PMCID: 2841923.

145. Tushir JS, Clancy J, Warren A, Wrobel C, Brugge JS, D’Souza-Schorey C. Unregulated ARF6 Activation in Epithelial Cysts Generates Hyperactive Signaling Endosomes and Disrupts Morphogenesis. Mol Biol Cell. 2010. PMCID: 2893997.

146. Wolfer A, Wittner BS, Irimia D, Flavin RJ, Lupien M, Gunawardane RN, Meyer CA, Lightcap ES, Tamayo P, Mesirov JP, Liu XS, Shioda T, Toner M, Loda M, Brown M, Brugge JS, Ramaswamy S. MYC regulation of a “poor-prognosis” metastatic cancer cell state. Proc Natl Acad Sci U S A. 2010;107(8):3698-703. PMCID: 2840447.

147. Zhou W, Hur W, McDermott U, Dutt A, Xian W, Ficarro SB, Zhang J, Sharma SV, Brugge J, Meyerson M, Settleman J, Gray NS. A structure-guided approach to creating covalent FGFR inhibitors. Chem Biol. 2010;17(3):285-95.

148. Irie HY, Shrestha Y, Selfors LM, Frye F, Iida N, Wang Z, Zou L, Yao J, Lu Y, Epstein CB, Natesan S, Richardson AL, Polyak K, Mills GB, Hahn WC, Brugge JS. PTK6 regulates IGF-1-induced anchorage-independent survival. PLoS One. 2010;5(7):e11729. PMCID: 2909213.

149. Knowlton ML, Selfors LM, Wrobel CN, Gu TL, Ballif BA, Gygi SP, Polakiewicz R, Brugge JS. Profiling Y561-dependent and -independent substrates of CSF-1R in epithelial cells. PLoS One. 2010;5(10):e13587. PMCID: 2964295.

150. Locasale JW, Grassian AR, Melman T, Lyssiotis CA, Mattaini KR, Bass AJ, Heffron G, Metallo CM, Muranen T, Sharfi H, Sasaki AT, Anastasiou D, Mullarky E, Vokes NI, Sasaki M, Beroukhim R, Stephanopoulos G, Ligon AH, Meyerson M, Richardson AL, Chin L, Wagner G, Asara JM, Brugge JS, Cantley LC, Vander Heiden MG. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat Genet 2011 Jul 31;43(9):869-74.

151. Iwanicki MP, Davidowitz RA, Ng MR, Besser A, Muranen T, Merritt M, Danuser G, Ince T, Brugge JS. Ovarian Cancer Spheroids Use Myosin-Generated Force to Clear the Mesothelium. Cancer Discovery. July 2011 1:144-157.

152. Grassian AR, Schafer ZT, Brugge JS. ErbB2 stabilizes epidermal growth factor receptor (EGFR) expression via Erk and Sprouty2 in extracellular matrix-detached cells. J Biol Chem. 2011;286(1):79-90. PMCID: 3013038.

153. Kim JH, Cho A, Yin H, Schafer DA, Mouneimne G, Simpson KJ, Nguyen KV, Brugge JS, Montell DJ. Psidin, a conserved protein that regulates protrusion dynamics and cell migration. Genes Dev. 2011;25(7):730-41. PMCID: 3070935.

154. Krajcovic M, Johnson NB, Sun Q, Normand G, Hoover N, Yao E, Richardson AL, King RW, Cibas ES, Schnitt SJ, Brugge JS, Overholtzer M. A non-genetic route to aneuploidy in human cancers. Nat Cell Biol. 2011;13(3):324-30.

155. Grassian AR, Metallo CM, Coloff JL, Stephanopoulos G, Brugge JS. Erk regulation of pyruvate dehydrogenase flux through PDK4 modulates cell proliferation. Genes Dev. 2011;25(16):1716-33.

156. Wang L, Brugge JS, Janes KA. Intersection of FOXO- and RUNX1-mediated gene expression programs in single breast epithelial cells during morphogenesis and tumor progression. Proc Natl Acad Sci U S A. 2011;108(40):E803-12. PMCID: 3189061.

157. Davidowitz RA, Iwanicki MP, Brugge JS. In vitro Mesothelial Clearance Assay that Models the Early Steps of Ovarian Cancer Metastasis. J Vis Exp. 2012(60).
158. Worster DT, Schmelzle T, Solimini NL, Lightcap ES, Millard B, Mills GB, Brugge JS, Albeck JG. Akt and ERK Control the Proliferative Response of Mammary Epithelial Cells to the Growth Factors IGF-1 and EGF Through the Cell Cycle Inhibitor p57Kip2. Sci Signal. 2012;5(214):ra19.

159. Mouneimne G, Hansen SD, Selfors LM, Petrak L, Hickey MM, Gallegos LL, Simpson KJ, Lim J, Gertler FB, Hartwig JH, Mullins RD, Brugge JS. Differential remodeling of actin cytoskeleton architecture by profilin isoforms leads to distinct effects on cell migration and invasion. Cancer Cell. 2012; 22(5):615-30. PMCID: 3500527.

160. Leung, C and Brugge JS. Outgrowth of Single Oncogene-expressing Cells from Suppressive Epithelial Environments. Nature, 2012 482(7385):410-3 PMCID: PMC3297969

161. Ng MR, Besser A, Danuser G, Brugge JS. Substrate stiffness regulates cadherin-dependent collective migration through myosin-II contractility. J Cell Biol. 2012;199(3):545-63. PMCID: 3483134.

162. Muranen, T, Selfors LM, Worster DT, Iwanicki MP, Song L, Morales FC, Gao S, Mills GB and
Brugge JS, Inhibition of PI3K/mTOR Leads to Adaptive Resistance in Matrix-Attached Cancer Cells.
Cancer Cell, 2012 Feb 14;21(2):227-39

163. Ng MR, Besser A, Danuser G, Brugge JS. Substrate stiffness regulates cadherin-dependent collective migration through myosin-II contractility. J Cell Biol. 2012;199(3):545-63. PMCID: 3483134.

164. Albeck JG, Mills GB, Brugge, JS, Frequency-Modulated Pulses of ERK Activity Transmit Quantitative Proliferation Signals. Mol Cell. 2013;49(2):249-61.

165. Buchwalter, G*, Hickey, M*, Cromer, C, Selfors,LM, Gunawardane, RN, Frishman, J., Jeselsohn, R, Lim, E, Chi, D, Fu, X, , Schiff, R, Brown, M#, Brugge, JS # PDEF Promotes Luminal Differentiation and Acts as a Survival Factor for ER-Positive Breast Cancer Cells Cancer Cell 2013 23: 753–767.*co-first authors, # co-corresponding authors
166. Kaanta AS, Virtanen C, Selfors L, Brugge JS, Neel BG. Evidence for a multipotent mammary progenitor with pregnancy-specific activity. Breast Cancer Research. 2013 Aug 15;15(4):R65
167. Vakifahmetoglu-Norberg H, Kim M, Xia HG, Iwanicki MP, Ofengeim D, Coloff JL, Pan L, Ince TA, Kroemer G, Brugge JS, Yuan J. Chaperone-mediated autophagy degrades mutant p53. Genes Dev. 2013 Aug 1;27(15):1718-30.
168. Davidowitz RA, Selfors, RM, Iwanicki MP, Karst A, Piao H, Ince TA, Drage MG, Dering J, Konecny GE, Matulonis U, Mills GB, Slamon D, Drapkin R, Brugge JS. Mesenchymal Gene Program Expressing Ovarian Cancer Spheroids Exhibit Mesenchymal Clearance, J Clin Invest. 2014 Jun;124(6):2611-25. PMCID: PMC4038562
169. Ng MR., Besser A, Brugge JS, Danuser G. Mapping the dynamics of force transduction at cell-cell junctions of epithelial clusters. Elife. 2014 Dec 5 PMCID: PMC4311494

