2010 - 2001 Awardees

Below is a list of the distinguished Horwitz Prize awardees from 2010 to 2001.

2010

  • Thomas J. Kelly, MD, PhD

    • Director, Sloan-Kettering Institute
    • Memorial Sloan-Kettering Cancer Center

    Thomas J. Kelly, MD, Ph.D. Thomas J. Kelly is director of the Sloan-Kettering Institute at the Memorial Sloan-Kettering Cancer Center, where he oversees a broad research program focused on the causes, diagnosis, and treatment of cancer. Prior to joining Sloan-Kettering in 2002, Dr. Kelly was professor and director of the Department of Molecular Biology and Genetics at the Johns Hopkins University School of Medicine and the founding director of the Johns Hopkins Institute for Basic Biomedical Sciences.

    Dr. Kelly received his PhD in biophysics from the Johns Hopkins University and his MD from the Johns Hopkins University School of Medicine. After postdoctoral studies at Johns Hopkins, where he determined the DNA sequences recognized by restriction enzymes, he conducted research on animal viruses at the National Institutes of Health as a member of the U.S. Public Health Service. He joined the faculty of the Johns Hopkins University School of Medicine in 1972. Dr. Kelly’s research has focused on how the genome is duplicated during the cell cycle, with particular emphasis on the ways DNA replication is initiated and controlled. Dr. Kelly’s laboratory developed the first cell-free systems for studying the biochemistry of DNA replication in human cells.

    Dr. Kelly was the co-recipient (with Bruce Stillman) of the 2004 Alfred P. Sloan Jr. Prize of the General Motors Cancer Research Foundation. He is a Member of the American Academy of Arts and Sciences, the National Academy of Sciences, the Institute of Medicine, and the American Philosophical Society.

    Dr. Kelly has served on many national advisory boards and is currently a member of the Advisory Committee to the Director, NIH, and the Scientific Management Review Board, NIH.

  • Bruce Stillman, PhD, FRS

    • President, Cold Spring Harbor Laboratory

    Bruce Stillman, Ph.D., F.R.S.A native of Australia, Bruce Stillman is a graduate of the University of Sydney and the Australian National University. He moved to Cold Spring Harbor Laboratory as a postdoctoral fellow in 1979 and has been there ever since. Dr. Stillman has been director of the Cancer Center at Cold Spring Harbor since 1992. In 1994, he was appointed Director and in 2003 was appointed President of Cold Spring Harbor Laboratory. Dr. Stillman’s research focuses on how chromosomes are duplicated in cells, a process that ensures accurate inheritance of genetic material from one cell generation to the next. Initially focusing on understanding the replication of DNA tumor viruses in mammalian cells, Dr. Stillman studied adenovirus and SV40 virus DNA replication. Dr. Stillman’s lab identified many core cellular DNA replication proteins and reconstituted SV40 DNA replication with purified proteins, thereby characterizing processes such as DNA polymerase clamp loading and switching from one DNA polymerase to another during DNA replication, a model that is used for many other DNA replication associated processes.

    Dr. Stillman also studied DNA-replication-coupled chromatin assembly and identified proteins required for inheritance of nucleosomes. He and his colleagues then characterized the structure of chromosomal origins of DNA replication in the budding yeast S. cerevisiae and discovered the multi-subunit Origin Recognition Complex (ORC), which binds in an ATP-dependent manner to sites in chromosomes where initiation of DNA replication occurs. ORC and other proteins load onto chromosomes competent protein complexes of MCM proteins that are used for initiation of DNA replication once the cell commits to cell division and enters S phase. Recently, Dr. Stillman's lab and others have reconstituted pre-RC assembly in vitro with purified proteins and have characterized how the pre-RC is activated by cell cycle-regulated protein kinases. Surprisingly, in mammalian cells ORC subunits also play roles in both centromere and centrosome activity during mitosis, thereby linking the initiation of DNA replication to processes that ensure accurate chromosome segregation.

