Chemistry

advertisement
Chemistry
A chiral catalyst for enantioselective carbon-hydrogen activation. Work
done in the laboratory of Jin-Quan Yu, Ph.D., associate professor.
Jin-Quan Yu, Ph.D., Associate Professor
CHEMISTRY
DEPAR TMENT OF
CHEMISTRY
S TA F F
K.C. Nicolaou, Ph.D.*
Chairman
Aline W. and L.S. Skaggs
Professor of Chemical
Biology
Darlene Shiley Chair in
Chemistry
Dariush Ajami, Ph.D.
Assistant Professor of
Molecular Assembly
Phil S. Baran, Ph.D.
Professor
Dale L. Boger, Ph.D.*
Richard and Alice Cramer
Professor of Chemistry
Tobin J. Dickerson, Ph.D.
Assistant Professor
Albert Eschenmoser, Ph.D.*
Professor
Sheng Ding, Ph.D.
Associate Professor
M.G. Finn, Ph.D.*
Professor
Valery Fokin, Ph.D.
Associate Professor
M. Reza Ghadiri, Ph.D.*
Professor
William A. Greenberg, Ph.D.
Assistant Professor of
Chemistry
Inkyu Hwang, Ph.D.
Assistant Professor
Kim D. Janda, Ph.D.**
Professor
Ely R. Callaway, Jr., Chair
in Chemistry
Director, Worm Institute of
Research and Medicine
Gunnar Kaufmann, Ph.D.
Assistant Professor of
Chemistry
2008
Jeffery W. Kelly, Ph.D.*
Lita Annenberg Hazen
Professor of Chemistry
Ramanarayanan
Krishnamurthy, Ph.D.
Associate Professor
Lucas J. Leman, Ph.D.
Assistant Professor of
Chemistry
Richard A. Lerner, M.D.***
President, The Scripps
Research Institute
Lita Annenberg Hazen
Professor of
Immunochemistry
Cecil H. and Ida M. Green
Chair in Chemistry
Roy Periana, Ph.D.*****
Professor
Evan T. Powers, Ph.D.
Associate Professor of
Chemistry
Julius Rebek, Jr., Ph.D.*
Professor
Director, The Skaggs Institute
for Chemical Biology
Edward Roberts, Ph.D.
Professor
THE SCRIPPS RESEARCH INSTITUTE
(Andrew) Bin Zhou, Ph.D.
Assistant Professor of
Immunochemistry
79
Deboshri Banerjee, Ph.D.
Elizabeth Barrett, Ph.D.****
Roland Barth, Ph.D.*****
SENIOR SCIENTIST
Clay Bennett, Ph.D.
Luis Martinez, Ph.D.*****
Moritz Biskup, Ph.D. †
Universität Karlsruhe
Karlsruhe, Germany
S TA F F S C I E N T I S T S
Lisa Eubanks, Ph.D.
Rajesh Grover, Ph.D.
Sarah Hanson, Ph.D.
Lubica Supekova, Ph.D.
Wen Xiong, Ph.D.
Anthony Boitano, Ph.D. †
Genomics Institute of the
Novartis Foundation
San Diego, California
Laure Bouchez, Ph.D.
Kristopher Boyle, Ph.D.
Christopher Burke, Ph.D
SERVICE FACILITIES
Antonio Burtoloso, Ph.D. †
University of Sao Paulo
Sao Paulo, Brazil
Raj K. Chadha, Ph.D.
Director, X-ray
Crystallography Facility
Mark Bushey, Ph.D. †
Exxon, Inc.
Union City, New Jersey
Dee H. Huang, Ph.D.
Director, Nuclear Magnetic
Resonance Facility
Darren Bykowski, Ph.D.*****
Gary E. Siuzdak, Ph.D.
Senior Director, Mass
Spectrometry Facility
Katerina Capkova, Ph.D.
I N S T R U M E N TAT I O N /
Floyd E. Romesberg, Ph.D.
Associate Professor
Petr Capek, Ph.D.
Arani Chanda, Ph.D.
Ke Chen, Ph.D.
Peng Chen, Ph.D.
William Roush, Ph.D.*****
Professor
SENIOR RESEARCH
Peter G. Schultz, Ph.D.*
Professor
Scripps Family Chair
Suresh Pitram, Ph.D.
Govardhan Cherukupalli,
Ph.D. †
Epix Pharmaceuticals
Lexington, Massachusetts
R E S E A R C H A S S O C I AT E S
Jodie Chin, Ph.D.
Ramzey Abujarour, Ph.D.
Srinivas Reddy Chirapu, Ph.D.
Rajesh Ambasudhan, Ph.D.
Chandramouli Chiruta, Ph.D.
Manuel Amorin Lopez, Ph.D.
Dong-Gyu Cho, Ph.D.
Mark Ams, Ph.D.
So-Hye Cho, Ph.D.
Yoshio Ando, Ph.D.
Sungwook Choi, Ph.D.
Deepshikha Angrish, Ph.D.
Joyanta Choudhury, Ph.D.
Shinji Ashida, Ph.D.
Sarwat Chowdhury, Ph.D.
Micahel Baksh, Ph.D.
Stepan Chuprakov, Ph.D.
K. Barry Sharpless, Ph.D.*
W.M. Keck Professor of
Chemistry
Anita D. Wentworth, Ph.D.
Assistant Professor
Paul Wentworth, Jr., Ph.D.
Professor
Chi-Huey Wong, Ph.D.*
Professor of Chemistry
Jin-Quan Yu, Ph.D.
Associate Professor
A S S O C I AT E
80 CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
Petr Cigler, Ph.D.
David Edmonds, Ph.D.
Neil Grimster, Ph.D.
Giltae Hwang, Ph.D.
T. Ryan Cirz †
Achaogen
South San Francisco,
California
Jem Efe, Ph.D.
Rajesh K. Grover, Ph.D.
Jan Elsner, Ph.D. †
Celgene Pharmaceuticals
San Diego, California
Jan Grunewald, Ph.D.
Michael Jahnz, Ph.D. †
NOXXON Pharma AG
Berlin, Germany
Daniel Ess, Ph.D.
Richard Guy, Ph.D.****
Ph.D. †
Scott Cockroft,
University of Edinburgh
Edinburgh, Scotland
Ph.D. †
David Colby,
Purdue University
West Lafayette, Indiana
Kevin Cole, Ph.D. †
Eli Lilly and Company
Indianapolis, Indiana
Christine Crane, Ph.D.
Matthew Cremeens, Ph.D. †
Gonzaga University
Redmond, Washington
Fernando Rodrigo Pinacho
Crisostomo, Ph.D. †
Burnham Institute for
Medical Research
La Jolla, California
Cyrine Ezzili, Ph.D.
Xingang Fang, Ph.D.
Simon Ficht, Ph.D. †
Sanofi-Aventis Deutschland
GmbH
Frankfurt, Germany
Joseph Rodolph Fotsing,
Ph.D. †
Senomyx, Inc.
San Diego, California
Bozena Frackowiak, Ph.D. †
Politechnika Opolska
Opole, Poland
Etzer Darout,
Pfizer Inc.
Groton, Connecticut
Ph.D. †
Amy DeBaillie,
Eli Lilly and Company
Indianapolis, Indiana
Graeme Freestone,
Metabasis Therapeutics, Inc.
San Diego, California
Amelia Fuller, Ph.D. †
Santa Clara University
Santa Clara, California
Jianmin Gao, Ph.D. †
Boston College
Chestnut Hill, Massachusetts
Haibo Ge, Ph.D.
Judith Denery, Ph.D.
Ross Denton, Ph.D. †
University of Cambridge
Cambridge, England
Savvas Georgiades, Ph.D.****
Ola Ghoneim, Ph.D. †
Qatar University
Doha, Qatar
Caroline Desponts, Ph.D.
Antonia Di Mola, Ph.D.
Deguo Du, Ph.D.
Anna Dubrovska, Ph.D.
Viktoriya Dubrovskaya, Ph.D.
Joshua Dunetz, Ph.D. †
Pfizer Inc.
Groton, Connecticut
Kyle Eastman, Ph.D.
Yuanjun He, Ph.D.
Jason Hein, Ph.D.
Dube Henry, Ph.D.
Nathan Gianneschi, Ph.D. †
University of California
San Diego, California
Cristina Gil-Lamaignere,
Ph.D. †
University Hospital Nuestra
Señora de la Candelaria
Santa Cruz de Tenerife, Spain
Rodolfo Gonzalez, Ph.D.
Scott Grecian, Ph.D.
Rong Jiang, Ph.D.*****
Guo Jiantoa, Ph.D.
Hiroyuki Kakei, Ph.D. †
Takeda Pharmaceutical
Company Limited
Osaka, Japan
Jaroslaw Kalisiak, Ph.D.
Seiji Kamioka, Ph.D.
Moumita Kar, Ph.D.
Marcos Hernandez, Ph.D.
Kwang Mi Kim, Ph.D.
Par Holmberg, Ph.D. †
Memorial Sloan Kettering
Cancer Center
New York, New York
Wen-Xu Hong, Ph.D.
Yu Fu, Ph.D.
Ph.D. †
Masaki Handa, Ph.D. †
Sagami Chemical Research
Center
Ayase, Kanagawa, Japan
Ph.D. †
Jeffrey Culhane, Ph.D.
Stephen Dalby, Ph.D.
Tanja Gulder, Ph.D.
Zhangyong Hong, Ph.D.
Richard J. Hooley, Ph.D. †
University of California
Riverside, California
Tamara Hopkins, Ph.D. †
Boehringer Ingelheim
Pharmaceuticals, Inc.
Ridgefield, Connecticut
Allen Horhota, Ph.D.
Tony Horneff, Ph.D.
F. Scott Kimball, Ph.D.
Jeremy Kister, Ph.D.
Keith Korthals, Ph.D.
Larisa Krasnova, Ph.D.
Arkady Krasovskiy, Ph.D.
Luke Lairson, Ph.D.
Jae Wook Lee, Ph.D.
Jinq-Chyi Lee, Ph.D. †
National Health Research
Institutes
Miaoli County, Taiwan
Jong Seok Lee, Ph.D.
Claas Hovelmann, Ph.D.
Ki-Bum Lee, Ph.D. †
Rutgers University
Piscataway, New Jersey
Fang Hu, Ph.D. †
Department of Molecular
Biology, Scripps Research
Sejin Lee, Ph.D. †
SK Drug Development Center
Daejong, Korea
Xiaoyi Hu, Ph.D.
Alexandre Lemire, Ph.D.****
Zheng-Zheng Huang, Ph.D. †
DuPont Central Research
and Development
Wilmington, Delaware
Edward Lemke, Ph.D.
Ben Hutchins, Ph.D.
Chuang-Chuang Li, Ph.D. †
Peking University
Peking, China
Jun-Li Hou, Ph.D.
Der-ren Hwang, Ph.D. †
Academia Sinica
Taipei, Taiwan
Christophe Letondor,
Ph.D.****
Fangzheng Li, Ph.D.*****
CHEMISTRY
2008
Hongming Li, Ph.D. †
Schering-Plough
Kenilworth, New Jersey
Joonwoo Nam, Ph.D. †
CytRx Corporation
San Diego, California
Ke Li, Ph.D. †
DuPont Central Research
and Development
Wilmington, Delaware
Tae-Gyu Nam, Ph.D.
THE SCRIPPS RESEARCH INSTITUTE
Troy Ryba, Ph.D. †
Broad Institute of MIT and
Harvard
Cambridge, Massachusetts
Ph.D. †
Sebastian Steiniger, Ph.D.
Antonia Stepan, Ph.D.
James Stover, Ph.D.
Youngha Ryu,
Texas Christian University
Fort Worth, Texas
Bernhard Stump, Ph.D.
Catherine Saccavini, Ph.D.
Hui Kai Sun, Ph.D.*****
Severin Odermatt, Ph.D.****
Nicholas Salzameda, Ph.D.
Shinobu Takizawa, Ph.D.
Yeon-Hee Lim, Ph.D. †
Schering-Plough
Kenilworth, New Jersey
Christian Olsen, Ph.D.
Antonio Sanchez-Ruiz, Ph.D.
Yazmin Osornio, Ph.D.****
Yoshikazu Sasaki, Ph.D.
Tongxiang Lin, Ph.D.
Junguk Park, Ph.D.
Stefan Schiller, Ph.D.
Adam Talbot, Ph.D. †
Institute of Chemical and
Engineering Sciences
Jurong Island, Singapore
Troy Lister, Ph.D. †
Novartis
Cambridge, Massachusetts
Nitin Patil, Ph.D.
Niklas Schone, Ph.D.
Annie Tam, Ph.D.
Yefeng Tang, Ph.D.
Christopher Liu, Ph.D. †
Cubix Pharmaceuticals
Lexington, Massachusetts
Richard Payne, Ph.D. †
University of Sydney
Sydney, Australia
Michael Schramm, Ph.D. †
California State University
Long Beach, California
Wenshe Liu, Ph.D. †
Texas A&M University
College Station, Texas
Xuemei Peng, Ph.D.****
Edward Sessions, Jr., Ph.D.