170. Godinho SA, Picone R, Burute M, Regina Dagher R, Ying Su Y, Leung CT, Kornelia Polyak K, Brugge JS, Thery M, and Pellman D. Oncogene-like induction of cellular invasion from centrosome amplification, Nature 2014: 510:167-71

171.Ince TA, Sousa AD, Jones MA, Harrell JC, Agoston ES, Krohn M, Selfors LM, Liu W, Chen K, Yong M, Buchwald P, Wang B, Hale KS, Cohick E, Sergent P, Witt A, Kozhekbaeva Z, Gao S, Agoston AT, Merritt MA, Foster R, Rueda BR, Crum CP, Brugge JS, Mills GB. Characterization of twenty-five ovarian tumour cell lines that phenocopy primary tumours. Nat Commun 2015.: 6:7419. PMCID: PMC4473807

172. Bui AD, Lee W, White AE, Harper JW, Schackmann RCJ, Overholtzer M, Selfors LM, Brugge JS. Cytokinesis involves a nontranscriptional function of the Hippo pathway effector YAP. 2016 Science Signaling 9(417):ra23.

173. Stover DG, Coloff JL, Barry WT, Brugge JS Winer Brugge JS, Selfors LM The Role of Proliferation in Determining Response to Neoadjuvant Chemotherapy in Breast Cancer: A Gene Expression-Based Meta-Analysis, Clin Can Res 2016 22(24):6039-6050

174. Coloff JL, Murphy P, Braun CR, Harris IS, Shelton LM, Kami K, Gygi SP, Selfors LM, Brugge JS Differential Glutamate Metabolism in Proliferating and Quiescent Mammary Epithelial Cells, Cell Metabolism 2016, 23: 867–880

175: Gallegos LL, Ng MR, Sowa ME, Selfors LM, White A, Zervantonakis IK, Singh P, Harper JW, Brugge JS. A protein interaction map for cell-cell adhesion regulators identifies DUSP23 as a novel phosphatase for β-catenin Scientific Reports 2016, 6:27114.

176. Muranen, T, Selfors LT, Julie Hwang, Gallegos LL, Coloff JL, Thoreen CC, Kang SA, Sabatini DM, Mills GB and Brugge JS The Balance of ERK and p38 MAPK Activity Determines the Sensitivity to PI3K/mTOR Inhibition Through Regulation of MYC and YAP, Cancer Res 2016, 76(24):7168-7180.

177. Iwanicki MP, Chen HY, Iavarone C, Zervantonakis IK, Muranen T, Novak M, Ince TA, Drapkin R and Brugge JS. Mutant p53 regulates ovarian cancer transformed phenotypes through autocrine matrix deposition. JCI Insight, 2016: 1(10) pii: e86829. PMCID PMC4963159

178. Zoeller JJ, Bronson RT, Selfors LM,. Mills GB, Brugge JS. Niche-localized tumor cells are protected from HER2-targeted therapy via upregulation of an anti-apoptotic program in vivo, npj Breast Cancer 2017: 3, doi: 10.1038/s41523-017-0020-z

179. Liu JF, Palakurthi S, Zeng Q, Zhou S, Ivanova E, Huang W, Zervantonakis IK, Selfors LM, Shen Y, Pritchard CC, Zheng M, Wulf GM, Paweletz C, English J, Kirschmeier P, Mills GB, Livingston D, Brugge JS, Matulonis U, Drapkin R Establishment of patient-derived tumor xenograft models of epithelial ovarian cancer for pre-clinical evaluation of novel therapeutics, 2016 Clin Cancer Res, 2017 Mar 1; 23(5): 1263–1273. PMCID: PMC5332350

180. Muranen T, Curry NL, Hwang J, DuBois CD, Hitchcock DS, Iwanicki MP, Coloff JL, Clish CB, Brugge JS and Kalaany NY. Starved epithelial cells uptake extracellular matrix for survival, 2017. Nature Comm, 8:13989 PMCID: PMC5234072

181. Zervantonakis IK, Iavarone C, Chen H-Y, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis U, Leverson JD, Sampath D, Mills GB, Brugge JS Targeting apoptotic vulnerabilities in patient-derived ovarian cancer xenografts to enhance the efficacy of PI3K pathway inhibitors, 2017 Nature Comm,;8(1):365. doi: 10.1038/s41467-017-00263-7 PMCID: PMC5573720

182. Takahashi N, Chen H-Y, Harris IS, Stover D, Bronson RT, Takemura RT, Kitao A, Deraedt T, Cichowski K, Welm AL, Mori, Y, Mills GB, Brugge JS. Cancer Cells Co-opt the Neuronal Redox-Sensing Channel TRPA1 to Promote Oxidative-Stress Tolerance. 2018. Cancer Cell 33: 6, p985-1003. https://doi.org/10.1016/j.ccell.2018.05.001

183. Wang Y, Woehrstein JB, Donoghue N, Dai M, Avendano MS, Schackmann RCJ, Wang SS, Tillberg, PW, Park D, Lapan, SW, Boyden ES, Brugge JS, Kaese, PS, Church GM, Agasti SS, Jungmann R, Yin P Rapid sequential in situ multiplexing with DNA-Exchange-Imaging bioR????iv 2017, https://doi.org/10.1101/112227

184. Selfors, LM, ,Stover, DG. Harris, IS, Brugge, JS, and Coloff, JL. Identification of cancer genes that are independent of proliferation and lineage programs. 2017 Proc Natl Acad Sci U S A. Dec 26; 114(52): E11276–E11284. Published online 2017 Dec 11. doi: 10.1073/pnas.1714877115

185. Hung YP, Teragawa C, Gilles T, Pargett M, Minguet M. Distor K, Gregg B, Yellen G, Brugge JS, Albeck JG. Akt regulation of glycolysis mediates bioenergetic stability in epithelial cells 2017 ELife, Dec 14;6. PMCID:PMC5730373

186. Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers.Sun C, Fang Y, Yin J, Chen J, Ju Z, Zhang D, Chen X, Vellano CP, Jeong KJ, Ng PK, Eterovic AKB, Bhola NH, Lu Y, Westin SN, Grandis JR, Lin SY, Scott KL, Peng G, Brugge J, Mills GB. Sci Transl Med. 2017 May 31;9(392). pii: eaal5148. doi: 10.1126/scitranslmed.aal5148. PMID:28566428

187. Iavarone C, Zervantonakis IK, Selfors LM, Palakurthi S, Liu JF, Drapkin R, Matulonis UA, Hallberg D, Velculescu VE, Leverson JD, Sampath D, Mills GB, Brugge JS. Combined MEK and BCL-2/XL inhibition is effective in high-grade serous ovarian cancer patient-derived xenograft models and BIM levels are predictive of responsiveness. Mol. Cancer Therapeutics 2019 Mar;18(3):642-655. PMID:30679390
188. Isaac S. Harris, IS, Endress, JE, Coloff, JL, Selfors,LS, Rosenbluth, JR, Takahashi N, McBrayer SK, Koduri B, Oser MG, Schauer NJ, Doherty LM, Hong AL, Younger ST, Doench JG, Hahn WC, Buhrlage SJ, Kaelin WG, Brugge JS, Deubiquitinases maintain protein homeostasis and survival of cancer cells upon glutathione depletion. Cell Metabolism, 2019 Feb 13. pii: S1550-4131(19)30020-8. doi: 10.1016/j.cmet.2019.01.020. [Epub ahead of print]

189. Zoeller JJ, Vagodny A, Taneja K, Tan BY, O’Brien N, Slamon DJ, Sampath D, Leverson JD, Bronson RT, Dillon DA, Brugge JS, Neutralization of BCL-2/XL enhances the cytotoxicity of T-DM1 in vivo Mol. Can. Therapeutics, in press

.