2009

  • Victor Ambros, PhD

    • Co-Director, RNA Therapeutics Institute
    • Silverman Professor of Natural Sciences
    • University of Massachusetts Medical School

    Victor Ambros, PhD, grew up in Vermont and graduated from MIT in 1975. He did his graduate research (1976–1979) with David Baltimore at MIT, studying poliovirus genome structure and replication. He began to study the genetic pathways controlling developmental timing in the nematode C. elegans as a postdoc in H. Robert Horvitz's lab at MIT and continued those studies while on the faculty of Harvard (1984–1992), Dartmouth (1992–2007), and the University of Massachusetts Medical School (2008–present). In 1993, he and co-workers Rosalind Lee and Rhonda Feinbaum identified the first microRNA, the product of the heterochronic gene lin-4 in C. elegans. Currently, the chief research interest of the Ambros lab is the roles of microRNA-mediated regulatory pathways in animal development and human disease.

  • Gary Ruvkun, PhD

    • Professor of Genetics
    • Harvard Medical School

    Gary Ruvkun is professor of genetics at Harvard Medical School. His lab uses C. elegans molecular genetics and genomics to study problems in developmental biology and physiology. Dr. Ruvkun is a graduate of UC Berkeley and Harvard. He began to work with C. elegans as a postdoc with Bob Horvitz at MIT and Walter Gilbert at Harvard, where he explored the heterochronic genes that control the temporal dimension of development in a collaborative study with Victor Ambros. The work led to the discovery of the first microRNA gene by the Ambros lab and the understanding that the mechanism of microRNA regulation of target mRNAs is post-transcriptional by the Ruvkun lab. A few years later the Ruvkun lab found the second microRNA gene, let-7 and showed that it is conserved across animal phylogeny.

    Dr. Ruvkun’s lab is now using functional genomic and genetic strategies to systematically discover the components of the RNAi and microRNA pathways in C. elegans. Most of the genes identified in these screens are conserved across eukaryotic phylogeny, suggesting universality of these 21-22 nucleotide pathways. Some of these components may be developed as drug targets to enhance RNAi in mammals, a technical improvement that may be necessary to elevate a laboratory tool to a therapeutic modality. Dr. Ruvkun’s lab has also discovered that C. elegans uses an insulin-signaling pathway to control its metabolism and longevity. The molecular genetic dissection of the insulin pathway has also been important for understanding and treating diabetes, a disease of insulin-signaling deficits. The new genes of the insulin pathway that have emerged from these studies represent new targets for diabetes drug development.

2008

  • F. Ulrich Hartl, MD

    • Director, Max-Planck-Institute of Biochemistry
    • Martinsried, Germany

    F. Ulrich Hartl studied medicine at Heidelberg University. After receiving his MD in 1982 and his doctoral degree in biochemistry in 1985 he moved to the laboratory of Walter Neupert in Munich, where he worked on the mechanism of protein transport into mitochondria, first as a postdoctoral fellow and from 1987 to 1991 as a research group leader. In 1988 he initiated the work on molecular chaperones and demonstrated, collaboratively with A. Horwich, the basic role of chaperones in assisting protein folding. His work in Walter Neupert’s department was interrupted by a stint in William Wickner’s laboratory at UCLA (1989–1990), where he worked on the mechanism of bacterial protein export. After returning to Munich he received his Habilitation in Biochemistry and soon after accepted an offer from Sloan-Kettering Cancer Center in New York to join the newly founded department of James Rothman as an associate member.

    From 1991 to 1997, Dr. Hartl investigated the mechanisms of protein-folding in the bacterial and eukaryotic cytosol. He reconstituted the pathway of chaperone-assisted folding in which the Hsp70 and the GroEL chaperone systems cooperate and discovered that GroEL and its co-factor GroES provide a nano-cage for single protein molecules to fold unimpaired by aggregation. In 1993 Hartl was promoted to member with tenure, and in 1994 he became an Investigator of the Howard Hughes Medical Institute. In 1997 he returned to Munich to head the Department of Cellular Biochemistry at the Max Planck Institute of Biochemistry (MPIB). At MPIB Dr. Hartl continues to investigate the mechanisms of cellular protein-folding using a range of methods from cell biology, biochemistry, and structural biology. In addition, he initiated research into neurodegenerative diseases caused by protein misfolding and aggregation. This work led to the finding that chaperones can effectively inhibit the formation of amyloid aggregates associated with neurodegeneration. Much of this work was done in collaboration with Manajit Hayer-Hartl.