Murali Peram Surakattula,
Ph.D. †
CytRx Corporation
San Diego, California
Shigeki Seto, Ph.D.
Pi-Hui Liang, Ph.D. †
Academia Sinica
Taipei, Taiwan
Michael Luzung, Ph.D.
Utpal Majumder, Ph.D.
Sreeman Mamidyala, Ph.D.
Andrew Nguyen, M.D., Ph.D.
Romain Noel, Ph.D.
George Nora, Ph.D.*****
Johan Paulsson, Ph.D.
Alexander Mayorov, Ph.D.
Charles Melancon, Ph.D.
Lionel Moisan, Ph.D. †
CEA
Gif-Sur-Yvette, France
Ana Montero, Ph.D.****
Miguel Morales, Ph.D.
Shun Su, Ph.D.
Mariola Tortosa, Ph.D. †
Instituto de Quimica Organica,
CSIC
Madrid, Spain
Craig Turner, Ph.D.****
Roshan Perera, Ph.D. †
University of Texas
Austin, Texas
Takeshi Masuda, Ph.D.
Michael Maue, Ph.D. †
Bayer CropScience AG
Monheim, Germany
Young Jun Seo, Ph.D.
81
Mary Jo Sever, Ph.D.
Alex Shaginian, Ph.D. †
Ardea Biosciences
San Diego, California
David Shaw, Ph.D.
Ramulu Poddutoori, Ph.D.
Weijun Shen, Ph.D.
Jonathan Pokorski, Ph.D.
Xiao Shengxiong, Ph.D.
Agustí Lledó Ponsati, Ph.D.
Bingfeng Shi, Ph.D.
Daniela Radu, Ph.D. †
DuPont Central Research
and Development
Wilmington, Delaware
Yan Shi, Ph.D.
Vincent Trepanier, Ph.D. †
Institute of Chemical and
Engineering Sciences
Jurong Island, Singapore
Jonathan Tripp, Ph.D.
Meng-Lin Tsao, Ph.D. †
University of California
Merced, California
Andrew Udit, Ph.D.
Hiroki Shigehisa, Ph.D.
Hiroyuki Shimamura, Ph.D.
Ronald Rahaim, Ph.D.*****
Siddhartha Shenoy, Ph.D.
Praveen Rao, Ph.D. †
University of Waterloo
Waterloo, Ontario, Canada
Matthew Tremblay, Ph.D.
Ryan Simkovsky, Ph.D.
Chinnappan Sivasankar,
Ph.D.****
Taiki Umezawa, Ph.D. †
Hokkaido University
Sapporo, Japan
Kenji Usui, Ph.D. †
Tokyo Institute of Technology
Tokyo, Japan
Carlos Valdez, Ph.D. †
Rigel Pharmaceuticals, Inc.
South San Francisco,
California
Adam Morgan, Ph.D. †
Concert Pharmaceuticals, Inc.
Lexington, Massachusetts
Per Restorp, Ph.D.
Ting-Wei Mu, Ph.D.
Jin-Kyu Rhee, Ph.D.
Michael Smolinski, Ph.D. †
Kinex Pharmaceuticals
Buffalo, New York
S. Vasudeva Naidu, Ph.D.
Fatima Rivas, Ph.D.
Xinyi Song, Ph.D.
Feng Wang, Ph.D.
Yuya Nakai, Ph.D.
Joshua Roth, Ph.D.*****
Simon Stamm, Ph.D.
Jian Wang, Ph.D.
Kimberly Reynolds, Ph.D.
Punna Venkateshwarlu, Ph.D.
82 CHEMISTRY
Jiangyun Wang, Ph.D. †
Institute of Biophysics
Beijing, China
Lin Wang, Ph.D.
Sheng-Kai Wang, Ph.D.
Weidong Wang, Ph.D.
Xisheng Wang, Ph.D.
Yajuan Wang, Ph.D.
Yuanhua Wang, Ph.D.
Timo Weide, Ph.D.
Albert Willis, Ph.D. †
Pharmagra Labs, Inc.
Brevard, North Carolina
Tao Wu, Ph.D. †
Institute of Chemical and
Engineering Sciences
Jurong Island, Singapore
Heiko Wurdak, Ph.D.
Jian Xie, Ph.D.
Wen Xiong, Ph.D.
Yue Xu, Ph.D.
Junichiro Yamaguchi, Ph.D.
Ryu Yamasaki, Ph.D. †
Tokyo University of Science
Tokyo, Japan
Ura Yasuyuki, Ph.D. †
Nara Women’s University
Nara, Japan
Yan Yin, Ph.D.
2008
Heyue Zhou, Ph.D.
Hongyan Zhou, Ph.D.
Shoutian Zhu, Ph.D.
Joerg Zimmermann, Ph.D.
V I S I T I N G I N V E S T I G AT O R S
Keisuke Fukuchi, Ph.D.
Sankyo Co., Ltd.
Tokyo, Japan
Christine Hernandez, Ph.D. †
University of Philippines
Diliman, Philippines
(Edmond) Shie-Liang Hsieh,
Ph.D.
National Yang-Ming University
Taipei, Taiwan
Masakazu Imamura, Ph.D. †
Astellas Pharma Inc.
Tsukuba, Ibaraki, Japan
Kuniyuki Kishikawa, Ph.D. †
Kyowa Hakko Kogyo Co., Ltd.
Sunto-gun, Shizuoka, Japan
Michael Meijler, Ph.D. †
Ben-Gurion University of the
Negev
Be’er Sheva, Israel
Takayoshi Suzuki, Ph.D. †
Nagoya City University
Nagoya, Japan
Yoshiyuki Yoneda, Ph.D. †
Daiichi Pharmaceutical Co.,
Ltd.
Tokyo, Japan
Ian Young, Ph.D.
S C I E N T I F I C A S S O C I AT E
Zhanqian Yu, Ph.D.
Xu Yuan, Ph.D.
Weiqiang Zhan, Ph.D.
Hongjun Zhang, Ph.D.
Xuejun Zhang, Ph.D.
Yanghui Zhang, Ph.D.
Yingchao Zhang, Ph.D. †
Hoffmann-La Roche, Inc.
Nutley, New Jersey
Jon Ashley
THE SCRIPPS RESEARCH INSTITUTE
* Joint appointment in The
Skaggs Institute for Chemical
Biology
** Joint appointments in The
Skaggs Institute for Chemical
Biology and the Department of
Immunology and Microbial
Science
*** Joint appointments in The
Skaggs Institute for Chemical
Biology and the Department of
Molecular Biology
**** Appointment completed
***** Scripps Florida
†
Appointment completed; new
location shown
CHEMISTRY
2008
Chairman’s Overview
s the “central science,” chemistry stands between
biology and medicine and between physics and
materials science and provides the crucial bridge
for drug discovery and
development. But chemistry has a much more
profound and useful role
in science and society. It
is the discipline that continually creates the myriad
of new materials that we
all encounter in our everyday lives: pharmaceuticals, high-tech materials,
polymers and plastics,
K.C. Nicolaou, Ph.D.
insecticides and pesticides,
fabrics and cosmetics, fertilizers, and vitamins—basically
everything we can touch, feel, and smell.
Chemistry at Scripps Research focuses on chemical
synthesis and chemical biology, the areas most relevant
to biomedical research and materials science. The members of our faculty are distinguished teacher-scholars who
maintain highly visible and independent research programs
in areas as diverse as biological and chemical catalysis,
synthesis of natural products, combinatorial chemistry,
molecular design, supramolecular chemistry, chemical
evolution, materials science, and chemical biology. The
chemistry graduate program attracts some of the bestqualified candidates from the United States and abroad.
Our major research facilities, under the direction of Dee
H. Huang (nuclear magnetic resonance), Gary Siuzdak
(mass spectrometry), and Raj Chadha (x-ray crystallography), are second to none and continue to provide crucial support to our research programs. In addition, the
Mabel and Arnold Beckman Center for the Chemical
Sciences constantly receives high praise from visitors
from around the world for its architectural design and
operational aspects, both highly conducive to research.
Research in the Department of Chemistry goes on
unabated, establishing international visibility and attracting attention, as evidenced by numerous lecture invitations, visits by outside scholars, and headline news in
the media. As of 2007, the Institute for Scientific Information ranked 2 members of the department as highly
cited researchers (in the top 100 worldwide).
Richard Lerner and his group continue their research
on antibodies, chemical synthesis, and the biological
A
THE SCRIPPS RESEARCH INSTITUTE
83
role of polyoxygen species. Scientists in Albert Eschenmoser’s group continue to work on the chemical etiology of nucleic acids and the origins of life.
Barry Sharpless and his group persist in their endeavors to discover and develop better catalysts for organic
synthesis and to construct, through innovative chemistry
and biology, libraries of novel compounds for biological
screening. Their click chemistry, which has had a major
impact in many areas of the molecular sciences, continues to be an important focus of their research.
Members of my own group continue to explore chemical synthesis and chemical biology, with a focus on the
total synthesis of new anticancer agents, antibiotics,
marine-derived neurotoxins, antimalarial compounds,
and other bioactive natural and designed molecules.
Julius Rebek and his group devise biomimetic receptors, including molecules that bind neurotransmitters and
membrane components, for studies in molecular recognition. Larger host receptors can surround 3 or more
molecular guests and act as chambers in which the
chemical reactions of the guests are accelerated. Scientists in the group also synthesize small molecules that act
as protein helix mimetics for pharmaceutical applications.
Peter Schultz and researchers in his laboratory are
expanding the number of genetically encoded amino acids
to include fluorescent, photocaged, metal-binding, chemically reactive, and posttranslationally modified amino
acids. These scientists have also adapted this technology to mammalian cells and are applying these tools in
basic and applied problems in cell biology. In addition,
members of the group have used cell-based screens to
identify small molecules that selectively differentiate
and expand embryonic and adult stem cells and reprogram lineage-committed cells, as well as novel genes
and small molecules that affect a number of physiologic
and disease processes.
Chi-Huey Wong and his group further advance the
fields of chemoenzymatic organic synthesis, chemical
glycobiology, and the development of enzyme inhibitors.
A new strategy for the synthesis of glycoproteins based
on sugar-assisted glycopeptide ligation has been developed.
The programmable 1-pot synthesis of oligosaccharides
developed by this group has been further used in the
assembly of glycoarrays for study of saccharides that
bind to proteins. Members of this group also developed
new probes to study posttranslational glycosylation and
identify glycoprotein biomarkers associated with cancer.
Researchers in Dale Boger’s laboratory continue their
work on chemical synthesis; combinatorial chemistry; het-
84 CHEMISTRY
2008
erocycle synthesis; anticancer agents, such as vinblastine,
cyclostatin, chlorofusion, and yatakemycin; and antibiotics,
such as vancomycin, teicoplanin, and ramoplanin.
Scientists in Kim Janda’s laboratory conduct research
grounded on organic chemistry as applied to specific biological systems. The targeted programs span a wide range
of interests, from immunopharmacotherapy to biological
and chemical warfare agents to filarial infections such
as “river blindness” to quorum sensing in bacteria and
new cancer therapeutic strategies. Recent achievements
include in vivo detection of botulinum neurotoxin antagonists, the development of peptides and antibodies as
drug delivery modules that home to cancer cells and
active vaccines for nicotine addiction and obesity that
are now in preclinical trials.
M. Reza Ghadiri and his group are making important
contributions in the design and study of a new generation
of antimicrobial agents, based on self-assembling peptide
nanotube architecture, to combat multidrug resistant
infections. In addition, members of the group continue
to make novel contributions in several ongoing basic
research endeavors, such as designing biosensors, developing molecular computation, designing self-reproducing
systems, understanding the origins of life, and creating
emergent chemical systems.
M.G. Finn and his group have pioneered the use of
virus particles as chemical reagents and building blocks
for nanochemical structures. This effort is directed toward
the development of new diagnostics for disease and catalysts for organic reactions. Members of Dr. Finn’s laboratory also develop and investigate new organic and
organometallic reactions and use these processes to
synthesize biologically active compounds.
Jeff Kelly and his group are exploring the interface
between the chemistry, biology, and pathobiology of
proteome maintenance. The aim of their projects is to
understand the physical and biological basis of protein
folding and the competitive misfolding and aggregation
processes that lead to age-associated neurodegenerative
diseases. Information on proteome maintenance is used
to develop new small-molecule therapeutic strategies for a
variety of diseases, including neurodegenerative diseases.
Anita Wentworth and the researchers in her group are
investigating the chemical basis of complex disease states
and are synthesizing peptide- and small molecule–based
therapeutic agents. These scientists focus on disease
states in which inflammatory and reactive oxygen species are prominent, such as atherosclerosis, Alzheimer’s
disease, and other diseases of ageing.
THE SCRIPPS RESEARCH INSTITUTE
Researchers in Floyd Romesberg’s laboratory are using
diverse techniques ranging from bioorganic and biophysical chemistry to bacterial and yeast genetics to understand and manipulate the process of evolution. Major
efforts include designing unnatural base pairs and using
directed evolution of DNA polymerases to efficiently synthesize unnatural DNA containing the base pairs, using
spectroscopy to understand biological function and how
it evolves, and understanding how induced and adaptive mutations contribute to evolution in eukaryotic and
prokaryotic cells.