Review Articles in Books or Journals
1. Brugge, J.S. and Chinkers, M. Tyrosine specific protein kinases. In: Annual Reports in Medicinal Chemistry, Vol. 18 (ed. H-J. Hess), Academic Press, New York, pp. 213-225 (1983).

2. Brugge, J.S. Interaction of the Rous sarcoma virus protein pp60src with the cellular proteins pp50 and pp90. In: Current Topics in Microbiology and Immunology, Vol. 123, Springer-Verlag, Berlin (1985).

3. Brugge, J.S. The p35/p36 substrates of protein-tyrosine kinases as inhibitors of phospholipase A2. Cell 46:149-150 (1986).

4. Golden, A. and Brugge, J.S. The cellular and viral src gene. In: The Oncogene Handbook Vol. 7 (eds. E.P Reddy, A.M. Skalka, and T. Curran). Elsevier Science Publishers, Amsterdam (1988).

5. Shattil, S.J. and Brugge, J.S. Protein tyrosine phosphorylation and the adhesive functions of platelets. Curr. Opin. Cell Biol., 3: 869-879 (1991).

6. Clark, E., and Brugge, J.S. Tyrosine phosphorylation in platelets and potential roles in intracellular signal transduction. Trends in Cardiovascular Medicine, 3: 218-227 (1993).

7. Brugge, J.S. New intracellular targets for therapeutic drug design. Science, 260: 918-919 (1993).

8. Shattil, S.J., Ginsberg, M.H., Brugge, J.S. Adhesive signaling in blood platelets. Curr. Opin. Cell Biol., 6: 695-704 (1994).

9. Clark, E.A., Shattil, S.J. and Brugge, J.S. Regulation of protein tyrosine kinases in platelets. Trends Biochem. Sci., 19: 464-469 (1994).

10. Clark, E.A. and Brugge, J.S. Integrin signal transduction pathways, the road taken. Science, 268: 233-239 (1995).

11. Clark, E.A. and Brugge, J.S. Tyrosine Phosphorylation. In: Platelet Function and Signal Transduction: A Practical Approach. (Watson, S. and Authi, K., eds) Oxford University Press, Oxford, England (1996).

12. Brugge, J.S., Clark, E.A., Shattil, S.J. Protein Tyrosine Phosphorylation in Platelets. In: Advances in Molecular and Cell Biology, “The Platelet”. (Bittar, E.E.ed) JAI Press, Greenwich, CT (1997).

13. Bolen, J.B. and Brugge, J.S. Leukocyte protein tyrosine kinases: potential targets for drug discovery. Annual Review of Immunology, 15: 371-404 (1997).

14. Thomas, S. and Brugge, J.S. Cellular functions regulated by Src family kinases. Annual Review of Cell and Developmental Biology. 13:513-609 (1997).

15. Brugge, J.S. Casting light on focal adhesions. Nature Genetics 19: 309-311 (1998).

16. Brugge, J.S. and McCormick, F. Cell Regulation: Intracellular Networking. Current Opinion in Cell Biology, 11: 173-176 (1999).

17. Miranti, C. Brugge, J.S. Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol. 4: E83-90.

18. Debnath, J, Muthuswamy, S.K., and Brugge, J.S. Morphogenesis And Oncogenesis Of MCF-10A Mammary Epithelial Acini Grown In Three-Dimensional Basement Membrane Cultures. Methods. Jul;30(3):256-68 (2003).

19. Seton-Rogers SE, Brugge JS. ErbB2 and TGF-beta: A Cooperative Role in Mammary Tumor Progression? Cell Cycle 3(5):597-600 (2004).

20. Shaw KR, Wrobel CN, Brugge JS: Use of three-dimensional basement membrane cultures to model oncogene-induced changes in mammary epithelial morphogenesis, J Mammary Gland Biol Neoplasia. Oct;9(4):297-310 (2004)

21. Debnath, J. and Brugge J.S. Modeling Epithelial Cancers in 3D Cultures, Nature Reviews Cancer 5: 675-688 (2005)

22. Walker SJ, Brugge JS. SePARating polarity and proliferation in ErbB2 oncogenesis. Nat Cell Biol 8 (11): 1220-2, 2006.

23. Carroll DK, Brugge JS, Attardi LD. p63, cell adhesion and survival. Cell Cycle 6 (3): 255-61, 2007.

24. Brugge, J.S., Hung, M.C., Mills, G.B. A New Mutational aktivation in the PI3K Pathway, Cancer Cell 12, 2007, 10-13

25. Mouneimne G, Brugge JS. Tensins: a new switch in cell migration. Dev Cell 13 (3): 317-9, 2007.

26. Schafer ZT, Brugge JS. IL-6 involvement in epithelial cancers. J Clin Invest 117 (12): 3660-3, 2007.

27. Overholtzer M, Brugge JS. The cell biology of cell-in-cell structures. Nat Rev Mol Cell Biol. 2008;9(10):796-809.

28. Mailleux AA, Overholtzer M, Brugge JS. [Entosis, a cell death process related to cell cannibalism between tumor cells.]. Med Sci (Paris) 24 (3): 246-248, 2008.

29. Mailleux AA, Overholtzer M, Brugge JS. Lumen formation during mammary epithelial morphogenesis: insights from in vitro and in vivo models. Cell Cycle 7 (1): 57-62, 2008.

30. Mouneimne G, Brugge JS. YB-1 translational control of epithelial-mesenchyme transition. Cancer Cell (2009) vol. 15 (5) pp. 357-9

31. Iwanicki MP, Brugge JS. Transcriptional regulation of metastatic [Id]entity by KLF17. Genome Biol. 2009;10(11):244.

32. Leung, C.T., and Brugge, J.S. (2009). Tumor self-seeding: bidirectional flow of tumor cells. Cell 139, 1226-1228.

33. Ng MR, Brugge JS. A stiff blow from the stroma: collagen crosslinking drives tumor progression. Cancer Cell. 2009;16(6):455-7.

34. Harrison SC, Brugge JS. Mechanistic biology in the next quarter century. Mol Biol Cell. 2010;21(22):3799-800. PMCID: 2982119.

35. Grassian AR, Coloff JL, Brugge JS. Extracellular Matrix Regulation of Metabolism and Implications for Turmorigenesis. Cold Spring Harb Symp Quant Biol. 2011 Nov 21. [Epub ahead of print]
PMID: 22105806.

36. Brugge JS. Rab25 Mediates Integrin Recycling for Tumor Cell Migration in 3D. Dev Cell. 2011 Nov 15;21(5):e1. PMID: 22075151

37. Albeck JG, Brugge JS. Uncovering a tumor suppressor for triple-negative breast cancers. Cell. 2011;144(5):638-40.

38. Gallegos L, Ng MR, Brugge JS. The myosin-II-responsive focal adhesion proteome: a tour de force? Nat Cell Biol. 2011;13(4):344-6.

39. Brugge JS. Into the deep: Refocusing on 3D. Nat Cell Biol. 2012;14(4):332.

40. Gallegos LL, Brugge JS. Live free or die: cell-cell adhesion regulates sensitivity to trail-induced apoptosis.Dev Cell. 2014 Jul 14;30(1):3-4.

42. Sever R and Brugge JS. Signal transduction in Cancer. Cold Spring Harb Perspect Med. 2015 Apr 1;5(4). PMID: 25833940

43. Harris IS and Brugge JS. Cancer: the enemy is my friend. Nature. 2015 Nov 12;527(7577). PMID 26503052

44. Coloff JL and Brugge JS. How tumor cells cope with the metabolic stress of leaving home Cell Res. 2016 Jul;26(7):757-8. PMCID: PMC5129879.