  • Arthur Horwich, MD

    • Sterling Professor of Genetics and Pediatrics
    • Yale University School of Medicine
    • Investigator, Howard Hughes Medical Institute

    Arthur Horwich received undergraduate and medical degrees from Brown University, then trained in pediatric medicine at Yale. As a postdoctoral fellow at the Salk Institute, he studied transforming T antigens with Walter Eckhart and Tony Hunter, then returned to Yale for further postdoctoral training with Leon Rosenberg. The latter studies were directed to understanding the posttranslational import of mitochondrial precursor proteins, examining signal peptides in the precursor proteins. Following appointment to the Yale faculty in Genetics in 1984, Dr. Horwich focused on the mitochondrial "machinery" that recognizes and translocates precursor proteins. In a genetic screen in yeast, uncovered Hsp60 (the yeast homologue of GroEL) as essential for folding newly imported proteins in work in collaboration with Ulrich Hartl. This led to further studies of chaperonin structure and mechanism using the bacterial GroEL-GroES chaperonin system, including X-ray crystallographic studies with Paul Sigler, cryoEM studies with Helen Saibil, and NMR studies with Kurt Wüthrich. Currently he is Sterling Professor of Genetics and Investigator of the Howard Hughes Medical Institute. He has been an attending physician in medical genetics and pediatrics at Yale-New Haven Hospital for the past 20 years. Honors include the 2001 Hans Neurath Award of the Protein Society, the 2004 Gairdner International Award, the 2006 Stein and Moore Award of the Protein Society, the 2007 Wiley Prize in Biomedical Sciences, and the 2008 Rosenstiel Award for Distinguished Work in Basic Medical Science. Dr. Horwich was elected to the National Academy of Sciences in 2003. He is an associate editor of Cell and Molecular Cell and a member of the editorial boards of the Journal of Cell Biology and Structure.

  • Rosalind Franklin, PhD

    • (1920–1958)
    • Honorary Horwitz Prize

2007

  • Joseph G. Gall, PhD

    • Staff Member, Department of Embryology
    • Carnegie Institution
    • American Cancer Society Professor of Developmental Genetics

    Joseph G. Gall received the BS degree in zoology from Yale University in early 1949 then directly entered Yale's graduate program zoology. He completed his PhD in 1952, working with the Drosophila geneticist and developmental biologist Donald F. Poulson. He took a teaching position in the zoology department at the University of Minnesota, where he remained until 1963. In the fall of 1963 he returned to Yale as a visiting professor in what was then the biology department, after the fusion of zoology and botany, and later became professor of biology with a joint appointment in molecular biophysics and biochemistry. Dr. Gall remained at Yale for 20 years, from 1963 to1983; during the last few years he held the Ross G. Harrison Chair in Biology. In 1983 he joined the embryology department of the Carnegie Institution in Baltimore as a staff member. In 1984 he was appointed American Cancer Society Professor of Developmental Genetics, a lifetime appointment.

    Dr. Gall has been an active member of the American Society for Cell Biology (ASCB) since its inception in 1960, serving as president 1967 – 68 and as a member of its council and several committees at various times.

    Dr. Gall's long-term research interests have been in the structure and function of the cell, particularly the nucleus. His earliest studies involved the giant “lampbrush” chromosomes found in oocytes of frogs and salamanders. These are the largest known chromosomes and permit various observations and manipulations that are difficult or impossible with smaller chromosomes. Among his more important findings, made at a time when the site of cellular RNA synthesis was still unclear (late 1950s, early 1960s), was that cellular RNA synthesis occurs on loops of DNA that extend out from the axis of the chromosome. Studies on the kinetics of DNase digestion showed that the chromosome consists of a single extremely long DNA molecule. Electron microscopic studies he carried out at about the same time on the nuclear envelope established the existence of the nuclear pore complex and its eight-fold symmetry. Other studies on centrioles clarified aspects of their replication during the cell cycle.