Phil Baran and his group are interested in how the
general challenge of chemoselectivity in organic chemistry
can be answered through the auspices of total synthesis. He and his coworkers have developed extremely
concise chemical solutions to the synthetic challenges
posed by numerous families of natural products. These
syntheses systematically tackle the issue of chemoselectivity and are characterized by striking brevity, new
biosynthetic postulates, the invention of new methods,
and a minimum use or complete absence of protecting
groups and superfluous oxidation state manipulations.
The Frontiers in Chemistry Lecturers (19th Annual
Symposium) for the 2007–2008 academic year were
M. Christina White, University of Illinois; Ben L. Feringa,
University of Groningen, the Netherlands; Ian Paterson,
Cambridge University; and Harry Noller, University of
California, Santa Cruz. In addition, we enjoyed hosting
the following professors: Samir Zard, Ecole Polytechnique, France, as the Bristol-Myers Squibb Lecturer;
E.J. Corey, Harvard University, as the Pfizer Lecturer;
and Robert Bergman, University of California, Berkeley,
as the Novartis Lecturer.
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
85
INVESTIGATORS’ R EPORTS
Practical Total Synthesis of
Natural Products
P.S. Baran, S. Ashida, N.Z. Burns, K. Chen, M.P. DeMartino,
K.J. Eastman, C.A. Guerrero, T. Gulder, B.D. Hafensteiner,
Y. Ishihara, P.J. Krawczuk, C. Li, D.W. Lin, J.W. Lockner,
M.R. Luzung, T.J. Maimone, T. Masuda, T.R. Newhouse,
D.P. O’Malley, H. Renata, J.M. Richter, N. Schone,
I.B. Seiple, R.A. Shenvi, J. Shi, H. Shigeshisa, S. Su,
A.F. Voica, J. Yamaguchi, I.S. Young
rom penicillin to paclitaxel (Taxol), natural products have an unparalleled track record in the betterment of human health. In fact, 9 of the top 20
best-selling drugs were either inspired by or derived from
natural products. Even the best-selling drug of all time,
atorvastatin (Lipitor), was based on a natural product
lead. Total synthesis, the art and science of recreating
these entities in the laboratory, invariably leads to fundamental discoveries in chemistry, biology, and medicine.
We focus on solving interesting challenges in the
total synthesis of natural products and on bridging gaps
in synthetic capabilities by inventing new reactions.
Through judicious target selection and creative retrosynthetic analyses, total synthesis becomes an engine for
discovery that drives the field of organic chemistry to
new levels of sophistication and practicality. Synthetic
organic chemistry requires tremendous ingenuity, artistic taste, experimental acumen, persistence, and character. Not surprisingly, drug development relies on the
expertise of researchers who have these characteristics.
Although we focus entirely on educating students in
fundamental chemistry, we also collaborate with expert
biologists to explore the medicinal potential of newly
synthesized natural products and the products’ analogs.
Recently completed total syntheses (Fig. 1) include
(1) the anticancer agent stephacidin; (2) the antibacterial agents ageliferin and axinellamine; (3) members
of the bioactive fischerindole, hapalindole, and welwitindolinone indole alkaloid family; (4) the anticancer agent
haouamine A; and (5) the structurally exotic marine
alkaloid chartelline C. Current natural product targets
(Fig. 2) include stylissadine A, sarcodonin, and vinigrol;
the neuroactive agent psychotrimine; and the potent
angiogenic agent cortistatin A.
F
F i g . 1 . Recently completed total syntheses.
PUBLICATIONS
Burns, N.Z., Baran, P.S. On the origin of the haouamine alkaloids. Angew. Chem.
Int. Ed. 46:205, 2007.
Chen, K., Richter, J.M., Baran, P.S. 1,3-Diol synthesis via controlled, radicalmediated C-H functionalization. J. Am. Chem. Soc. 130:7247, 2008.
Grube, A., Immel, S., Baran, P.S., Köck, M. Massadine chloride: a biosynthetic
precursor of massadine and stylissadine. Angew. Chem. Int. Ed. 46:6721, 2007.
Köck, M., Grube, A., Seiple, I., Baran, P.S. The pursuit of palau’amine. Angew.
Chem. Int. Ed. 46:6586, 2007.
Maimone, T.J., Voica, A.F., Baran, P.S. A concise approach to vinigrol. Angew.
Chem. Int. Ed. 47:3054, 2008.
86 CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
SYNTHETIC METHODS
Central to much of our work are investigations to
develop and apply the hetero Diels-Alder reaction, including the use of heterocyclic and acyclic azadienes (Fig. 1),
the thermal reactions of cyclopropenone ketals, inter-
F i g . 1 . N-Sulfonyl-1-aza-1,3-butadiene Diels-Alder reaction.
F i g . 2 . Recent natural product targets.
O’Malley, D.P., Yamaguchi, J., Young, I.S., Seiple, I.B., Baran, P.S. Total synthesis
of (±)-axinellamines A and B. Angew. Chem. Int. Ed. 47:3581, 2008.
Richter, J.M., Whitefield, B., Maimone, T.J., Lin, D.W., Castroviejo, P., Baran,
P.S. Scope and mechanism of the direct indole and pyrrole couplings adjacent to
carbonyl compounds: total synthesis of acremoauxin A and oxazinin 3. J. Am.
Chem. Soc. 129:12857, 2007.
Shenvi, R.A., Guerrero, C.A., Shi, J., Li, C.C., Baran, P.S. Synthesis of (+)-cortistatin A. J. Am. Chem. Soc. 130:7241, 2008.
Yamaguchi, J., Seiple, I.B., Young, I.S., O’Malley, D.P., Maue, M., Baran, P.S.
Synthesis of 1,9-dideoxy-pre-axinellamine. Angew. Chem. Int. Ed. 47:3578, 2008.
Synthetic and Bioorganic
Chemistry
D.L. Boger, E. Anderson, S. Baraldi, K. Boyle, C. Burke,
R. Clark, D. Colby, C. Crane, J. DeMartino, J. Elsner, C. Ezzili,
J. Garfunkle, H. Ge, D. Hochstatter, I. Hwang, R. Jones,
H. Kakei, D. Kato, F.S. Kimball, J. Lajiness, S. Lee, C. Liu,
K. MacMillan, J. Nam, K. Otrubova, P. Patel, W. Robertson,
Y. Sasaki, M. Schnermann, S. Seto, C. Slown, S. Stamm,
J. Stover, S. Takizawa, A. Tam, P. Va, L. Whitby, J. Xie, A. Zuhl
he research interests of our group include the total
synthesis of natural products, development of new
synthetic methods, heterocyclic chemistry, bioorganic and medicinal chemistry, the study of DNA-agent
interactions, and the chemistry of antitumor antibiotics.
We place a special emphasis on investigations to define
the structure-function relationships of natural or designed
organic agents.
T
molecular and intramolecular acyl radical–alkene addition
reactions, medium- and large-ring cyclization technology, and solution-phase combinatorial chemistry. In
each instance, the development of the methods represents the investigation of chemistry projected as a key
element in the synthesis of a natural or designed agent.
T O TA L S Y N T H E S I S O F N AT U R A L P R O D U C T S
Efforts are under way on the total synthesis of a
number of natural products that constitute agents in
which we have a specific interest. Representative agents
currently under study include (+)-CC-1065 and functional analogs; the duocarmycin class of antitumor
antibiotics, including yatakemycin; tropoloalkaloids;
prodigiosin and roseophilin; the deoxybouvardin and
RA-I class of antitumor agents; vancomycin, teicoplanin,
ristocetin, chloropeptins, and related agents; ramoplanin;
the luzopeptins, quinoxapeptins, thiocoraline, BE-22179,
and sandramycin; bleomycin A2 and functional analogs;
HUN-7293; chlorofusin; CI-920 (fostriecin) and cytostatin; the combretastatins; storniamide A; phomazarin;
ningalins; lamellarin O; lukianol A; piericidins; nothapodytine and mappicine; rubrolone; vindoline; and vinblastine (Figs. 2 and 3).
BIOORGANIC CHEMISTRY
The agents listed in the previous paragraph were
selected on the basis of their properties; in many
instances, they are agents related by a projected property. For example, (+)-CC-1065, the duocarmycins,
and yatakemycin are antitumor antibiotics and related
sequence-selective DNA minor groove alkylating agents.
Representative of such efforts, studies to determine the
structural features of yatakemycin and the duocarmycins
that contribute to the sequence-selective DNA alkylation
properties of these agents have resulted in the identifi-
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
87
Fig. 3. Additional recent total syntheses.
F i g . 2 . Recent total syntheses.
cation of a unique source of catalysis for the DNA alkylation reaction. Efforts are under way to develop DNA
cross-linking agents of a predefined cross-link, to fur-
ther understand the nature of the noncovalent and covalent interactions between agents and DNA, and to apply
this understanding to the de novo design of DNA-binding
and DNA-effector agents. Techniques for the evaluation
of the agent-DNA binding and alkylation properties, col-
88 CHEMISTRY
2008
laborative efforts in securing biological data, nuclear
magnetic resonance structures of DNA-agent complexes,
molecular modeling, and studies of DNA-agent interactions are integral parts of the program.
Additional ongoing studies include efforts to define
the fundamental basis of the DNA-binding or cleavage
properties of bleomycin A2, sandramycin, and the luzopeptins; to design inhibitors of the folate-dependent
enzymes glycinamide ribonucleotide transformylase and
aminoimidazole carboxamide ribonucleotide transformylase as potential antineoplastic agents; to establish
the chemical and biological characteristics responsible
for the sleep-inducing properties of the endogenous
lipid oleamide; to inhibit tumor growth through inhibition
of angiogenesis; to inhibit aberrant gene transcription
associated with cancer; and to control intracellular signal transduction through the discovery of antagonists
or agonists that affect protein-protein interactions, including receptor dimerization.
PUBLICATIONS
Eubanks, L.M., Hixon, M.S., Jin, W., Hong, S., Clancy, C.M., Tepp, W.H., Baldwin, M.R., Malizio, C.J., Goodnough, M.C., Barbieri, J.T., Johnson, E.A., Boger,
D.L., Dickerson, T.J., Janda, K.D. An in vitro and in vivo disconnect uncovered
through high-throughput identification of botulinum neurotoxin A antagonists [published correction appears in Proc. Natl. Acad. Sci. U. S. A. 104:6490, 2008].
Proc. Natl. Acad. Sci. U. S. A. 104:2602, 2007.
Hardouin, C., Kelso, M.J., Romero, F.A., Rayl, T.J., Leung, D., Hwang, I., Cravatt, B.F., Boger, D.L. Structure-activity relationships of the α-ketooxazole inhibitors of fatty acid amide hydrolase. J. Med. Chem. 50:3359, 2007.
Ishikawa, H., Boger, D.L. Total synthesis of (–)- and ent-(+)-4-desacetoxy-5desethylvindoline. Heterocycles 72:95, 2007.
Jin, W., Trzupek, J.D., Rayl, T.J., Broward, M.A., Vielhauer, G.A., Weir, S.J.,
Hwang, I., Boger, D.L. A unique class of duocarmycin and CC-1065 analogues
subject to reductive activation. J. Am. Chem. Soc. 129:15391, 2007.
Lee, S.Y., Clark, R.C., Boger, D.L. Total synthesis, stereochemical reassignment,
and absolute configuration of chlorofusin. J. Am. Chem. Soc. 129:9860, 2007.
Nam, J., Shin, D., Rew, Y., Boger, D.L. Alanine scan of [L-Dap2]ramoplanin A2
aglycon: assessment of the importance of each residue. J. Am. Chem. Soc.
129:8747, 2007.
Romero, F.A., Du, W., Hwang, I., Rayl, T.J., Kimball, F.S., Leung, D., Hoover,
H.S., Apodaca, R.L., Breitenbucher, J.G., Cravatt, B.F., Boger, D.L. Potent and
selective α-ketoheterocycle-based inhibitors of the anandamide and oleamide
catabolizing enzyme, fatty acid amide hydrolase. J. Med. Chem. 50:1058, 2007.
Tichenor, M.S., MacMillan, K.S., Stover, J.S., Wolkenberg, S.E., Pavani, M.G.,
Zanella, L., Zaid, A.N., Spalluto, G., Rayl, T.J., Hwang, I., Baraldi, P.G., Boger,
D.L. Rational design, synthesis, and evaluation, of key analogues of CC-1065 and
the duocarmycins. J. Am. Chem. Soc. 129:14092, 2007.
Tichenor, M.S., MacMillan, K.S., Trzupek, J.D., Rayl, T.J., Hwang, I., Boger, D.L.
Systematic exploration of the structural features of yatakemycin impacting DNA
alkylation and biological activity. J. Am. Chem. Soc. 129:10858, 2007.