45. Coloff JL, Brugge JS. Metabolic changes promote rejection of oncogenic cells. Nat Cell Biol. 2017 Apr 27;19(5):414-415. doi: 10.1038/ncb3521. PubMed PMID: 28446818.

Chapters in Books
1. Yonemoto, W., Lipsich, L., Darrow, D. and Brugge, J.S. An analysis of the interaction of the Rous sarcoma virus transforming protein, pp60src, with a major heat shock protein. In: Heat Shock Proteins from Bacteria to Man (ed. Schlesinger, M.J., Ashburner, M., Tisseries, A.) Cold Spring Harbor Laboratory (1982).

2. Brugge, J.S., Darrow, D., Lipsich, L.A. and Yonemoto, W. The association of the transforming protein of Rous sarcoma virus with two cellular phosphoproteins. In: Oncogenes: Evaluation of Basic Findings and Clinical Potential (ed. R. Rauscher) Alan Liss, New York, pp. 135-148 (1982).

3. Brugge, J.S., Yonemoto, W., Lipsich, L. and Darrow, D. Studies on the interaction between the transforming proteins of avian sarcoma viruses and two cellular proteins. “Tumor Viruses and Differentiation”, (ed. Scolnick, E. and Levine, A.), Alan Liss, New York, pp. 399-410 (1983).

4. Lipsich, L.A., Yonemoto, W., Bolen, J., Israel, M., and Brugge, J.S. Structural and functional studies on the Rous sarcoma virus transforming protein. In: Cancer Cells 2: Oncogenes and Viral Genes (eds. Van de Woude, G.F., Levine, A.J., Topp, W.C., Watson, J.D.) Cold Spring Harbor, New York, pp. 43-52 (1984).

5. Yonemoto, W., Bolen, J.B., Israel, M.A., Lipsich, L. and Brugge, J.S. Use of monoclonal antibodies to probe the functional activity of the cellular src gene product in polyoma virus transformed cells. In: Monoclonal Antibodies and Cancer Therapy, Vol. 27, 551-565 (ed. R.A. Reisfeld and S. Sell (1985).

6. Coussens, P.M. and Brugge, J.S. Comparison of the structural and functional properties of the viral and cellular and src gene products. In: Induction and Recognition of the Transformed Cell (Eds. M.I. Greene and T. Hamaoka), Plenum Press, New York (1986).

7. Brugge, J.S. Regulation of the expression of the cellular src proto- oncogene product pp. 401-415 (eds. N.O. Kjeldgaard and J. Forchhammer). Alfred Benson Symposium 24 on Viral Carcinogenesis (1987).

8. Brugge, J.S., Yonemoto, W., Lustig, A., and Golden, A. Investigations of the expression of the cellular src gene product, pp. 241-252. In Oncogenes and Cancer. Proceedings of the International Symposium of the Princess Takamatsu Cancer Research Fund, S.A. Aaronson et al. (Eds.), Japan Sci. Soc. Press, Tokyo/VNU Press, Utrecht, (1987).

9. Brugge, J.S. Expression of the c-src proto-oncogene product in neural cells, pp. 213-226 In: NATO Research Workshop on Cellular and Molecular Aspects of Neural Development and Regeneration, Vol. 22 (A. Gorio et al., eds.) Springer Verlag (Heidelberg) (1988).

10. Brugge, J.S. and Levy, J.B. Investigations of a variant form of the tyrosine protein kinase, pp60c-src in neuronal cells. In: “Protein Phosphorylation and Neuronal Function, (eds. E. Costa and E. Barnard) Raven Press, NY (1989).

11. Grassian AR, Coloff JL, Brugge JS. Extracellular Matrix Regulation of Metabolism and Implications for Tumorigenesis. Cold Spring Harbor Symposia on Quantitative Biology. 2011

Books
Brugge, J., Curran, T., Harlow, E., McCormick, F. Origins of Human Cancer: A Comprehensive Review. Cold Spring Harbor Press, New York.

Invited Lectures at Meetings

1984
University of Pennsylvania Symposium on Cellular Oncogenes

1985
UCLA Symposium on Monoclonal Antibodies and Cancer Therapy, Park City, UT
West Coast Workshop on Oncogenes and Their Products, Lake Tahoe, CA
Workshop on Oncogenes, Tissue Culture Association Meeting, New Orleans, LA
FASEB Summer Conference on Mechanisms of Carcinogenesis, Vermont
Chairman – Cold Spring Harbor RNA Tumor Virus Meeting

1986

Alfred Benson Symposium on Viral Carcinogenesis, Copenhagen, Denmark
FASEB Summer Conference on Membrane Receptors, Vermont
Cell Growth and Differentiation, Rensselaersville, NY (New York Acid Technologies Foundation Conference)
17th International Symposium of the Princess Takamatsu Cancer Research Fund, Tokyo, Japan

1987

Oncogenes and Growth Factors, European Molecular Biology Organization, Heidelberg, Germany
NATO Workshop: Cellular and Molecular Aspects of Neural Development and Regeneration,
Miami, FL
Special Session on Oncogenes, Annual Meeting of the American Association of Neurosurgeons,
Dallas, TX
Session Chairperson: 3rd Annual Meeting on Oncogenes, Frederick, MD
FASEB Meeting on Mechanisms of Carcinogenesis, Saxtons River, VT
Review and Update of Neurobiology, Session on Oncogenes in Neural Development, Woods Hole, MA
Symposium on Proto-oncogenes in Neural Cells, Annual Meeting of American Association of Neurosciences, New Orleans, LA

1988

Symposium on Receptors and Growth Factors–Annual Meeting of Federation of American Society of Experimental Biologists, Las Vegas, Nevada
Chairperson: Session on src-related tyrosine kinases, Annual Meeting on Oncogenes, Frederick, MD
Cellular & Molecular Aspects of the Early Events in Neurogenesis, Jacques Monod Conference, Brittany,France
Protein Phosphorylation & Neuronal Function, Fidia Research Foundation Symposium, Toronto, Canada

1989

CIBA Foundation Symposium on Proto-Oncogenes in Development, London, England
FASEB Meeting on “Molecular Carcinogenesis,” Copper Mountain, Colorado
Gordon Conference on “Neural Plasticity,” Brewster Academy, New Hampshire
NATO Conference on “Regulation of Cellular Growth,” Mallorca, Spain
National Cancer Institute Workshop on “Tumor Antigens & Proto-oncogene interactions,” Gaithersburg, Maryland
University of North Carolina Symposium on “Viruses and Cancer,” Chapel Hill, North Carolina

1990

Symposium on Tyrosine Kinases: The First Decade, Salk Institute, San Diego, California
Princeton University Seminar, Department of Molecular Biology, Princeton, New Jersey
Guest Lecturer: Cold Spring Harbor Course on the Molecular Genetics of Nervous System Diseases
International Society for Differentiation Sixth Annual Conference on Differentiation of Normal and Neoplastic Cells, Vancouver, Canada
Ninth Summer Symposium: Molecular Pathways of Cell Growth Control, Pennsylvania State University, University Park, Pennsylvania
Chairman: Session on src protein: 6th Annual Oncogene Meeting, Frederick, Maryland

1991

Gordon Conference: Fibronectin and Cell Matrix Proteins, February, 1991
Symposium: Molecular Mechanisms of Signal Transduction, SUNY Stony Brook, May 22-25, 1991
FASEB Conference: Mechanisms of Carcinogenesis, July 21-26, 1991
Cold Spring Harbor Course: “Cloning of Neural Genes”, July 11, 1991
European Molecular Biology Organization Symposium on Mechanisms of Signal Transduction,Heidelberg, Germany, Sept. 15-19, 1991