    After moving to Yale, Dr. Gall began studies on ribosomal RNA (rRNA) and the genes that code for it (rDNA). In 1967–68, through a combination of biochemical and cytological observations, he demonstrated that these genes are able to leave the chromosome and replicate independently during the early stages of oocyte formation in amphibians and other animals. This phenomenon of gene amplification was independently discovered by Igor Dawid and Donald Brown of the Carnegie Institution. At about the same time, a former postdoctoral student of Dr. Gall’s, Oscar Miller, demonstrated the activity of the amplified genes in a set of electron microscopic observations.

    The studies on gene amplification were followed almost immediately by development of the technique of in situ hybridization, in collaboration with Dr. Gall’s graduate students Mary Lou Pardue and Susan Gerbi. This technique allowed the identification of specific DNA or RNA sequences at the cellular or subcellular level. Their original technique used radioactive probes. The procedure was later modified by others to use fluorescent probes, which permit even finer localization and simultaneous use of multiple probes. In situ hybridization is now one of the most widely used cytological techniques. It permits localization of genes to specific chromosome regions and of RNA sequences to specific cells or groups of cells.

    Among several important observations Dr. Gall’s team made with the in situ hybridization technique was that the heterochromatic regions of chromosomes consist of simple sequences called “satellite” DNA. They also showed how in situ hybridization could be used with the giant chromosomes of Diptera for precise gene localization. A few years later, gene cloning made numerous sequences available for mapping studies.

    Dr. Gall’s interest in rDNA amplification during oocyte formation led him to investigate the similar phenomenon he discovered in the ciliated protozoan Tetrahymena. Work on this organism led to the demonstration that the rDNA genes exist as free molecules in the macronucleus. A postdoctoral fellow, Elizabeth Blackburn, found that the ends of these molecules had a unique structure consisting of a hexanucleotide repeat GGGGTT. Later studies by Blackburn and others established that this repeat, or very similar ones, are found at the ends or telomeres of chromosomes from nearly every type of animal and plant investigated. The in situ hybridization technique was valuable in making this determination.

    In recent years, the focus of Dr. Gall’s research has been the organization of transcription in the nucleus. He has studied the small nuclear RNAs (snRNAs), which are known to play important roles in the processing of all types of messenger RNA (mRNA) and ribosomal RNA (rRNA). His lab is concentrating on several nuclear organelles that contain snRNAs, including the nucleolus, Cajal bodies, and nuclear speckles. Their most recent studies suggest that Cajal bodies may be sites for preassembly and/or modification of macromoleuclar complexes that carry out nuclear transcription and RNA processing.

    The research findings from Dr. Gall’s laboratory have been reported in 150 articles in various scientific journals. His research has been combined with his long-standing interest in the history of biology, particularly cell biology and microscopy. He has collected early books in these areas and in 1996 published a book, “Views of the Cell: A Pictorial History.” The book brings together 60 historical images and their descriptions that Dr. Gall originally prepared as covers for Molecular Biology of the Cell, the official journal of the American Society for Cell Biology. In 2001, Dr. Gall co-edited with J. Richard McIntosh a book of readings in cell biology titled “Landmark Papers in Cell Biology,” published jointly by the American Society for Cell Biology and Cold Spring Harbor Laboratory Press.

  • Elizabeth H. Blackburn, PhD

    • Morris Herzstein Professor in Biology and Physiology
    • Department of Biochemistry and Biophysics,
    • University of California, San Francisco

    Elizabeth H. Blackburn is a leader in the area of telomere and telomerase research, with broad experience in the different aspects of telomere function and biology. She discovered the ribonucleoprotein enzyme, telomerase. Her laboratory is a leader in manipulating telomerase activity in cells, and she has amassed considerable knowledge and experience in the effects this has on cells.

    Dr. Blackburn and her research team at the University of California, San Francisco, are working with various cells, including human cancer cells, with the goal of understanding telomerase and telomere biology. Her work on telomeres and telomerase has been published extensively in peer-reviewed journals.