Xu, L., Chong, Y., Hwang, I., D’Onofrio, A., Amore, K., Beardsley, G.P., Li, C.,
Olson, A.J., Boger, D.L., Wilson, I.A. Structure-based design, synthesis, evaluation, and crystal structures of transition state analogue inhibitors of inosine
monophosphate cyclohydrolase. J. Biol. Chem. 282:13033, 2007.
THE SCRIPPS RESEARCH INSTITUTE
Phage Escape for the Prediction
of Protein Evolution
T.J. Dickerson, J. Denery, L. Eubanks, A. Hoyt, K.D. Janda,
A. Moreno, Y. Nakai, A. Nguyen, A. Nunes, A. Rohrbach,
C. Saccavini
he relationship between host and pathogen is in
a constant state of flux, with each side continually
evolving in a struggle to maximize the chance for
survival. A plethora of defensive systems has evolved to
counteract and/or eliminate invading pathogens. These
systems exert pressure upon the pathogen, leading to
mechanisms that combat the host, including immune
evasion and drug resistance. Subsequently, the host
must counter these improved pathogens with a selected
adaptive immune response. This cycle can continue
indefinitely, with pathogen and host counteracting the
survival strategy of each other.
From a structural point of view, the contest is usually played out between the relatively unstructured protein
loops of both systems, where generation of 3-dimensional structural diversity of these parts of the protein
does not affect overall protein function. The accessible
protein-sequence diversity of both pathogen and host
immune system is exceedingly expansive, thereby making an a priori analysis of where the mutation can occur
and how antibodies subsequently respond an extremely
difficult challenge. However, adaptive immunity is inherently a reactive system and cannot operate in a proactive
mode; challenge from an exogenous antigen is required
before a response is made. Thus, despite the seemingly
limitless diversity that can be accessed by the mammalian immune system, only a small part of this theoretical
diversity is present at any given time. Only after a specific challenge is the full extent of the immune repertoire
brought to bear upon an invading pathogen. A technique
that could be used to predict the advance of evolving
human disease and predetermine suitable therapeutic
strategies before a pathogen becomes a public health
threat would be valuable both for understanding pathogen evolution and for transforming drug discovery into a
process that predetermines therapeutic strategies before
the onset of epidemics or pandemics.
Recently, we developed a technology termed “phage
escape” that allows a preemptive determination of the
evolutionary pathways used by the causative organisms
of specific diseases. This technology has been applied
to the evolution of the hemagglutinin viral surface pro-
T
CHEMISTRY
2008
tein from the highly pathogenic avian influenza virus
H5N1 (“bird flu”). During the past year, we successfully prepared all of the experimental tools needed to
perform a “checkmate analysis” of influenza virus hemagglutinin type 5. Currently, we are generating a map of
the evolution of this protein (Fig. 1).
THE SCRIPPS RESEARCH INSTITUTE
89
Chemical and Functional
Genomic Approaches to Stem
Cell Biology and Regenerative
Medicine
S. Ding, R. Abu-Jarour, R. Ambasudhan, R. Coleman,
C. Desponts, J. Efe, H.S. Hahm, S. Hilcove, J. Hsu, W. Li,
T. Lin, Y. Shi, W. Xiong, Y. Xu, X. Yuan, H. Zhou
ecent advances in stem cell biology may make
possible new approaches for the treatment of a
number of diseases, including cardiovascular
disease, neurodegenerative disease, musculoskeletal
disease, diabetes, and cancer. These approaches could
involve cell replacement therapy and/or drug treatment
to stimulate the body’s own regenerative capabilities
by promoting survival, migration/homing, proliferation,
and differentiation of endogenous stem/progenitor cells.
However, such approaches will require identification of
renewable cell sources of engraftable functional cells, an
improved ability to manipulate proliferation and differentiation of the cells, and a better understanding of
the signaling pathways that control the fate of the cells.
Equipped with a high-throughput screening platform
and large arrayed molecular libraries—combinatorial
chemical libraries, genome-scale cDNA libraries (for
gain of function), and small interfering RNA libraries
(for loss of function)—we are developing and integrating
chemical and functional genomic tools to study stem
cell biology and regeneration. We screen these libraries
to identify and further characterize small molecules and
genes that can control stem cell fate in various systems,
including (1) self-renewal regulation of embryonic and
adult stem cells; (2) directed and step-wise differentiation of embryonic stem cells toward neuronal, cardiac,
and pancreatic lineages; (3) directed neuronal differentiation and subtype neuronal specification of human
and rodent neural stem cells; (4) cellular plasticity and
reprogramming of lineage-restricted somatic cells to
more primitive precursor cells; (5) functional proliferation of cardiomyocytes and pancreatic beta cells from
adults; (6) developmental signaling pathways and epigenetic mechanisms (histone and DNA de/methylation);
and (7) development of new technologies for stem cell
derivation and gene targeting.
In addition, we are characterizing the molecular
mechanism of these identified small molecules and
R
F i g . 1 . Work flow for using phage escape technology to identify
escape mutants. In an immunologic checkmate analysis, phagebound protein variants (escape mutant, right middle) are able to
escape a known collection of neutralizing antibodies (top center).
PUBLICATIONS
Brogan, A.P., Dickerson, T.J., Janda, K.D. Nornicotine-organocatalyzed aqueous
reduction of α,β-unsaturated aldehydes. Chem. Commun. (Camb.) Issue 46:4952,
2007.
Capková, K., Yoneda, Y., Dickerson, T.J., Janda, K.D. Synthesis and structureactivity relationships of second-generation hydroxamate botulinum neurotoxin A
protease inhibitors. Bioorg. Med. Chem. Lett. 17, 6463, 2007.
Dickerson, T.J., McKenzie, K.M., Hoyt, A.S., Wood, M.R., Janda, K.D., Brenner,
S.B., Lerner, R.A. Phage escape libraries for checkmate analysis. Proc. Natl. Acad.
Sci. U. S. A. 104:12703, 2007.
Eubanks, L.M., Dickerson, T.J. Investigating novel therapeutic targets and molecular mechanisms to treat botulinum neurotoxin A intoxication. Future Microbiol.
2:677, 2007.
Ino, A., Dickerson, T.J., Janda, K.D. Positional linker effects in haptens for cocaine
immunopharmacotherapy. Bioorg. Med. Chem. Lett. 17:4280, 2007.
Lowery, C.A., Dickerson, T.J., Janda, K.D. Interspecies and interkingdom communication mediated by bacterial quorum sensing. Chem. Soc. Rev. 37:1337, 2008.
Treweek, J., Wee, S., Koob, G.F., Dickerson, T.J., Janda, K.D. Self-vaccination by
methamphetamine glycation products chemically links chronic drug abuse and cardiovascular disease. Proc. Natl. Acad. Sci. U. S. A. 104:11580, 2007.
Willis, B., Eubanks, L.M., Dickerson, T.J., Janda, K.D. The strange case of the
botulinum neurotoxin: using chemistry and biology to modulate the most deadly
poison. Angew. Chem. Int. Ed. 47:8360, 2008.
Willis, B., Eubanks, L.M., Wood, M.R., Janda, K.D., Dickerson, T.J., Lerner, R.A.
Biologically templated organic polymers with nanoscale order. Proc. Natl. Acad.
Sci. U. S. A. 105:1416, 2008.
Yao, Y., Dickerson, T.J., Hixon, M.S., Dyson, H.J. NMR detection of adventitious
binding of xylose to the quorum-sensing protein SdiA of Escherichia coli. Bioorg.
Med. Chem. Lett. 17:6202, 2007.
90 CHEMISTRY
2008
genes by using various approaches, including detailed
investigations of structure-activity relationship, affinity
chromatography for target identification, transcriptome
profiling, proteomics analysis, chemical/genetic epistasis,
and biochemical and functional assays in vitro and in
vivo. So far, we have identified and are characterizing
functional small molecules and/or genes in each of the
previously mentioned distinct biological processes that
involve regulation of stem/progenitor cells.
More recent examples include identification and
characterization of distinct small molecules for selfrenewal of human embryonic stem cells and clonal
expansion/survival; dopaminergic neuron specification
from mouse embryonic stem cells; derivation of rat
embryonic stem cells; reprogramming of somatic cells
to a pluripotent state; definitive endoderm and pancreatic induction; chemically defined monolayer conditions
for self-renewal of embryonic stem cells and their directed
differentiation to cardiac lineages; proliferation of human
beta cells; and regulating Wnt signaling. These studies
may ultimately facilitate the therapeutic application of
stem cells and the development of small-molecule drugs
to stimulate tissue and organ regeneration in vivo.
PUBLICATIONS
Shi, Y., Do, J.-T., Desponts, C., Hahm, H.-S., Schöler, H.R., Ding, S. A combined
chemical and genetic approach for the generation of induced pluripotent stem cells.
Cell Stem Cell 2:525, 2008.
Xu, Y., Shi, Y., Ding, S. A chemical approach to stem-cell biology and regenerative
medicine. Nature 453:338, 2008.
THE SCRIPPS RESEARCH INSTITUTE
F i g . 1 . Structural simplification of α- L -threofuranosyl-(3′g2′)
nucleic acid, which was inspired by studies on (3′g4′)-lyxopyranosyl nucleic acid, gives rise to acyclic informational oligomeric
systems. Two examples are shown: glycerol nucleic acid and glyceric acid nucleic acid.
The structure of this oligomeric system is based on a
structural simplification of the oligonucleotides containing lyxopyranosyl (2′g4′)– and threofuranosyl (2′g3′)–
linked phosphodiester backbones, which we studied
previously. Among the oligomer systems depicted in Figure 1, the nucleic acid derived from the glycerol backbone
is not considered to be a potentially prebiotic system, in
contrast to the oligomer system derived from glyceric acid
and tagged via amide bonds with 5-aminopyrimidines.
We have completed the synthesis of such a glyceric acid–derived oligomer containing six 5-aminouracil
units (6-mer) and have studied its base-pairing properties with DNA, RNA, and α-L-threofuranosyl-(3′g2′)
nucleic acid. Base pairing was strong between the
6-mer and poly(dA) (Fig. 2), was somewhat weaker
Chemical Etiology of Nucleic
Acid Structure
A. Eschenmoser, R. Krishnamurthy, G.K. Mittapalli,
R.R. Kondreddi, Y. Osornio, V.S. Naidu
n the general context of our project to map the landscape of potentially primordial informational oligomer
systems, we have been working during the past year
on the following topics.
I
2 ′g 3 ′- P H O S P H O D I E S T E R – L I N K E D G LY C E R I C A C I D
O L I G O M E R S B A S E D O N 5 - A M I N O P Y R I M I D I N E – TA G G E D
BACKBONES
We have undertaken the synthesis and study of the
base-pairing properties of oligomers derived from a
2′,3′-phosphodiester–linked glyceric acid backbone that
has 2,4-disubsituted 5-aminopyrimidines, attached to
the carboxyl group of glyceric acid via an amide bond
at the 5-amino group, as recognition elements (Fig. 1).
F i g . 2 . UV (left) and circular dichroism (right) spectroscopic data
for base pairing between 5-aminouracil–tagged 2-phosphoglycerate
hexamer and DNA, poly(dA); c = 5+5 µM. Measurements were made
in phosphate buffer.
with the corresponding poly(rA), and even occurred with
α-L-threofuranosyl-(3′g2′) nucleic acid. We are expanding the study to include the complementary partner
strand tagged with 2,4,5-triaminopyrimidine and have
explored different pathways for synthesizing the suitably protected building blocks necessary for the automated oligonucleotide synthesis.
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
91
SYNTHESIS OF OLIGODIPEPTIDES OF
A S PA R T Y L - 3 - A M I N O A L A N I N E D I P E P T I D E TA G G E D
WITH OROTIC ACID
In our search for alternative heterocycles that would
be potentially prebiotic and offer opportunities for
becoming chemoselectively attached to a backbone,
we considered orotic acid as a candidate. Orotic acid
and its 5-substituted derivatives have been identified
as products from the hydrolysis of polymeric material
formed from hydrogen cyanide. In addition, the involvement of orotic acid as a nucleobase in the biosynthetic
assembly of pyrimidine nucleotides justifies and warrants exploration of its base-pairing properties. We are
synthesizing the necessary building blocks for the synthesis and study of the base-pairing properties of oligomers consisting of aspartyl-3-aminoalanine dipeptide
units tagged with orotic acid (Fig. 3). The choice of the
F i g . 4 . Hypothetical autocatalytic cycle for the dimerization of
glyoxylate to dihydroxyfumarate and the biomolecules to be derived
from the constituents of that cycle.
with the constraints of a primordial chemistry. We are
conducting exploratory studies for assessing the chemistry of selected intermediates postulated to be formed
from the chemistry of glyoxylate and dihydroxyfumarate.
Some of the preliminary results are encouraging.
F i g . 3 . Orotic acid as a recognition element. Also shown is the
oligodipeptide tagged with orotic acid.
oligodipeptide backbone on which orotic acid would be
attached was influenced by the results of our previous
studies. Because the carboxyl group is now on the
heterocycle, amide bond–mediated tagging of the carboxyl group of orotic acid requires a 3-aminoalanine
as a tagging unit.