1992

Keystone Conference: Positive Growth Control, January, 1991
Cold Spring Harbor Symposium on Quantitative Biology, May, 1992
Gordon Conference on Hemostatis, June, 1992
Gordon Conference on Hormone Action, August, 1992
Symposium on Growth Factors, Lake Placid, NY, September, 1992

1993

Winter Immunology Conference, Asilomar, CA, January, 1993
Keystone Conference on Cell Adhesion, Keystone, CO, January, 1993
Gordon Conference on Molecular Pathology, March 1993
Symposium: Integrins and Cell Signaling, University of North Carolina, April 1993
Massachusetts Biotechnology Council Symposium on Structure Based Drug Design, April 1993
Ninth Annual Oncogene Meeting – Chairman of Session, Baltimore, MD, June, 1993
Gordon Conference on Cell Contact and Adhesion, June, 1993
Gordon Conference on Molecular and Genetic Basis for Cell Proliferation, Meriden, NH, July 1993
Science Innovation Conference on New Research Techniques, Boston, MA, August 1993
Gordon Conference on Fertilization and Activation of Development, Plymouth, NH, August 1993
International Society for Neurochemistry Biennial Meeting, Montpellier, France, August 1993
Symposium: Cell Matrix Interaction, ASBMR Fifteenth Annual Meeting, Tampa FL, September 1993
Ben May Institute Symposium on Signal Transduction , Chicago, IL, October, 1993
Forbeck Research Foundation Forum on Growth Factors, Hilton Head, SC, October 1993
Symposium: Cancer Research, Bristol Meyers Squibb, Los Angeles, CA, November 1993
Symposium: Tyrosine Kinase Pathways, American Society of Nephrology, Boston, MA, November 1993

1994

Symposium: Keystone Conference on Transmembrane Signal Transduction, Keystone, CO, February, 1994
Symposium: Keystone Conference on Biology of Physicochemical Interactions at the Cell Surface, Taos, NM, February 1994
Symposium: Keystone Conference on Molecular Basis of Cancer Therapy, Tamarron, CO, March, 1994
Symposium: Oncogenes and Growth Control, EMBO, Heidelberg, Germany, April 1994
Symposium: Signal Transduction Targets for Drug Discovery, Harvard Medical School, Boston, MA, May, 1994
Symposium: New Strategies for Drug Design, SUNY StonyBrook, NY, May 1993
Gordon Conference on Second Messengers, Kimball Union Academy, Meriden, NH, June,1994
Gordon Conference on Hemostasis, Proctor Academy, Andover, NH, June, 1994
10th Annual Oncogene Meeting, Session Chairperson, Hood College, Frederick, MD, June, 1994
Gordon Conference on Cardiac Regulatory Mechanisms, Salve Regina University, Newport, RI, July 1994
Symposium: Cancer & Development, Chairperson, MGH Cancer Center, Boston, MA, September 1994
Nature Conference on Immune System: A Model Organ, Boston, MA, November 1994
American Heart Association’s 11th Annual Conference on Thrombosis & Hemostasis, Dallas, TX, Symposium: Chairperson and Speaker, Protein-Protein Interactions in Signal Transduction, American Society for Cell Biology 34th Annual Meeting, San Francisco, CA, December 1994
Symposium: Speaker, Cell Adhesion and Signaling, Americal Society for Cell Biology 34th Annual Meeting, San Francisco, CA, December 1994

1995

Symposium: Session Chairperson and Speaker, Keystone Conference on Oncogenes: 20 Years Later Keystone, CO, January 1995
Session Chairperson, Conference on Tyrosine Phosphorylation and Cell Signaling, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, May 1995
Symposium: Chairperson and Speaker, Signaling from the Extracellular Matrix, 77th Annual Meeting of the Endocrine Society, Washington, DC, June 1995
Gordon Conference on Medicinal Chemistry, Session on Signal Transduction targets, Speaker, August, 1995
Symposium on Signal Transduction Whitehead Institute, Mass. Inst. of Technology, Session Chairperson , Oct., 1995
Symposium for Medical Research, Jane Coffin Childs Memorial Fund, Speaker, Oct., 1995
Colloquium on Life Sciences, Society and Industry, NYAS and Carnegie Foundation, Session on Implementations of Innovations, Speaker, Oct., 1995

1996

Symposium on Protein Phosphorylation and Cancer, Harvard Medical School, Speaker, Jan.,1996
Miami Bio/Technology Winter Symposium, Therapeutic Strategies in Molecular Medicine, Speaker, Feb., 1996
Am Ass’n of Neurochemistry Annual Meeting, Symposium on Cell Adhesion, March, 1996
Keystone Symposium on Signal Transduction Through Tyrosine Kinases, Speaker, March,1996
Am Soc for Biochem. and Mol. Biology, Chairperson and speaker, Symposium: Nonreceptor Protein tyrosine kinases, June, 1996
Oncogene Meeting, Session Chairperson
Harvard School of Medicine, Pathology Department Retreat, Keynote address, July, 1996
Roussel Institute, Table Ronde, Signal Transduction, July, 1996
Massachussetts General Hospital Cancer Center Retreat, Keynote address, September, 1996
Strategic Research Institute, Signal Transduction Meeting, Keynote address, September, 1996
M.D. Anderson Cancer Center 49th Annual Research Symposium on Regulatory Mechanisms in Growth and Differentiation, Speaker, October 1996
Inflammation Research Association 8th International Conference, Speaker, October, 1996
American Society for Cell Biology 6th International Congress on Cell Biology, Plenary Symposium Chairperson and speaker, December, 1996

1997:

Keystone Conference an Signaling through Cell Adhesion Receptors, Keystone Co.
Symposium on Oncogenes and Protein Phosphorylation, Rockefeller University, NYC,NY
Gordon Conference on Cell Growth and Proliferation, Colby State College, N.H.
FASEB conference on Protein Kinases and Phosphatases, Snowmass, CO.
Table Ronde Conference on Cell Adhesion, Paris, France
Scripps Research Institute, San Diego, CA

1998

Pew Foundation Scholars meeting, Keynote Address, Puerto Vallerata, Mexico
Keystone Conference on Extracellular Matrix Signaling, Steamboat Springs CO
Gordon Conference on Vascular Biology, NH
Gordon Conference on Cell Proliferation, NH

1999

Nature Biotechnology Winter Symposium, Miami FL
Keystone Conference on Oncogene Networks in signal Transduction, Keystone CO
Cold Spring Harbor Meeting on Tyrosine Phosphorylation, CSH, NY
FASEB meeting on Protein Phosphorylation and Second Messengers, Kimball Union Academy, NH
Gordon Conference, Molecular Biology of the Cell, Tilton Academy, NH
FASEB meeting on Protein Kinases, Co-organizer, Snowmass CO
Gordon Conference on Cell Structure and Gene Expression, Kimball Union Academy, NH
BatSheva de Rothschild Symposium on The Dialogue between Cell Adhesion, Protein Degradation and Transcriptional Regulation in Cancer, Dead Sea, Israel
ASCB, MiniSymposium Chairman and Speaker, Washington DC

2000

Keystone Symposium on Dynamics of the Cytoskeleton, Keystone Colorado
Keystone Symposium on Regulation of Signaling Pathways by Integrins and Growth Factors, Keystone CO
Gordon conference on Second Messengers & Protein Phosphorylation, Meriden, NH
Dubrovnik Confernce on Signal Transduction , Dubrovnik, Croatia
Gordon Conference on Signaling through Adhesion Receptors, Newport RI
Cold Spring Harbor Meeting on Cancer Genetics and Tumor Suppressor Genes
Albert Einstein on Epithelial-Stromal Interactions and Tumor Progression Workshop
ASCB , San Francisco CA