    Dr. Blackburn earned her BSc (1970) and MSc (1972) degrees from the University of Melbourne in Australia, and her PhD (1975) from the University of Cambridge in England. She did her postdoctoral work in molecular and cellular biology at Yale from 1975 to 1977.

    In 1978, Dr. Blackburn joined the faculty of the Department of Molecular Biology at the University of California at Berkeley. In 1990, she joined the Department of Microbiology and Immunology at UC San Francisco, where she served as department chair from 1993 to 1999. Dr. Blackburn is currently a faculty member in the Department of Biochemistry and Biophysics at UCSF. She is also a non-resident fellow of the Salk Institute.

    Throughout her career, Dr. Blackburn has been the recipient of many prestigious awards. These include the Eli Lilly Research Award for Microbiology and Immunology (1988), the National Academy of Science Award in Molecular Biology (1990), and an Honorary Doctorate of Science from Yale University (1991). She was a Harvey Society Lecturer at the Harvey Society in New York (1990), and the recipient of the UCSF Women's Faculty Association Award (1995). Most recently, she was awarded the Australia Prize (1998), Harvey Prize (1999), Keio Prize (1999), American Association for Cancer Research-G.H.A. Clowes Memorial Award (2000), American Cancer Society Medal of Honor (2000), AACR-Pezcoller Foundation International Award for Cancer Research (2001), General Motors Cancer Research Foundation Alfred P. Sloan Award (2001), E.B. Wilson Award of the American Society for Cell Biology (2001), 26th Annual Bristol-Myers Squibb Award for Distinguished Achievement in Cancer Research (2003), Dr. A.H. Heineken Prize for Medicine (2004), Kirk A. Landon-AACR prize for Basic Cancer Research (2005), and Albert Lasker Medical Research Award in Basic Medical Research (2006).

    Dr. Blackburn was named California Scientist of the Year in 1999, elected president of the American Society for Cell Biology for the year 1998, and served as a board member of the Genetics Society of America (2000–2002). She is an elected Fellow of the American Academy of Arts and Sciences (1991), the Royal Society of London (1992), the American Academy of Microbiology (1993), and the American Association for the Advancement of Science (2000). She was elected Foreign Associate of the National Academy of Sciences in 1993 and a member of the Institute of Medicine in 2000.

  • Carol W. Greider, PhD

    • Daniel Nathans Professor and Director
    • Department of Molecular Biology and Genetics
    • Professor of Oncology
    • The Johns Hopkins University School of Medicine

    Carol W. Greider received a BA from the University of California at Santa Barbara in 1983 and a PhD from the University of California at Berkeley in 1987. In 1984, working with Elizabeth Blackburn, she discovered telomerase, an enzyme that maintains telomeres, or chromosome ends. Dr. Greider first isolated and characterized telomerase from the ciliate Tetrahymena. In 1988 Dr. Greider went to Cold Spring Harbor Laboratory where, as an independent Cold Spring Harbor Fellow, she cloned and characterized the RNA component of telomerase. In 1990, Dr. Greider was appointed an assistant investigator at Cold Spring Harbor Laboratory. She expanded the focus of her telomere research to include the role of telomere length in cell senescence, cell death, and cancer. With Calvin Harley, she showed that human telomeres shorten progressively in primary human cells. This work, along with that of other researchers, led to the idea that telomere maintenance and telomerase may play important roles in cellular senescence and cancer. Dr. Greider was appointed associate investigator at Cold Spring Harbor Laboratory in 1992 and investigator in 1994. Her lab continued to work on both the biochemistry of telomerase and the role of telomere maintenance in cancer in human and mouse cells.