E X P L O R I N G T H E C H E M I S T R Y O F G LY O X Y L AT E A N D
D I H Y D R O X Y F U M A R AT E
A research project such as mapping the landscape
of potentially primordial informational oligomer systems
eventually demands the conception of, and the commitment to, a detailed chemical scenario for the type of
organic chemistry that is supposed to have led to such
oligomers under primordial conditions. Figure 4 depicts
the chemical nature of the scenario we have decided to
study experimentally. In the reaction cycle shown, glyoxylate would autocatalytically convert itself into its
dimer dihydroxyfumarate. Dihydroxyfumarate is a known
compound that we postulate can act as a common starting material for a large variety of biomolecules, such as
sugars, α-amino acids, and pyrimidines, and for other
organics of etiologic interest by reactions that are essentially unexplored thus far but are deemed compatible
PUBLICATIONS
Eschenmoser, A. On a hypothetical generational relationship between HCN and
constituents of the reductive citric acid cycle. Chem. Biodivers. 4:554, 2007.
Eschenmoser, A. The search for the chemistry of life’s origin. Tetrahedron
63:12821, 2007.
Koch, K., Schweizer, B., Eschenmoser. A. Reactions of the HCN-tetramer with
aldehydes. Chem. Biodivers. 4:541, 2007.
Organic, Medicinal, and
Biological Chemistry
M.G. Finn, A. Accurso, S. Brown, S.-H. Cho, V. Hong, J. Lau,
S. Lee, Y.-H. Lim, S. Presolski
n addition to our work on biological polyvalency and
immunology with engineered virus particles, supported by the Skaggs Institute for Chemical Biology,
we focus on the development of catalysts and the synthesis of biologically useful structures. Two of these projects are described in the following sections.
I
C O P P E R - C ATA LY Z E D A Z I D E - A L K Y N E C L I C K
CHEMISTRY
We have continued our development of new catalysts and conditions for the copper-catalyzed azide-
92 CHEMISTRY
2008
alkyne cycloaddition reaction, which has become a principal example of click chemistry in the synthesis of possible drugs, dendrimers, polymers, and functionalized
surfaces in laboratories around the world. In the past
year, using an active but highly air-sensitive catalyst, we
developed a convenient electrochemical protocol for
performing bioconjugations. This procedure enables
investigators who lack sophisticated inert-atmosphere
equipment to perform the reaction under demanding
conditions. We have also discovered new derivatives of
the (benzimidazole-methyl)amine ligands reported last
year, which accelerate the copper-catalyzed azide-alkyne
cycloaddition reaction to a remarkable degree. A comprehensive picture is rapidly emerging of the types of
ligands effective under the diverse conditions in which
this cycloaddition reaction is applied.
An important application of click chemistry is the
synthesis and modification of polymeric materials. We
found that metal adhesives can be formed by the simple
application of mixtures of polyvalent azide and alkyne
compounds to copper-containing surfaces. By incorporating amino groups to help speed the click reaction, and
flexible cross-linking molecules to protect against stress
fracturing in the resulting polymers, greatly improved
adhesives have been created. Figure 1 illustrates the
F i g . 1 . Graduate student Vu Hong sits on a 20-L container filled
with water, supported by 2 copper plates glued together with an
adhesive prepared by graduate student Adrian Accurso.
THE SCRIPPS RESEARCH INSTITUTE
strength of one of these formulations, which has potential in such applications as protective coatings, electrically conducting junctions, and antifouling agents.
A G E N T S W I T H A C T I V I T Y A G A I N S T H E PAT I T I S B
VIRUSES: MISDIRECTING PROTEIN-PROTEIN
INTERACTIONS
Modulation of protein-protein contacts by small
molecules is an attractive strategy for the development
of biologically active compounds. In many instances,
the target interprotein interaction covers a substantial
landscape with high thermodynamic stability. Virus
particles rely on the assembly of protein subunits that
engage in well-defined protein-protein interactions.
However, these interactions are necessarily weak until
the late stages of assembly; such cooperativity is necessary to ensure that protein is efficiently used in the
multistep construction process.
Viral capsid intermediates are therefore a class of
protein-protein interface targets that may be easier for
small molecules to affect. We have investigated this possibility for hepatitis B virus (HBV), a pathogen that
infects 400 million people worldwide. The antiviral activity of the heteroaryldihydropyrimidine class of compounds has been known for several years. We found that
their mechanism of action is the distortion, rather than
the inhibition, of the protein assembly process. In a type
of molecular jujitsu, these small molecules use the
natural interactions of the capsid proteins against the
virus, by increasing the energy of protein subunit association and the rate of subunit aggregation. This increase
causes the viral proteins to assemble in error-prone
fashion, forming large and irregular structures rather
than the symmetric particles necessary for the proper
function of the virus (Fig. 2).
By making many heteroaryldihydropyrimidine analogs, we found compounds that nucleate the misincorporation of hundreds of protein molecules at a time,
allowing these compounds to be highly effective inhibitors in cell culture. We continue to improve the performance of these structures and test the best analogs
for activity against HBV in animal models. Furthermore, we think that assembly misdirection of this kind
is a general antiviral strategy. By attacking the relatively easy target of protein-protein interactions present in early virus assembly intermediates, selective
agents can be developed in a new and effective way.
PUBLICATIONS
Destito, G., Yeh, R., Rae, C.S., Finn, M.G., Manchester, M. Folic acid-mediated targeting of cowpea mosaic virus particles to tumor cells. Chem. Biol. 14:1152, 2007.
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
93
Zhang, Q., Ma, X., Ward, A., Hong, W.-X., Jaakola, V.-P., Stevens, R.C., Finn,
M.G., Chang, G. Designing facial amphiphiles for the stabilization of integral membrane proteins. Angew. Chem. Int. Ed. 46:7023, 2007.
Organometallic Catalysis in
Synthesis, Bioorganic Chemistry,
and Materials Research
V.V. Fokin, A. Chanda, S. Chuprakov, J. Fotsing, S. Grecian,
T. Horneff, L. Krasnova, S.-W. Kwok, J. Raushel, T. Weide
ur goals are to discover new reactions and to
develop their practical applications in organic
synthesis, chemical biology, and materials science. Transformations catalyzed by transition metals
are central to our research. With many variables for
optimization, these reactions have great potential to
become useful on both laboratory and industrial scales.
Although we often use automation techniques to screen
extensive sets of catalysts, ligands, and conditions
(sometime on the basis of just a hunch that “it should
work”), mechanistic investigations of the initial reactivity
are prominent in our research. Analysis of reaction kinetics, in situ infrared monitoring, reaction heat flow
calorimetry, and other physicochemical methods are
routinely used to understand the mechanistic underpinnings of the processes under investigation. Computational studies of reactive intermediates and mechanistic
pathways compliment these experimental techniques.
Equipped with reactions that proceed reliably with a
range of readily available starting materials and are
experimentally simple to perform, we endeavor to
assemble molecules with interesting and useful function, ranging from new biologically active small molecules to macromolecular scaffolds such as dendrimers
for studying multivalency effects and polymers for use
in coatings and adhesives.
We have focused on metal-catalyzed reactions of
acetylenes because such processes can reliably produce
a plethora of heterocyclic systems. Copper(I)- and ruthenium(II)-catalyzed reactions of acetylenes with 1,3-dipoles
developed in our laboratories are shown in Figure 1.
Reactions of alkynes with organic azides take a special
place in the arsenal of catalytic dipolar cycloadditions
we use. Although azides are energetic compounds, they
are quite inert: stable to water, oxygen, most functional
groups found in biological molecules, and many of the
common organic reagents and conditions. With acety-
O
F i g . 2 . Negative-stain electron micrographs of the assembly prod-
ucts of HBV capsid protein induced by different heteroaryldihydropyrimidine derivatives. HBV capsids are typically 35 nm in diameter;
the structures shown here are many times that size.
Hong, V., Udit, A.K., Evans, R.A., Finn, M.G. Electrochemically protected
copper(I)-catalyzed azide-alkyne cycloaddition. Chembiochem 9:1481, 2008.
Kaltgrad, E., O’Reilly, M.K., Liao, L., Han, S., Paulson, J., Finn, M.G. On-virus
construction of polyvalent glycan ligands for cell-surface receptors. J. Am. Chem.
Soc. 130:4578, 2008.
Kaltgrad, E., Sen Gupta, S., Punna, S., Huang, C.-Y., Chang, A., Wong, C.-H.
Finn, M.G., Blixt, O. Anti-carbohydrate antibodies elicited by polyvalent display on
a viral scaffold. Chembiochem 8:1455, 2007.
Liu, Y., Díaz, D.D., Accurso, A.A., Sharpless, K.B., Fokin, V.V., Finn, M.G. Click
chemistry in materials synthesis, III: metal-adhesive polymers from Cu(I)-catalyzed
azide-alkyne cycloaddition. J. Polym. Sci. A Polym. Chem. 45:5182, 2007.
Miermont, A., Barnhill, H., Strable, E., Lu, X., Wall, K.A., Wang, Q., Finn, M.G.,
Huang, X. Cowpea mosaic virus capsid: a promising carrier for the development of
carbohydrate based antitumor vaccines. Chem. Eur. J. 14:4939, 2008.
Portney, N.G., Tseng, R.J., Destito, G., Strable, E., Yang, Y., Manchester, M.,
Finn, M.G., Ozkan, M. Microscale memory characteristics of virus-quantum dot
hybrids. Appl. Phys. Lett. 90:214104, 2007.
Prasuhn, D.E., Jr., Singh, P., Strable, E., Brown, S., Manchester, M., Finn, M.G.
Plasma clearance of bacteriophage Qβ particles as a function of surface charge. J.
Am. Chem. Soc. 130:1328, 2008.
Rodionov, V.O., Presolski, S., Díaz, D.D., Fokin, V.V., Finn, M.G. Ligand-accelerated Cu-catalyzed azide-alkyne cycloaddition: a mechanistic report. J. Am. Chem.
Soc. 129:12705, 2007.
Rodionov, V.O., Presolski, S., Gardinier, S., Lim, Y.-H., Finn, M.G. Benzimidazole
and related ligands for Cu-catalyzed azide-alkyne cycloaddition. J. Am. Chem. Soc.
129:12696, 2007.
Strable, E., Prasuhn, D.E., Jr., Udit, A.K., Brown, S., Link, A.J., Ngo, J.T., Lander, G., Quispe, J., Potter, C.S., Carragher, B., Tirrell, D.A., Finn, M.G. Unnatural
amino acid incorporation into virus-like particles. Bioconjug. Chem. 19:866, 2008.
Zhang, Q., Horst, R., Geralt, M., Ma, X., Hong, W.-X., Finn, M.G., Stevens, R.C.,
Wüthrich, K. Microscale NMR screening of new detergents for membrane protein
structural biology. J. Am. Chem. Soc. 130:7357, 2008.
94 CHEMISTRY
F i g . 1 . Catalytic syntheses of heterocycles from alkynes and
1,3-dipolar species.
lenes, organic azides produce 1,2,3-triazoles, exceptionally stable 5-membered heterocycles. These reactions,
however, are very slow without a catalyst.
The copper- and ruthenium-catalyzed cycloadditions of azides and alkynes provide ready access to
1,2,3-triazoles of various substitution patterns. The
copper-catalyzed variant has become the premier click
reaction and is used in diverse applications by chemists
across the world in organic synthesis, medicinal chemistry, chemical biology, and materials science. In addition to its exceptional reliability and tolerance to a wide
range of functional groups, the reaction has provided
valuable insights into the unique and, until recently,
unexplored reactivity patterns of organic azides and
in situ generated copper acetylides. Indeed, catalytic
syntheses of isoxazoles and pyrazoles are the recent
additions to the family of such reactions. The ruthenium-catalyzed reaction (Fig. 1) enables “fusion” of
organic azides and both terminal and internal alkynes
with a complementary regioselectivity and appears to
proceed through a completely different mechanism.
This past year, we discovered the exquisite catalytic activity of pentamethylcyclopentadienyl ruthenium(II)
chloride complexes in reactions of alkynes with nitrile
oxides. These cycloadditions result in facile formation
of 3,4-disubstituted and fully substituted isoxazoles.
Until now, no general methods for regioselective synthesis of these heterocycles were available. When thermal cycloadditions of nitrile oxides with alkynes are
successful, they favor the formation of the 3,5-disubstituted isomers. Furthermore, examples of reactive
partners for nitrile oxides are limited to a handful of
highly activated, electron-deficient alkynes. Unactivated
or sterically hindered acetylenes usually do not react
2008
THE SCRIPPS RESEARCH INSTITUTE
at all. In contrast, in the presence of ruthenium complexes, nitrile oxides and both terminal and internal
alkynes are “fused” within minutes to hours at room
temperature. In addition to the immediate practical benefits, this transformation suggests that different dipoles
can be activated and engaged in catalysis by ruthenium
complexes. Such processes are being investigated.
Both copper- and ruthenium-catalyzed cycloadditions and their applications in the synthesis of biologically active compounds and novel materials have been
the subject of our intense attention during the past year.