2001

Gordon Conference on Fibronectin, Integrins & Related Molecules
American Society for Biochemistry and Molecular Biology, Orlando FL
HHMI Workshop on Signal Transduction, Chevy Chase, MD
2nd Messengers and Protein Phosphorylation , Melbourne Australia
Seventeenth Annual Meeting on Oncogenes:Cancer Cell Signal Trasduction, Frederick, MD
Gordon Conference on Second Messengers & Protein Phosphorylation, Kimball Union Academy, NH
FASEB Meeting on Protein Kinases, Snowmass CO
American Cancer Society Symposium, San Diego CA
Migration and Metastasis Workshop, Washington DC
ASCB, Washington, DC

2002

APCR Workshop, Bethesda MA
Gordon Conference on Adhesion Signaling
Cancer Intervention, Van Andel Institute
Astra-Zeneca Growth Factors and Cancer Symposium
American Society for Matrix Biology, Symposium speaker
ASCB, San Francisco, CA

2003

Gordon Conference on Molecular Pharmacology
Keystone Symposium on Signaling Via Cell-Cell Interactions
American Society for Matrix Biology
American Society for Biochemistry & Molecular Biology
American Association for Cancer Research Annual Meeting Opening Symposium “New Opportunities for Accelerating the Conquest of Human Cancer”
Gordon Conference on Epithelial Differentiation & Keratinization
FASEB Summer Research Conference: Protein Phosphoryation
International Congress of Biochemistry & Molecular Biology
The Salk Institute: Oncogenes and Growth Control Meeting
Gordon Research Conference – Epithelial Cell Differentiation
Gordon Research Conference – Mammary Gland Biology
AACR meeting on Breast Cancer Molecular Pathogenesis
Diabetes Genome Anatomy Project PPG meeting Woods Hole
ACS Professors Meeting
Breast Cancer SPORE Roundtable
ASCB, San Francisco

2004

AACR Meeting on Advances in Cancer Research Hawaii
Keystone Symposium on Signaling in Vertebrate Organogenesis Santa Fe NM
American Society for Biochemistry and Molecular Biology, Annual Meeting Boston MA
Annual Meeting on Oncogenes Frederick MD
Gordon Conference on Signaling by Adhesion Receptors Rhode Island
Gordon Research Conference on Growth Factor Signaling Keynote Speaker Oxford England
12th International Conference on Second Messengers & Phosphoproteins Toronto Canada
57th Annual Symposium on Fundamental Cancer Research
52nd Annual Scientific Meeting American Association of Cytopathology, Basic Science Award Lecture Chicago Illinois
Plenary Symposium, ASCB, Washington, DC

2005

AACR Special Conference on Advances in Breast Cancer Research: Genetics, Biology and Clinical Applications
Second International Meeting on Tumor Progression and Therapeutic Resistance Boston MA
ASCB Engineering Cell Biology, Keynote Speaker, Seattle, WA
Beatson Int’l Cancer Conference, Glasgow, Scotland
Cold Spring Harbor Symposium on Quantitative Biology, CSH, NY
General Motors Cancer Research Symposium Washington DC
Era of Hope Breast Cancer Meeting, Keynote speaker, Phil.PA
AACR Charlotte Friend Memorial Lecture, Los Angeles, CA
Keystone meeting on Molecular Targets for Cancer, Santa Fe NM
Keystone meeting on Tumor Microenvironment Banff, Alberta Canada
Keystone meeting on Cell Migration and Adhesion, Snowbird Utah
Chicago Signal Transduction Symposium, Chicago Ill
Cell Press Symposium on Growth Control, Boston MA
Breast Cancer Research Foundation Symposium, NYC, NY
International Assn of Breast Cancer Research, Montreal, CA
Symposium on Winning the War Against Cancer; From Genomics to Bedside and Back, Grand Rapids, MI
Mammary Gordon Conference, Lucca, Italy
American Assn’ Breast Cancer Research Annual Meeting, Symposium

2007

Nature Conference: Cancer Therapeutics: The Road Ahead, Capri, Italy
FASEB Conference on Growth Factor Receptor Tyrosine Kinases in Mitogenesis, Morphogenesis and Tumorigenesis, Tucson, AZ
Weekend to End Breast Cancer International Workshop, Toronto, Ontario
American Association for Cancer Research annual meeting
Chair and speaker, major symposium: Tumor Microenvironment
Chair and speaker, Methods workshop: Three-Dimensional Culture Models
IDEAS Boston conference
Keystone Symposium on Apoptotic and Non-Apoptotic Cell Death Pathways
Baylor College of Medicine, Virology Dept. Reunion
American Association for Cancer Research/Japan Cancer Association Joint Meeting
FEBS workshop on Invadopodia, Podosomes and Focal Adhesion, Keynote Lecture Ciente, Italy
Irish Cancer Society Lectureship

2008

Keystone Symposium on Cell Death and Sensescence, Breckinridge, CO
American Association for Cancer Research, San Diego, CA; Tumor Microenvironment Symposium, Speaker and Chair, Triple Negative Breast Cancer Chair
Ibsen Foundation, Cancer Metabolism Meeting, Costa Rica
Mammary Gland Biology, Gordon Research Conference, Lucca, Italy

2009

European Organization Research Threatment Cancer 2009 Boston MA
AACR Advances in Breast Cancer Research San Diego CA
AACR Frontiers in Cancer Research, Boston MA
Damon Runyon Fellows’ Retreat, Dennis MA
European Cell Death Organization Conference, Paris France
Les Treilles Cancer: from Basic Rearch to Clinical Benefits Provence France
GRC Cell Contact and Adhesion Waterville Valley Resort,MA
Mammary Gland Biology GRC Newport, RI
AACR Annual Meeting Symposium Speaker Denver Colorado
Cell Death Pathways Keystone Conference Whistler, CA
AACR Translation of the Cancer Genome Conference
Keystone Symposia Omics Meets Cell Biology, Keystone CO

2010

AACR/MRS Joint Meeting Tumor Microenvironment Philadelphia PA
GRC on Signaling by Adhesion Receptors, Colby College
AACR Annual Meeting Washington DC
Cell Death Pathways: Apoptosis, Autophagy and Necrosis/Metabolism and Cancer, Vancouver CA

2011

AACR Special Conference on Advances in Breast Cancer Research: Genetics, Biology, and Clinical Applications, October 12-15, 2011
TOR and PI3K meeting Basel Switzerland September 2011
Salk Institute Mechanisms and Models August 2011
GRC Epithelial Differentiation and Keratinization Mt. Snow, VT. July 2011
Cold Spring Harbor Symposium on Quantitative Biology Energy, Metabolism, the Cell Cycle, and Cancer
Frontiers in Cancer Research, Boston MA
AACR Annual Meeting Orlando FL April 2011
Keystone Symposium on Epithelial Cancers and Stem Cells Keystone Colorado 2011
Keystone Symposium on PI-3-Kinase Signaling Pathways, 2011
BCRF’s Palm Beach Breast Cancer Symposium

2012
AACR Cancer Annual Meeting Cancer Metabolism Symposium, Chicago Ill
Keystone meeting on Cancer and Metabolism, Banff Alberta
Center for Cancer Research Eminent Lecturer NCI Bethesda MD.
Gordon Research Conference – Cell Adhesion Receptor Signaling
CSH Mechanism & Models of Cancer
27th Annual Aspen Cancer Conference
MD Anderson Postdoctoral Association Symposium
Pfizer Annual Postdoctoral Symposium
NCI workshop on Tumor Models, Philadelphia PA
ASCB Annual Meeting Invited speaker Special Interest Symposium on Migration and Invasion