    In 1997, Dr. Greider moved her laboratory to the Department of Molecular Biology and Genetics at the Johns Hopkins University School of Medicine. In 1999, she was appointed professor of molecular biology and genetics, and in 2001 she was appointed professor of oncology. At Hopkins Dr. Greider’s group continued to study the biochemistry of telomerase and determined the secondary structure of the human telomerase RNA. She also expanded her work on a mouse model of telomere dysfunction and showed that the shortest telomere in a cell triggers a DNA damage response. In 2004 she was appointed the Daniel Nathans Professor and Director of the Department of Molecular Biology and Genetics. Dr. Greider has won nemerous awards for her work on telomerase: the Gardiner Award (1998), the Rosenstiel Award (1999), the Passano Foundation Award (1999), and the Richard Lounsbery Award (2003). In 2003, Dr. Greider was elected to the National Academy of Sciences and to the American Academy of Arts and Sciences. In 2006 she received the Wiley Prize and the Albert Lasker Award for Basic Medical Research. Dr. Greider currently directs a group of ten researchers who are focused on understanding telomeres and telomerase and their role in chromosome stability, stem cell failure, and cancer.

2006

  • Roger D. Kornberg, PhD

    • Professor of Medicine
    • Department of Structural Biology
    • Stanford School of Medicine

    Roger Kornberg, professor of medicine in the department of structural biology at Stanford School of Medicine, received a PhD in chemistry from Stanford in 1972 for his demonstration of the diffusional motions of lipids in bilayer membranes, termed flip-flop and lateral diffusion. He was a postdoctoral fellow and member of the scientific staff at the Laboratory of Molecular biology in Cambridge, England, from 1972–75, where he discovered the nucleosome. He moved to his present position in 1978, where his research has focused on the mechanism and regulation of eukaryotic gene transcription. Notable findings include the demonstration of the role of nucleosomes in transcriptional regulation, the establishment of a yeast RNA polymerase II transcription system and the isolation of all the proteins involved, the discovery of the mediator of transcriptional regulation, the development of two-dimensional protein crystallization and its application to transcription proteins, and the atomic structure determination of an RNA polymerase II transcribing complex. Dr.Kornberg’s closest collaborator has been his wife, Dr. Yahli Lorch. They have three children, Guy, Maya, and Gil.

2005

  • Ada Yonath, PhD

    • Helen and Milton A. Kimmelman Center for Biomolecular Structure and Assembly
    • Weizmann Institute of Science

    Ada Yonath director of the Helen and Milton A. Kimmelman Center for Biomolecular Structure and Assembly of the Weizmann Institute of Science, is a professor in the Department of Structural Biology.

    Dr. Yonath uses ribosomal crystallograph to study the mechanisms underlying protein biosynthesis, a research line she pioneered more than twenty years ago despite considerable skepticism of the international scientific community. She determined the complete high-resolution structures of both ribosomal subunits and discovered, within the otherwise asymmetric ribosome, the universal symmetrical region that provides the framework and navigates the process of polypeptide polymerization. Consequently, she showed that the ribosome is a ribozyme that places its substrates in stereochemistry suitable for peptide bond formation and for substrate-mediated catalysis. Two decades ago, she visualized the path taken by the nascent proteins, namely the ribosomal tunnel, and recently revealed the dynamics elements enabling its involvement in elongation arrest, gating, intracellular regulation, and nascent chain trafficking into their folding space.

    Additionally, Dr. Yonath elucidated the modes of action of more than twenty different antibiotics targeting the ribosome, illuminated mechanisms of drug resistance and synergism, and deciphered the structural basis for antibiotic selectivity and showed how it plays a key role in clinical usefulness and therapeutic effectiveness, thus paving the way for structure-based drug design.

    To enable ribosomal crystallography, Dr. Yonath introduced a novel technique, cryo bio-crystallography, which in an amazingly short time, became routine and revolutionized structural biology, allowing intricate projects otherwise considered formidable.

    Dr. Yonath earned her PhD at the Weizmann Institute of Science and did postdoctoral studies at MIT and Carnegie Mellon University. In 1970, she established what was for nearly a decade the only protein crystallography laboratory in Israel. After returning from a sabbatical year at the University of Chicago, from 1986 to 2004 she headed a Max-Planck Research Unit in Hamburg, Germany, in parallel to her research activities at the Weizmann Institute.