We have investigated the mechanism of these processes
and have endeavored to develop new ligands to make
the reactions more efficient. We have used the reactions
to synthesize libraries of compounds for HIV protease
inhibition, in collaboration with J.H. Elder and A.J. Olson,
Department of Molecular Biology; nicotinic receptor
agonists and antagonists, in studies with P. Taylor,
University of California, San Diego; metallo-β-lactamase
inhibitors, in collaboration with P. Hodder, Translational
Research Institute; and polymeric materials and dendritic constructs for polyvalent display and imaging
applications (Fig. 2).
F i g . 2 . Small molecules synthesized by using azide-alkyne
cycloadditions.
CHEMISTRY
2008
PUBLICATIONS
Boren, B.C., Narayan, S., Rasmussen, L.K., Zhang, L., Zhao, H., Lin, Z., Jia, G.,
Fokin, V.V. Ruthenium-catalyzed azide-alkyne cycloaddition: scope and mechanism. J. Am. Chem. Soc. 130:8923, 2008.
Finn, M.G., Kolb, H.C., Fokin, V.V., Sharpless, K.B. Concept and applications of
click chemistry: from the standpoint of advocates. Kagaku to Kogyo 60:976, 2007.
Fokin, V.V. Click imaging of biochemical processes in living systems. ACS Chem.
Biol. 2:775, 2007.
Hawker, C.J., Fokin, V.V., Finn, M.G., Sharpless, K.B. Bringing efficiency to materials synthesis: the philosophy of click chemistry. Aust. J. Chem. 60:381, 2007.
Kalisiak, J., Sharpless, K.B., Fokin, V.V. Efficient synthesis of 2-substituted-1,2,3triazoles. Org. Lett. 10:3171, 2008.
THE SCRIPPS RESEARCH INSTITUTE
95
bond–directed ring stacking into open-ended hollow
tubular structures that have marked antibacterial and
antiviral activities in vitro. The effectiveness of this
novel supramolecular class of bioactive species as
selective antibacterial agents was highlighted by the
high efficacy of one of these antimicrobials against
lethal methicillin-resistant Staphylococcus aureus infections in mice. Currently, we are exploring rational design
of cyclic glycopeptides and selections from combinatorial libraries to discover novel antiviral supramolecular
compounds (Fig. 1).
Liu, Y., Díaz, D.D., Accurso, A.A., Sharpless, K.B., Fokin, V.V., Finn, M.G. Click
chemistry in materials synthesis, III: metal-adhesive polymers from Cu(I)-catalyzed
azide-alkyne cycloaddition. J. Polym. Sci. A Polym. Chem. 45:5182, 2007.
Rasmussen, L.K., Boren, B.C., Fokin, V.V. Ruthenium-catalyzed cycloaddition of
aryl azides and alkynes. Org. Lett. 9:5337, 2007.
Rodionov, V.O., Presolski, S.I., Díaz, D.D., Fokin, V.V., Finn, M.G. Ligand-accelerated Cu-catalyzed azide-alkyne cycloaddition: a mechanistic report. J. Am. Chem.
Soc. 129:12705, 2007.
Vestberg, R., Malkoch, M., Kade, M., Wu, P., Fokin, V.V., Sharpless, K.B., Drockenmuller, E., Hawker, C.J. Role of architecture and molecular weight in the formation of tailor-made ultrathin multilayers using dendritic macromolecules and click
chemistry. J. Polym. Sci. A Polym. Chem. 45:2835, 2007.
Yoo, E.J., Ahlquist, M., Bae, I., Sharpless, K.B., Fokin, V.V., Chang, S. Mechanistic
studies on the Cu-catalyzed three-component reactions of sulfonyl azides, 1-alkynes
and amines, alcohols, or water: dichotomy via a common pathway J. Org. Chem.
73:5520, 2008.
Design of Functional
Synthetic Systems
M.R. Ghadiri, M. Amorin, J. Beierle, A. Chavochi, J. Chu,
B. Frezza, N. Gianneschi, L. Leman, A. Loutchnikov,
A. Montero, C. Olsen, J. Picuri, D. Radu, Y. Ura
e are engaged in multidisciplinary research
to uncover new chemical and biochemical
approaches for the design of functional molecular, supramolecular, and complex self-organized systems. Our efforts span disciplines ranging from synthetic
organic, bioorganic, and physical organic chemistry to
nanotechnology, biophysics, enzymology, and molecular
biology. Current research includes the design of synthetic
peptide catalysts, antimicrobial self-assembling peptide
nanotubes, semisynthetic allosteric enzymes, self-replicating molecular systems and emergent networks, single-molecule DNA sensing, molecular computation, and
prebiotic chemistry.
W
ANTIMICROBIAL PEPTIDE NANOTUBES
We have shown that appropriately designed cyclic
peptide subunits can self-assemble through hydrogen
F i g . 1 . Antiviral agents based on self-assembling cyclic peptide
nanotubes. Cyclic D ,L-α-peptides act on endosomal membranes to
prevent the development of low pH in endocytic vesicles, arrest the
escape of virions from the endosome, and abrogate adenovirus
infection.
DESIGN OF SIGNAL SELF-AMPLIFYING DNA SENSORS
We constructed a novel sequence-specific DNA detection system based on rationally designed semisynthetic
enzymes. The system is composed of covalently associated inhibitor-DNA-enzyme modules that function via
DNA hybridization–triggered allosteric enzyme activation and signal amplification through substrate turnover
(Fig. 2). The functional capacity of the system is high-
F i g . 2 . Schematic representation of an intrasterically inactivated
inhibitor-DNA-enzyme construct (left) and the DNA hybridization–
triggered enzyme activation (right). The construct can be used to
sense low concentrations of cDNA because of its built-in capacity
for signal amplification via rapid substrate turnover.
lighted by the sequence-specific detection of approximately 10 fmol of DNA in less than 3 minutes under
physiologic conditions. Our studies suggest that ratio-
96 CHEMISTRY
2008
nally designed intrasterically regulated enzymes may be
a promising new class of reagents for highly sensitive,
rapid, and 1-step detection of label-free DNA sequences
that does not depend on polymerase chain reactions.
THE SCRIPPS RESEARCH INSTITUTE
ous solutions. The synthetic networks have some of the
basic architectural and dynamic features of the living
networks, reorganize in response to changes in environmental conditions and inputs (Fig. 4), and perform basic
SINGLE-MOLECULE DNA SEQUENCING
We are interested in the study of matter at the level
of single molecules. For these studies we use the transmembrane protein α-hemolysin as a rapid and highly
sensitive sensor element for stochastic analysis of the
molecules lodged or trapped inside the protein pore;
the analysis relies on detecting the perturbations in the
conductance levels produced in the ion channel in the
native protein. Using this technique, we developed an
approach by which a single-stranded DNA molecule can
be trapped in a specific configuration inside an α-hemolysin channel, manipulated, and studied with high sensitivity at the single-molecule level. We have been able
to detect up to 9 consecutive DNA polymerase–catalyzed
single-nucleotide primer extensions (Fig. 3) with high
F i g . 4 . Adaptive reorganization in a synthetic peptide network. The
graph structure or wiring of a synthetic peptide network responds dramatically to changes in the environmental stimuli (pH or salt content).
Boolean logic functions. We suggest that the ability
to rationally construct predictable chemical circuitry
might be useful in advancing the modeling and better
understanding of some of the basic dynamic information-processing characteristics of the more complex cellular networks.
M O L E C U L A R C O M P U TAT I O N
F i g . 3 . Single-molecule monitoring of DNA polymerase–catalyzed
single-nucleotide primer extensions with high sensitivity via an
α-hemolysin–DNA–rotaxane device.
sensitivity and spatial resolution (≤2.4 Å). The singlebase resolution of this approach and the ability to control the passage of DNA in single-base steps satisfy the
2 minimal requirements of a nanopore-based sequencing device.
COMPLEX SYNTHETIC NETWORKS
Living cells use complex networks of evolutionarily
selected biomolecular interactions and chemical transformations to process multiple extracellular input signals rapidly and simultaneously. We are interested in
understanding and experimentally modeling the organizational and functional properties of biological networks. We have developed a general strategy for the
design and construction of self-organized synthetic peptide networks based on the sequence-selective autocatalytic and cross-catalytic template-directed coiled
coil peptide fragment condensation reactions in aque-
A fundamental goal of computing is to reproduce
in a molecular setting the familiar properties of microelectronics, such as digital logic, component modularity,
and hierarchical design capacity. In this regard, significant advances have been made in the design of molecular logic gates by using small-molecule and rotaxane
complexes, deoxyribozymes, enzymatic biochemical
networks, peptide networks, and other systems. However, the molecular logic gates must be integrated into
more complex networks in which outputs from each gate
can serve as inputs to downstream gates.
We recently described the construction of a basis set
of DNA-based logic gates (AND, OR, AND-NOT) capable
of communicating with one another. These gates were
rewired into a higher-order circuit that enforces a net
XOR (Exclusive OR) Boolean behavior (Fig. 5), showing
that the components can be modularly recombined to
implement novel logic processing. Our results support
the notion that with a basis set of only a few logic gates
and within the limits imposed by the availability of
uniquely addressable oligonucleotide sequences, design
of molecular circuits capable of performing a large variety of digital logic operations might be within reach.
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
97
A Merging of Chemistry
and Biology
K.D. Janda, J. Ashley, K. Capková, S. De Lamo Marin,
J. Denery, T. Dickerson, A. Di Mola, B. Ellis, L. Eubanks,
K. Fukuchi, C. Hernandez, G. Kaufmann, C. Lowery,
F i g . 5 . A multilevel circuit built from OR, AND, and AND-NOT
gates that performs a net XOR (Exclusive-OR) analysis on the inputs.
PREBIOTIC CHEMISTRY
The emergence of a polymer that could store genetic
information, replicate, and exhibit phenotypic properties
subject to selective environmental pressures marked a
crucial stage in the transition from the prebiotic world
to biology; however, the nature of such a polymer remains
unresolved. We have discovered an oligomer family that
quickly and efficiently self-assembles via reversible covalent anchoring of nucleobase recognition units onto simple
peptide backbones. The resulting oligomers specifically
self-pair and cross-pair with complementary strands
of RNA and DNA in Watson-Crick fashion. Moreover,
the oligomers undergo dynamic component exchange,
template-directed assembly processes, and dynamic
sequence modification in response to changing selective pressures. Such oligomers could therefore have
participated in a number of processes that would be
advantageous for primordial genetic systems, such as
dynamic sequence repair and adaptation.
PUBLICATIONS
Cockroft, S.L., Chu, J., Amorin, M., Ghadiri, M.R. A single-molecule nanopore
device detects DNA polymerase activity with single-nucleotide resolution. J. Am.
Chem. Soc. 130:818, 2008.
Frezza, B.M., Cockroft, S.L., Ghadiri, M.R. Modular multi-level circuits from
immobilized DNA-based logic gates. J. Am. Chem. Soc. 129:14875, 2007.
Gianneschi, N.C., Ghadiri, M.R. Design of molecular logic devices based on a programmable DNA-regulated semisynthetic enzyme. Angew. Chem. Int. Ed.
46:3955, 2007.
Leman, L.J., Weinberger, D.A., Huang, Z.-Z., Wilcoxen, K.M., Ghadiri, M.R.
Functional and mechanistic analyses of biomimetic aminoacyl transfer reactions in
de novo designed coiled coil peptides via rational active site engineering. J. Am.
Chem. Soc. 129:2959, 2007.
S. Mahajan, A. Mayorov, G. McElhaney, J. Mee, A. Moreno,
Y. Nakai, A. Nguyen, A. Nunes, J. Park, A. Rohrbach,
C. Saccavini, N. Salzameda, S. Steiniger, J. Treweek,
A. Willis, Y. Xu, Y. Yoneda, B. Zhou, H. Zhou
uring the past year, we used various applications
of organic chemistry to address biological problems. Representative examples of our results
are given for 3 research programs: inhibition of bacterial virulence via the disruption of bacterial communication, discovery of a link between drug abuse and
cardiovascular disease, and selection and characterization of human neutralizing antibodies against Bacillus
anthracis toxin.
D
I N F E C T I O N C O N T R O L B Y A N T I B O D Y - M E D I AT E D
I N T E R F E R E N C E W I T H B A C T E R I A L C O M M U N I C AT I O N
The ability of microorganisms to coordinate their
gene expression according to population density has
been termed quorum sensing. This chemical exchange
of information among single-cell organisms is mediated
by secreted signaling molecules termed autoinducers.
Important biological and clinical aspects of quorum
sensing include the regulation of bacterial virulence
factors and the formation of biofilms; hence, inhibition of signaling associated with quorum sensing could
provide a promising new strategy for the attenuation
of bacterial infections. Indeed, analogs of autoinducers
have been used as small-molecule antagonists in several quorum-sensing circuits as a means of signaling
interference. Alternatively, we have pioneered an antibody-based strategy to combat quorum sensing through
disruption of signal transmission.
Recently, we applied our antibody-based technology
to the interference of the quorum-sensing circuits of
Staphylococcus aureus. This microorganism is the most
common cause of hospital-acquired infections, including
diseases ranging from skin infections and food poisoning
to life-threatening nosocomial infections. The increasing
resistance of S aureus isolates to glycopeptide antibiotics, most prominently vancomycin, is a major concern
in intensive care units, and an alternative strategy to
combat this pathogen is urgently required.