2013

Keystone meeting on Cancer Metabolism Keystone CO,
AACR Annual Meeting, Drug Resistance Symposium
AACR Annual Meeting, Keynote talk WICR Professional Advancement Mentoring presentation Princeton, Governor’s Conference, Princeton, NJ Cold Spring Harbor, Cancer Biology and Therapeutics, Cold Spring Harbor, NY Hippo Tumor Suppressor Meeting, Monterey, CA, Keynote Gordon Research Conference, Cell Growth and Proliferation, Mt. Snow, VT
AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics, Boston MA
AACR Frontiers in Basic Cancer Research 2013
GIRC International Congress Invitation – Resistances to Targeted Therapies meeting, Grenoble France
Society for Melanoma Research Keynote presentation Philadelphia PA
NCI workshop on Tumor Cell Heterogeneity, Vanderbilt Medical School
Forbeck Forum, Forbeck Foundation, Hilton Head South Carolina

2014

14th Annual E.E. Just Scientific Symposium
Hunter Cell Biology meeting – Keynote Plenary lecture, Australia April 2014,
AACR annual meeting: Meet the Professor presentation
Aspen Cancer conference: Session chair and talk- drug resistance
AACR special conference chair PI3K, Co-Chair, session chair, talk
Gordon Research conference on Cell Death, session chair and talk
AACR Precision Medicine Series: Drug Sensitivity and Resistance: Improving Cancer Therapy
Keynote Plenary lecture Hunter Cell Biology meeting –, Australia
San Antonio Breast Cancer meeting – Brinker Award lecture presentation

2015
Keystone Symposium: “The Biological Code of Cell Signaling – A Tribute to Tony Pawson”
AACR NCI EORTC conference on Molecular Targets and Cancer Therapeutics
Astellas Science Day presentation

2016
CSHL Symposium on Quantitative Biology addressing Targeting Cancer
LINCS Conference NIH

2017

AACR Annual Meeting – Session on Adaptive Resistance to Therapy
Breast Cancer Think Tank
Weizmann Institute Symposium
AACR Meeting Addressing Critical Questions in Ovarian Cancer Research Keynote speaker
Aspen Cancer Conference
Gordon Research Conference on Mammary Biology
University of Notre Dame Symposium, Bridging the Gap from Bench to Bedside

2018
Breast Cancer Think Tank
FASEB meeting on Signaling in Cancer- Keynote Lecture
Gordon Conference on Adhesion Signaling – Keynote Lecture
Mechanisms and Models of Breast Cancer – Keynote Lecture, Cold Spring Harbor Laboratory,

Invited Seminars (since 1984):

1984

University of Texas, Department of Biology, San Antonio, TX
Baylor College of Medicine, Department of Virology, Houston, TX
St. Jude’s Research Center, Memphis, TN
University of Miami School of Medicine, Department of Physiology and Biophysics, Miami, FL
Harvard Medical School, Department of Microbiology, Cambridge, MA

1985

Oncogene, Inc., Seattle, WA
Rockefeller University, New York City, NY
College of Physicians and Surgeons, Department of Microbiology, New York City, NY
University of Tennessee, Department of Microbiology, Knoxville, TN
University of Massachusetts School of Medicine, Department of Biochemistry, Worcester, MA
Brookhaven National Laboratory, Biology Department, Brookhaven, NY
Princeton University, Department of Molecular Biology, Princeton, NJ
Columbia University, Department of Biological Sciences, New York City, NY

1986

University of North Carolina, Chapel Hill, NC
Columbia University, College of Physicians and Surgeons, New York City, NY
Sloan Kettering Memorial Cancer Institute, New York City, NY
Pennsylvania State University, University Park, PA
University of California School of Medicine, Los Angeles, CA
University of California School of Medicine, San Francisco, CA
Albany Medical School,Albany, NY
Rockefeller University, New York City, NY

1987

Boston University School of Medicine, Department of Biochemistry, Boston, MA
Harvard University, Department of Cellular and Molecular Biology, Cambridge, MA
Harvard Medical School, Department of Genetics, Cambridge, MA
Rutgers University School of Medicine, New Brunswick, NJ
University of Colorado Health Sciences Center, Denver, CO
Washington University School of Medicine, St. Louis, MO
Fox Chase Cancer Center, Philadelphia, PA
Frederick Cancer Research Facility, Frederick, MD
Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
National Institutes of Health, Bethesda, MD
Giessen Institute for Research, Giessen, Germany
Southwestern Medical School, Dallas, TX
Harvard School of Public Health, Cambridge, MA

1988

Cleveland Clinic Institute, Cleveland, OH
University of Rochester, Rochester, NY
Harvard Medical School, Department of Microbiology, Cambridge, MA
University of Miami School of Medicine, Miami, FL
Syntex Research Center, Palo Alto, CA
University of California at Irvine School of Medicine, Irvine, CA
University of Cincinnati School of Medicine, Cincinnati, OH
University of Pennsylvania School of Medicine, Philadelphia, PA
Fox-Chase Institute for Cancer Research, Philadelphia, PA

1989

Ludwig Institute for Cancer Research, Montreal, Canada
Columbia University College of Physicians and Surgeons, New York, NY

1990
Northwestern University, Evanston, IL
University of Virginia School of Medicine, Charlottesville, VA
Burroughs Welcome Research Laboratories, Research Triangle Park, NC
Jefferson University, Philadelphia, PA
Pennsylvania State University School of Medicine, University Park, PA
National Jewish Research Institute, Denver, CO
Mayo Clinic Research Foundation, Rochester, MI
Department of Genetics, Baylor College of Medicine, Houston, TX

1991

National Institutes of Health, Eye Institute, Bethesda, MD
Department of Molecular Biology, Rhone-Poulenc Rorer, Inc., King of Prussia, PA
Department of Molecular Biology, University of Michigan School of Medicine, Ann Arbor, MI
Rutgers University Center for Advanced Biotechnology and Medicine, Piscataway, NJ
University of Vermont School of Medicine, Berlington, VT
Villanova University, Philadelphia, PA
Yale University School of Medicine, New Haven, CT
Medical College of Ohio, Toledo, Ohio
University of Delaware, Wilmington, DE

1992

Rockefeller University, New York, NY
Washington University School of Medicine, St. Louis, MO
Fox Chase Cancer Research Institute, Philadelphia, PA
Tufts University, Boston, MA
Harvard University, Boston, MA
Massachusetts General Hospital Cancer Center, Boston, MA

1993

University of California, San Francisco
University of Southern California School of Medicine, Pasadena, CA
University of North Carolina at Chapel Hill, Chapel Hill, NC
Dana Faber Cancer Institute, Boston, MA

1994

Harvard Medical School, Cambridge, MA
University of Massachusetts Medical Center, Worcester, MA

1995

National Institutes of Health: Directors Seminar, Bethesda, MD
New York University Medical Center: Honors Program Lecture, New York, NY
Genetics Institute, Cambridge, MA

1996

Roussel Institute, Paris, France
St. Elizabeth’s Hospital, Tuft’s University Medical School, Boston, MA
Southwestern Medical Center, University of Texas, Dallas, TX, Bashour Distinguished Lecturer
Mt. Sinai School of Medicine, New York City, NY, Dean’s Lecture Series
Massachussets Institute of Technology, Department of Biology, Boston, MA
Harvard Medical School, Department of Cell Biology, Boston, MA

1997

Boston Biomedical Research Institute, Boston, MA
Scripps Research Institute, San Diego, CA