    Dr. Yonath is a member of the National Academy of Sciences, USA; the American Academy of Arts and Sciences; the Israel Academy of Sciences and Humanities; the European Academy of Sciences and Art; and the European Molecular Biology Organization. Her awards and honors include the Israel Prize, the first European Crystallography Prize, an NIH Certificate of Distinction, the Harvey Prize, the Kilby Prize, the Cotton Medal of the US Chemical Society, the Anfinsen Award of the International Protein Society, the Zurich University's Paul Karrer Gold Medal, the University of Southern California's Massry Award and Medal, the Datta Medal of the Federation of European Biochemical Societies, and the Fritz Lipmann Award of the German Biochemical Society. At the Weizmann Institute, Dr. Yonath is the Martin S. and Helen Kimmel Professorial Chair.

2004

  • Tony Hunter, PhD

    • Salk Institute for Biological Studies

    Tony Hunter received his PhD in biochemistry from the University of Cambridge, England, for his research on mammalian protein synthesis. He is an American Cancer Society research professor and director of the Molecular and Cell Biology Laboratory at the Salk Institute for Biological Studies. He is also an adjunct professor in the Division of Biological Sciences at the University of California at San Diego. His research focuses on how cells regulate their growth and division and on how mutations in genes that regulate growth lead to cancer.

    In 1979, Dr. Hunter's lab discovered that a phosphate can be attached to tyrosine residues in proteins, a discovery that enabled researchers to study tyrosine kinases and their functions in signal transduction, cell growth and development, and cancer and other diseases. Dr. Hunter's and Dr. Pawson's work has led to the development of drugs for halting cancer cell proliferation and has potential for other significant therapies.

    Dr. Hunter's current research interests include the tyrosine kinases of the Src and growth factor receptor families, as well as the signaling pathways downstream of these tyrosine kinases that regulate cell growth, cell migration, and differentiation. His group also studies the cyclin-dependent protein kinases and other protein kinases that regulate progression through the cell cycle, how protein ubiquitination and degradation is used as a means of regulating signaling pathways and the cell cycle, and protein trafficking.

    He is on the editorial boards of several journals, including Cell, Molecular Cell, the EMBO Journal and the Proceedings of the National Academy of Sciences and has received many awards for his research, including a National Cancer Institute Outstanding Investigator Award.

    He is a Fellow of the Royal Society of London, an associate member of the European Molecular Biology Organization, a fellow of the American Academy of Arts and Sciences, a foreign associate of the National Academy of Sciences, and a member of the Institute of Medicine.

  • Anthony Pawson

    • University of Toronto

    Anthony Pawson was an undergraduate at the University of Cambridge, England (1970–1973) and obtained his PhD at the Imperial Cancer Research Fund in London with Dr. Alan Smith, working on retroviral gene expression. He undertook postdoctoral work at the University of California at Berkeley with G. Steven Martin (1976–1980), where he identified a variety of retroviral oncogene products, and provided early evidence for the role of tyrosine phosphorylation in malignant transformation. He moved to the University of British Columbia, Vancouver as an Assistant Professor in 1981, and took up his present position at the Samuel Lunenfeld Research Institute of Mt. Sinai Hospital, Toronto, in 1985. Over the last 20 years has explored the mechanisms through which cell surface receptors control intracellular signaling pathways, and the organization of cell regulatory systems, building on his identification of the SH2 domain as the prototypic interaction module.

    Tony Pawson is a University Professor of the University of Toronto, Director of Research at the Samuel Lunenfeld Research Institute of Mt. Sinai Hospital, and a Distinguished Scientist of the Canadian Institutes for Health Research. He has received a number of awards, including the Gairdner Foundation International Award, the AACR/Pezcoller International Award for Cancer Research, the Heineken Prize for Biochemistry and Biophysics (Royal Netherlands Academy of Arts and Sciences,) and the Killam Prize for Health Sciences. He is a fellow of the Royal Societies of London and Canada, a foreign associate of the National Academy of Sciences (US), an associate member of EMBO, and a recipient of the Order of Canada.

2003

  • Roderick MacKinnon

    • Rockefeller University

2002

  • James E. Rothman

    • Sloan-Kettering Institute
  • Randy W. Schekman

    • University of California, Berkeley

2001

  • Avram Hershko

    • Technion-Israel Institute of Technology, Israel
  • Alexander Varshavsky

    • California Institute of Technology, CA