98 CHEMISTRY
2008
Staphylococcus aureus uses a set of 4 cyclic autoinducing peptides (AIP-1–AIP-4) to regulate its quorumsensing machinery, which is responsible for orchestrating
the expression of virulence genes. Thus, inhibition of the
S aureus system would result in decreased pathogenicity. We generated a monoclonal antibody, AP4-24H11,
to sequester AIP-4 (Fig. 1). This antibody was elicited
THE SCRIPPS RESEARCH INSTITUTE
In addition to causing severe dopaminergic neurotoxic effects, chronic methamphetamine self-administration induces increasing drug tolerance that correlates with
escalating intake. Although the molecular mechanism
behind pharmacologic tolerance is not fully elucidated,
we hypothesized that methamphetamine covalently
modifies endogenous proteins in a process known as
glycation (Fig. 2) before reaching the brain and medi-
F i g . 1 . Structure of the S aureus autoinducer AIP-4 and AP4
hapten used to generate the quorum-quenching antibody AP4-24H11.
against a rationally designed hapten (AP4, Fig. 1) and
efficiently interfered with the quorum sensing of S aureus
in vitro, as determined by real-time polymerase chain
reaction analysis and inactivation of AP4-24H11 by synthetic AIP-4. Importantly, AP4-24H11 suppressed both
S aureus–induced dermal injury in a mouse model of
abscess formation in vivo and provided complete protection against a lethal S aureus challenge. These
findings provide a strong foundation for further investigations of immunopharmacotherapy as treatment of
bacterial infections in which quorum sensing controls
the expression of virulence factors.
A L I N K B E T W E E N C H R O N I C M E T H A M P H E TA M I N E
U S E A N D C A R D I O VA S C U L A R D I S E A S E
The rapid spread of methamphetamine abuse across
the United States is as alarming as the propensity of
the drug to induce severe addiction and the healthrelated consequences of addiction. Whereas before
2001 methamphetamine use occurred predominantly
in the western United States, its use now is extending
rapidly throughout the United States and across different ethnic groups. The threat that methamphetamine
now poses to society underscores the need to more
thoroughly examine the ramifications of chronic methamphetamine abuse.
F i g . 2 . Reaction scheme of methamphetamine protein glycation
as initiated by glucose and methamphetamine.
ating its well-characterized stimulant effects. Glycation
reactions, collectively termed the Maillard reaction,
have been studied for decades in the food industry in
the development of flavor and color; however, Maillard
products can also assume a biologically hazardous role
when synthesized in vivo. Acquiring the ability to crosslink proteins, these irreversible reaction products, termed
advanced glycation end products (AGEs), have gained
notoriety for their participation in a range of pathologic changes.
Protein glycation by methamphetamine induces an
immune response against these modified proteins, which
could lead to sequestration of drug and, ultimately, the
development of tolerance. We have shown that this drugdependent glycation mechanism is operative in vivo. We
detected antibodies against methamphetamine-derived
AGEs in rats that chronically self-administered the drug,
and we noted a direct relationship between the level of
methamphetamine intake and the respective antibody
titers against methamphetamine-glycated proteins.
Additionally, we detected increased levels of proinflammatory and other cytokine molecules, particularly
vascular endothelial growth factor. AGE-associated upreg-
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
99
ulation of this growth factor has been associated with
the onset of heart disease, but these effects had not
been previously associated with methamphetaminederived AGEs. Because AGEs can alter protein function
in vivo and participate in various diseases, methamphetamine-derived AGEs provide an unrecognized
molecular mechanism for the development of vasculitis and other cardiovascular maladies with high incidence in chronic methamphetamine users.
HUMAN NEUTRALIZING ANTIBODIES AGAINST
ANTHRAX TOXIN
A less-than-adequate therapeutic plan for the treatment of anthrax in the 2001 bioterrorism attacks has
highlighted the importance of developing alternative or
complementary therapeutic approaches for biothreat
agents. Vaccination against B anthracis for protection
against anthrax has been known for more than a century. However, the prolonged vaccination schedules and
induction times required for an immune response are
serious drawbacks, because the therapeutic window for
treatment of anyone exposed to a deliberate release of
B anthracis is limited. Alternatively, recently developed
antibiotic prophylaxis for the treatment of persons
exposed to anthrax, although important, would also be
of lesser value if the infection were caused by an antibiotic-resistant strain.
Passive immunization has provided an attractive
avenue as a treatment both before and after exposure
to B anthracis. Indeed, in many animal studies, passive
transfer of antiserum successfully provided protection
against anthrax. Furthermore, passive immunization
could have advantages over active vaccination and
antibiotic treatments via few toxic effects, high specificity, the capability for stockpiling large quantities of
the antiserum, and immediate protection against a
biological attack.
Using a phage-displayed human single-chain variable fragment (scFv) antibody library, we selected and
characterized several human monoclonal neutralizing
antibodies against the toxin of B anthracis. In total,
15 clones with distinct sequences and high specificity
for the protective antigen region of the anthrax toxin
(Fig. 3) were analyzed by using biophysical and cellbased cytotoxicity assays. From this panel of antibodies, a set of neutralizing antibodies was identified, and
the potency of protection was established by using a
macrophage cytotoxicity assay. Among the neutralizing
antibodies identified, 1 clone had excellent affinity for
the protective antigen region of the anthrax toxin and
F i g . 3 . Targeting of the protective antigen (PA) region of B
anthracis toxin by human monoclonal antibodies.
provided superior protection from lethal toxin in the
cell cytotoxicity assay. Our results add to the ever-growing arsenal of immunologic and functional analysis of
monoclonal antibodies to the exotoxins of anthrax. In
addition, the antibodies may be new candidates for
prophylactic and therapeutic agents.
PUBLICATIONS
Brogan, A.P., Dickerson, T.J., Janda, K.D. Nornicotine-organocatalyzed aqueous
reduction of α,β-unsaturated aldehydes. Chem. Commun. (Camb.) Issue 46:4952,
2007.
Capková, K., Yoneda, Y., Dickerson, T.J., Janda, K.D. Synthesis and structureactivity relationships of second-generation hydroxamate botulinum neurotoxin A
protease inhibitors. Bioorg. Med. Chem. Lett. 17:6463, 2007.
Debler, E.W., Kaufmann, G.F., Meijler, M.M., Heine, A., Mee, J.M., Pljevaljcic,
G., Di Bilio, A.J., Schultz, P.G., Millar, D.P., Janda, K.D., Wilson, I.A., Gray, H.B.,
Lerner, R.A. Deeply inverted electron-hole recombination in a luminescent antibody-stilbene complex. Science 319:1232, 2008.
Dickerson, T.J., McKenzie, K.M., Hoyt, A.S., Wood, M.R., Janda, K.D., Brenner,
S., Lerner, R.A. Phage escape libraries for checkmate analysis. Proc. Natl. Acad.
Sci. U. S. A. 104:12703, 2007.
Ino, A., Dickerson, T.J., Janda, K.D. Positional linker effects in haptens for cocaine
immunopharmacotherapy. Bioorg. Med. Chem. Lett. 17:4280, 2007.
Kaufmann, G.F., Park, J., Janda, K.D. Bacterial quorum sensing: a new target for
anti-infective immunotherapy. Expert Opin. Biol. Ther. 8:719, 2008.
Kaufmann, G.F., Park, J., Mee, J.M., Ulevitch, R.J., Janda, K.D. The quorum
quenching antibody RS2-1G9 protects macrophages from the cytotoxic effects of
the Pseudomonas aeruginosa quorum sensing signalling molecule N-3-oxo-dodecanoyl-homoserine lactone. Mol. Immunol. 45:2710, 2008.
Lowery, C.A., Dickerson, T.J., Janda, K.D. Interspecies and interkingdom communication mediated by bacterial quorum sensing. Chem. Soc. Rev. 37:1337, 2008.
Park, J., Jagasia, R., Kaufmann, G.F., Mathison, J.C., Ruiz, D.I., Moss, J.A., Meijler, M.M., Ulevitch, R.J., Janda, K.D. Infection control by antibody disruption of
bacterial quorum sensing signaling. Chem. Biol. 14:1119, 2007.
Park, J., Kaufmann, G.F., Bowen, J.P., Arbiser, J.L., Janda, K.D. Solenopsin A, a
venom alkaloid from the fire ant Solenopsis invicta, inhibits quorum-sensing signaling in Pseudomonas aeruginosa. J. Infect. Dis. 198:1198, 2008.
100 CHEMISTRY
2008
Richardson, H.N., Zhao, Y., Fekete, E.M., Funk, C.K., Wirsching, P., Janda, K.D.,
Zorrilla, E.P., Koob, G.F. MPZP: a novel small molecule corticotropin-releasing factor
type 1 receptor (CRF1) antagonist. Pharmacol. Biochem. Behav. 88:497, 2008.
Treweek, J., Wee, S., Koob, G.F., Dickerson, T.J., Janda, K.D. Self-vaccination by
methamphetamine glycation products chemically links chronic drug abuse and cardiovascular disease. Proc. Natl. Acad. Sci. U. S. A. 104:11580, 2007.
Willis, B., Eubanks, L.M., Dickerson, T.J., Janda, K.D. The strange case of the
botulinum neurotoxin: using chemistry and biology to modulate the most deadly
poison. Angew. Chem. Int. Ed. 47:8360, 2008.
Willis, B., Eubanks, L.M., Wood, M.R., Janda, K.D., Dickerson, T.J., Lerner, R.A.
Biologically templated organic polymers with nanoscale order. Proc. Natl. Acad.
Sci. U. S. A. 105:1416, 2008.
Xu, Y., Hixon, M.S., Dawson, P.E., Janda, K.D. Development of a FRET assay for
monitoring of HIV gp41 core disruption. J. Org. Chem. 72:6700, 2007.
Yoneda, Y., Steiniger, S.C., Capkova, K., Mee, J.M., Liu, Y., Kaufmann, G.F.,
Janda, K.D. A cell-penetrating peptidic GRP78 ligand for tumor cell-specific prodrug therapy. Bioorg. Med. Chem. Lett. 18:1632, 2008.
Zarebski, L.M., Vaughan, K., Sidney, J., Peters, B., Grey, H., Janda, K.D.,
Casadevall, A., Sette, A. Analysis of epitope information related to Bacillus
anthracis and Clostridium botulinum. Expert Rev. Vaccines 7:55, 2008.
Zhou, B., Carney, C., Janda, K.D. Selection and characterization of human antibodies neutralizing Bacillus anthracis toxin. Bioorg. Med. Chem. 16:1903, 2008.
Zhou, B., Pellett, S., Tepp, W.H., Zhou, H., Johnson, E.A., Janda, K.D. Delineating the susceptibility of botulinum neurotoxins to denaturation through thermal
effects. FEBS Lett. 582:1526, 2008.
Zhou, H., Zhou, B., Ma, H., Carney, C., Janda, K.D. Selection and characterization of human monoclonal antibodies against Abrin by phage display. Bioorg. Med.
Chem. Lett. 17:5690, 2007.
Maintaining the Proteome to
Ameliorate Human Disease
J.W. Kelly, S. Choi, E. Culyba, M.T.A. Dendle, D. Du,
C. Fearns, A. Fuller, T.-W. Mu, A. Murray, D. Ong, J. Paulsson,
E.T. Powers, P. Rao, M. Saure, R. Simkovsky, S. Siegel,
J. Solomon, K. Usui, Y. Wang, I. Yonemoto, Z. Yu
aintenance of the proteome (proteostasis) both
inside and outside human cells is essential
for development, reproduction, and successful aging. Deficiencies in proteostasis lead to many
metabolic, oncologic, neurodegenerative, and cardiovascular diseases. Understanding the mechanisms of
proteostasis, especially defects in the pathways of the
proteostasis network that occur with aging, enables the
design of new therapeutic strategies to ameliorate ageonset protein misfolding diseases, a main goal of our
research. We use animal and cell-based disease models
and biophysical approaches in combination with medicinal chemistry and structure-based drug design. Our
collaborators, W.E. Balch, Department of Cell Biology;
J. Buxbaum, Department of Molecular and Experimental
M
THE SCRIPPS RESEARCH INSTITUTE
Medicine; J.R. Yates, Department of Chemical Physiology; E. Masliah, University of California, San Diego; and
A. Dillin, the Salk Institute for Biological Studies, La
Jolla, California, play an essential role in our multidisciplinary approach.