1998
Distinguished Lectureship, Lawrence Berkeley Laboratory, Berkeley, CA
Sloan Kettering Cancer Institute, NNY, NY.
Moffat Cancer Center, University of S. Florida, Tampa, FL
Vollum Institute, University of Oregon Health Sciences Center, Portland Oregon
Harvard School of Public Health, Boston MA
Yale University School of Medicine, New Haven CO
Birnham Institute, San Diego CA

1999

University of Pennsylvania School of Medicine, Philadelphia PA
Brandeis University, Waltham, MA
Purdue University. Lafayette, IN
University of North Carolina, Chapel Hill, NC
Stanford University, McCormick Distinguished Lectureship, Palo Alto CA

2000

Vollum Institute, Portland, Oregon
Columbia University, New York, NY
MD Anderson Cancer Center, Houston TX
University of Colorado, Boulder, CO
Massachusetts General Hospital, Boston, MA
Jackson Laboratories, Bar Harbor MA
University of California, Berkley, Berkeley, CA
Cornell University, Ithaca NY

2001

Brigham and Women’s Hospital, Dept of Genetics, Boston MA
Center for Blood Research, Harvard Medical School, Boston MA
Beth Israel-Deaconess Hospital, Dept. of Pathology, Boston MA
Kathleen Robison Huntsman Distinguished Lectureship University of Utah, Salt Lake City UTAH
Duke University School of Medicine, Durham, NC
Biogen, Boston MA

2002

Taurog Lectureship, UT Southwestern, Dallas Texas
University of California at San Francisco, San Francisco CA
University of California at San Diego, San Diego CA
Northwestern University School of Medicine, Chicago IL
Cell Signalling, Inc. Beverly MA
University of North Carolina, Chapel Hill NC
Harvard Medical School, Dept of Pathology
Washington University St. Louis MO
SUNY StonyBrook
Pennsylvania State University
MGH Course on Epithelial Cell Biology
Institut de Recherches Cliniques de Montreal
Temple U School of Medicine Pathology & Laboratory Medicine
University of Pennsylvania –Biomedical Graduate Studies Retreat, plenary speaker
Vanderbilt Ingram Cancer Center
Penn State College of Medicine: Department of Neuroscience & Anatomy

2003

Yale University Molecular Biophysics & Biochemistry Department
The Scripps Research Institute Department of Cell Biology
SUNY at Stony Brook
Tufts U School of Medicine: Sackler School of Graduate Biomedical Sciences: Schmidt Distinguished Lectureship
Marine Biology Laboratory: Forkosh-Waxler Lecture in Physiology
University of California at San Francisco Women in Life Sciences Lectureship
MGH Cutaneous Biology Research Center
HHMI Outreach Seminar for High School Teachers
Genentech Pharmaceuticals

2004

University of Alabama Birmingham Alabama
St. Jude’s Children’s Research Hospital, Memphis,Tennessee
M.D. Anderson Cancer Seminar, Experimental Therapeutics Seminar Series
Duke University Medical Center, Durham, North Carolina
Beth Israel Deaconess Medical Center
NIH Directors Lecture Series, Bethesda, MD
Fox Chase Cancer Center, Philadelphia, PA
Memorial Sloan Kettering Cancer Center, New York, NY
Albert Einstein College of Medicine, New York, NY
University of New Mexico School of Medicine, Albuquerque NM
University of California at San Francisco, San Francisco, CA
Cambridge University, Cambridge England
Fred Hutchinson Cancer Research Institute, Seattle WA

2005
Massachusetts Institute of Technology, Biology Colloquium Seminar, Boston MA
National Cancer Institute Rosalind Franklin Award Lecture, Washington, DC
University of Pennsylvania, Philadelphia, PA
UMDNJ – Robert Wood Johnson Medical School, Rutgers, NJ
Brigham & Women Hospital Renal Division, Boston, MA
Institute for Breast Cancer Research, Toronto, ON
Duke University Graduate School, Durham NC

2006

University of Nebraska: Eppley Cancer Institute Omaha, Nebraska
Stanford University School of Medicine: Palo Alto, CA

2007

London Research Institute, London, England
Northwestern University, Evanston Ilinois
Albert Einstein College of Medicine, NYC, NY
National Cancer Institute Center for Cancer Research, Grand Rounds

Scholars Lecture In Physiology, Marine Biological Laboratory, Wood Hole, MA
Drexel University College of Medicine Walter Rubin Memorial Keynote Lecturer Philadelphia, PA
University of North Carolina, Steelman Lectureship, Chapel Hill, NC

2008

University of California-San Francisco
Department of Pathology, Harvard Medical School
Tufts University, Sackler School of Graduate Biomedical Sciences
UCLA David Geffen School of Medicine Annual Lecturer 2008
IRCM Boehringer-Ingelheim Lectureship, Montreal
University of Virginia Distinguished Lectureship 2008

2009

Novartis
Broad Metabolism initiative
Keystone in Whistler
Epithelial Morphogenesis, Homeostasis and Oncogenic Transformation
Weizmann Institute
Astra Zeneca
University of Massachusetts

2010

Sloan-Kettering Institute
Weill Medical College
BCRF’s Palm Beach Symposium and Luncheon
Northeastern University colloquium series
Princeton University
University of Colorado Health Sciences Center
University of Cincinnati School of Medicine
UCSF School of Medicine

2011

Yale University School of Medicine
University of Alabama School of Medicine
Vanderbilt University School of Medicine
Queen’s University, Belfast, Ireland
University of Pennsylvania School of Medicine
Wistar Institute, Philadelphia, Pennsylvania

2012

UCSD School of Medicine
UCLA David Geffen School of Medicine
Stanford University School of Medicine
David. M. Kovitz Visiting professorship University of Calgary, CA
Mary Bartlett Bunge Lectureship University of Miami Miller School of Medicine

2013

University of Virginia School of Medicine
University of North Carolina School of Medicine
Duke University School of Medicine
University of Arizona College of Medicine, Tucson, AZ
Johns Hopkins Medical Institute, Bard Lecture, Baltimore, MD
Massachusetts General Hospital Cancer Center, Boston, MA
University of Southern California, Pasadena CA
Cedars Sinai Hospital, Los Angeles CA

2014

Mt Sinai Schol of Medicine, NYC, NY
Herbert Irving Comprehensive Cancer Center¹s Distinguished Seminar
9th Annual Robert L. Sinsheimer Distinguished Lecture
OSHU Center for Spacial Systems Biomedicine Annual Retreat Keynote lecture
2nd Annual WISAY Distinguished Woman Scientist Award Lecture
Cell Signalling seminar

2015

Congressional Briefing on Cancer Precision Medicine
BIDMC Cancer Center Distinguished Lecture Distinguished Speaker
Landry Cancer Symposium

2016

Astra Zeneca Pharmaceuticals
Stanford Medical School
Van Andel Research Institute

2017

University of Cincinnati Medical School
University of Florida Medical School
AbbVie Pharmaceutical
Yale College of Medicine
University of Colorado Health Science Center Dean’s Distinguished Lectureship
Fox Chase Cancer Center
Yale University School of Medicine
Notre Dame University
Keynote address: AACR Special Conference: Addressing Critical Questions in Ovarian Cancer Research and treatment

2018
MRC Cancer Unit’s Annual Lecturer, University of Cambridge
2018 Jeffrey M. Trent Lecture in Cancer Research
Beatson Institute
University of Pennsylvania School of Medicine
Duke University
Fred Hutchinson Cancer Center, Distinguished Lectureship, July 2018
CSHL Mechanisms and Models of Cancer, Keynote lecture, August 2018
Symposium Celebrating the Scientific Career of Robert Goldman, Northwestern Medical School, September 2018
Lippard Lectureship, MIT and MGH, November 2018
MD Anderson Cancer Symposium, Keynote Lecture, October 2018