A M E L I O R AT I O N O F LY S O S O M A L S T O R A G E D I S E A S E S
Lysosomal storage diseases are loss-of-function
diseases often caused by a mutation in one of the
lysosomal enzymes, which results in excessive misfolding of the enzyme within the endoplasmic reticulum and cytosolic degradation instead of proper folding
and trafficking of the enzyme to the lysosome. In 2
separate studies, we found that the innate proteostasis capacity of a cell can be enhanced with small molecules termed proteostasis regulators to fold mutated
enzymes that would otherwise misfold and be degraded,
resulting in increased trafficking of the mutated enzyme
to the lysosome and increased function. In the first study,
we found that diltiazem and verapamil, L-type calcium
channel blocker drugs approved by the Food and Drug
Administration, increased folding capacity in the endoplasmic reticulum, trafficking, and activity of mutant
lysosomal enzymes associated with 3 distinct lysosomal
storage diseases: Gaucher disease, α-mannosidosis,
and type IIIA mucopolysaccharidosis. These compounds
likely act by calcium ion–mediated enhancement of
endoplasmic reticulum lumenal chaperone function. In
the second study, we discovered that 2 proteostasis
regulators partially restored folding, trafficking, and function of mutant enzymes in Gaucher and Tay-Sachs cell
lines by activating the unfolded protein response, a
signaling pathway that influences proteostasis in the
secretory pathway. Moreover, we found that the combination of a proteostasis regulator and a pharmacologic chaperone, a chemical that binds directly to a
given enzyme and thereby stabilizes the enzyme, synergistically restored enzyme function, because of their
distinct mechanisms of action.
U N D E R S TA N D I N G T H E E T I O L O G Y O F A L Z H E I M E R ’ S
DISEASE
We are interested in understanding the molecular
and mechanistic basis for the age-onset nature of Alzheimer’s disease. Genetic and biochemical evidence
implicates aggregation of amyloid β-peptide (Aβ), enabled
by an age-onset decrease in proteostatic capacity, as
the cause of neurodegeneration in this disease; however, precise identification of the toxic structure and
the mechanism of neurotoxic effects remain elusive. In
patients with Alzheimer’s disease, the correlation between
CHEMISTRY
2008
disease severity and the concentration of spherical
aggregates, annular structures, protofibrils and other
soluble oligomeric species is better than the correlation between disease severity and the concentration
of fibrillar amyloid.
Previously, we showed that mutating the phenylalanine 19–phenylalanine 20 backbone amide bond to
an E-olefin bond allows the formation of spherical aggregates to the exclusion of fibrils. In a more extensive
amide-to-ester mutagenesis scan through the hydrophobic core (residues 17–21) of Aβ 1-40, we compared the
mutants with wild-type Aβ 1-40 and the E-olefin Aβ
1-40 mutant. Even though the E-olefin mutant, the
amide-to-ester mutant, and wild-type Aβ 1-40 form
aggregates of different morphologies, all 3 types of aggregates were similarly toxic to PC12 neuronal cells. This
finding suggests that a common, but low-abundance,
aggregate morphology mediates toxic effects or that several different aggregate morphologies are similarly toxic.
O X I D I Z E D M E TA B O L I T E E N H A N C E M E N T O F
A M Y L O I D F O R M AT I O N
One of the central mysteries of Alzheimer’s disease
is how Aβ forms amyloid in vivo when both thermodynamic and kinetic barriers against aggregation exist.
We propose that covalent modification of Aβ by smallmolecule oxidation products can explain, at least in part,
the ability of Aβ to form amyloid at physiologic concentrations and thus place a load on the proteostasis network. Using Aβ conjugates site-specifically modified
with a cholesterol aldehyde at aspartic acid 1, lysine
16, or lysine 28, we found that modification lowered
the critical concentration for aggregation into the nanomolar range, within the physiologic concentration range
of Aβ, and dramatically increased the rate of aggregation. Aβ modified at lysine 16 formed amorphous aggregates fastest and at the lowest concentrations (within
2 hours at 20 nM).
The same cholesterol aldehyde is found in human
atherosclerotic lesions and rapidly promotes apolipoprotein C -II amyloid formation in vitro. Thus, enhancement of amyloid formation by oxidized metabolites
appears to be common to several diseases and suggests
that strategies to prevent such modification may have
therapeutic potential for a spectrum of human diseases.
T R E AT I N G T R A N S T H Y R E T I N A M Y L O I D O G E N E S I S
Transthyretin is 1 of 27 secreted human proteins,
including amyloid, known to misfold and misassemble
into extracellular aggregates. The rate-limiting step in
amyloid formation by transthyretin is the dissociation
THE SCRIPPS RESEARCH INSTITUTE
101
of the tetramer. We have developed kinetic stabilizers of
the tetrameric structure of transthyretin that have novel
chemistries and mechanisms of actions. To improve
inhibitors of transthyretin amyloidogenesis, we are optimizing each of the 3 substructural elements that make
up a typical inhibitor: the 2 aryl rings and the linker
that joins the rings. We evaluated structural modifications to the aryl ring 1 by screening a library of 2-arylbenzoxazoles that have thyroid hormone–like aryl
substituents on the 2-aryl ring. The 3,4,5-substituted
thyroxine-like aryl ring appears to be the optimal solution for the structure of aryl ring 1. In addition, we
synthesized 40 bisaryl compounds to optimize the structure of the linker. We found that direct connection of
the 2 aryls, or linkage through nonpolar E-olefin or
–CH 2 CH 2 – substructures, generates the most potent
and selective inhibitors of transthyretin amyloidogenesis.
Five high-resolution (1.4–1.8 Å) x-ray crystallography structures of transthyretin reveal that the 3,5dimethyl-4-hydroxyphenyl ring preferentially occupies
the inner cavity of the thyroxine-binding site and that
the 3,5-dibromo-4-hydroxyphenyl aryl prefers the outer
cavity because the phenol is deprotonated with flanking electron withdrawing group substitution. A study
to optimize the remaining aryl ring is well under way.
Tetramers of transthyretin can also be kinetically
stabilized by trans-suppression, as we showed previously with T119M transthyretin subunit incorporation
into the tetramer, which stabilizes heterotetramers
containing T119M and V30M transthyretin subunits.
In an analogous manner, heterotetramers composed
of murine transthyretin and human transthyretin subunits are kinetically stable and nonamyloidogenic.
This information is important for evaluating transgenic models of human transthyretin amyloidosis in
which the transgenic animals have a low copy number
of the mutant amyloidogenic human transthyretin gene.
U N D E R S TA N D I N G A N D A M E L I O R AT I N G G E L S O L I N
AMYLOIDOSIS
Gelsolin amyloid disease is another age-onset degenerative malady linked to protein aggregation that is
thought to be due to an age-associated decline in
proteostasis. A mutation in gelsolin (D187N) leads to
aberrant folding and cleavage by furin within the Golgi
apparatus during trafficking. Subsequent cleavage of the
gelsolin fragment by the matrix metalloprotease MT1MMP outside the cell results in 5- and 8-kD fragments
of gelsolin, which deposit as amyloid in the extracellular matrix. In collaboration with W.E. Balch, Depart-
102 CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
ment of Cell Biology, we have developed 2 transgenic
mouse models of human D187N gelsolin amyloidosis
that recapitulate the aberrant endoproteolytic cascade
and the aging-associated decline in proteostasis that
result in extracellular amyloidogenesis in humans.
Total Synthesis, New Synthetic
Technologies, and Chemical
Biology
PUBLICATIONS
Balch, W.E., Morimoto, R.I., Dillin, A., Kelly, J.W. Adapting proteostasis for disease intervention. Science 319:916, 2008.
K.C. Nicolaou, A. Agua, R. Aversa, W. Brenzovich,
A. Burtoloso, J. Chen, K. Cole, S. Dalby, R. Denton,
D. Edmonds, S. Ellery, A. Estrada, B. Fraga, M. Frederick,
M. Freestone, C. Gelin, J. Goodwin-Tindall, M. Hesse,
P. Huang, V. Jeso, M. Kar, A. Krasovskiy, A. Lemire, A. Li,
H. Li, Y. Lim, T. Lister, N. Mainolfi, U. Majumder, C. Mathison,
A. Morgan, A. Nold, A. Ortiz, N. Patil, B. Pratt, R. Reingruber,
F. Rivas, A. Sanchez Ruiz, D. Sarlah, D. Shaw, A. Stepan,
A. Talbot, Y. Tang, V. Trepanier, G. Tria, T. Umezawa,
J. Wang, T. Wu, W. Zhan, H. Zhang
Bieschke, J., Siegel, S.J., Fu, J., Kelly, J.W. Alzheimer’s Aβ peptides containing
an isostructural backbone mutation afford distinct aggregate morphologies but analogous cytotoxicity: evidence for a common low-abundance toxic structure(s)? Biochemistry 47:50, 2008.
Dillin, A., Kelly, J.W. The yin-yang of sirtuins. Science 317:461, 2007.
Fowler, D.M., Koulov, A.V., Balch, W.E., Kelly, J.W. Functional amyloid: from bacteria to humans. Trends Biochem. Sci. 32:217, 2007.
Jäger, M., Dendle, M., Kelly, J.W. A cross-strand Trp-Trp pair stabilizes a WW
domain at the expense of function. Protein Sci. 16:2306 2007.
Jäger, M., Nguyen, H., Dendle, M., Gruebele, M., Kelly, J.W. Influence of hPin1
WW N-terminal domain boundaries on function, protein stability, and folding. Protein Sci. 16:1495, 2007.
Johnson, S.M., Connelly, S., Wilson, I.A., Kelly, J.W. Biochemical and structural
evaluation of highly selective 2-arylbenzoxazole-based transthyretin amyloidogenesis inhibitors. J. Med. Chem. 51:260, 2008.
Kelly, J.W. Compromised cellular folding fidelity results in numerous clinically
important diseases. Nature 446:112, 2007.
Liu, F., Du, D., Fuller, A.A., Davoren, J.E., Wipf, P., Kelly, J.W., Gruebele, M. An
experimental survey of the transition between two-state and downhill protein folding scenarios. Proc. Natl. Acad. Sci. U. S. A. 105:2369, 2008.
Mu, T.-W., Fowler, D.M., Kelly, J.W. Partial restoration of mutant enzyme homeostasis in three distinct lysosomal storage disease cell lines by altering calcium
homeostasis. PloS Biol. 6:e26, 2008.
Münch, J., Rücker, E., Ständker, L., Adermann, K., Goffinet, C., Schindler, M.,
Wildum, S., Chinnadurai, R., Rajan, D., Specht, A., Giménez-Gallego, G.,
Sánchez, P.C., Fowler, D.M., Koulov, A., Kelly, J.W., Mothes, W., Grivel, J.C.,
Margolis, L., Keppler, O.T., Forssmann, W.G., Kirchhoff, F. Semen-derived amyloidogenic prostatic acidic phosphatase fragments dramatically enhance HIV infection. Cell 131:1059, 2007.
Reixach, N., Foss, T.R., Santelli, E., Pascual, J., Kelly, J.W. Human-murine transthyretin heterotetramers are kinetically stable and non-amyloidogenic: a lesson in
the generation of transgenic models of diseases involving oligomeric proteins. J. Biol.
Chem. 283:2098, 2008.
Stewart, C.R., Wilson, L.M., Zhang, Q., Pham, C.L.L., Waddington, L.J., Staples,
M.K., Stapleton, D., Kelly, J.W., Howlett, G.J. Oxidized cholesterol metabolites
found in human atherosclerotic lesions promote apolipoprotein C-II amyloid fibril
formation. Biochemistry 46:5552, 2007.
Wiseman, R.L., Koulov, A., Powers, E.T., Kelly, J.W., Balch, W.E. Protein energetics
in maturation of the early secretory pathway. Curr. Opin. Cell Biol. 19:359, 2007.
Wiseman, R.L., Powers, E.T., Buxbaum, J.N., Kelly, J.W., Balch, W.E. An adaptable
standard for protein export from the endoplasmic reticulum. Cell 131:809, 2007.
Yu, Z., Sawkar, A.R., Kelly, J.W. Pharmacologic chaperoning as a strategy to treat
Gaucher disease. FEBS Lett. 274:4944, 2007.
e focus on the total synthesis of natural products, the discovery and development of new
synthetic technologies, and chemical biology.
Naturally occurring substances are selected as synthetic
targets for their novel molecular architectures, important
biological properties, and interesting mechanisms of
action. The projects are designed to optimize the opportunities for discovery and invention in the areas of
chemistry, biology, and medicine. The natural products
thiostrepton, azaspiracid-1–azaspiracid-3, abyssomycin C,
the bisanthraquinones and the marinomycins exemplify
this philosophy. Current projects include studies on the
antibiotics nocathiacin I, platensimycin, and platencin;
the antitumor agents lomaiviticins A and B and uncialamycin; the anti-HIV agent biyouyanagin A; and the
marine biotoxin maitotoxin (Fig. 1).
In addition, we are developing synthetic technologies and strategies for chemical synthesis and chemical
biology studies. Our overall aims are to advance the
art and science of chemical synthesis and to develop
enabling technologies for biology and medicine while
maximizing educational opportunities and training of
young men and women in chemistry.
W
PUBLICATIONS
Nicolaou, K.C., Chen, J.S., Zhang, H., Montero, A. Asymmetric synthesis and biological properties of uncialamycin and 26-epi-unicialamycin. Angew. Chem. Int. Ed.
47:185, 2008.
Nicolaou, K.C., Cole, K.P., Frederick, M.O., Aversa, R.J., Denton, R.M. Chemical
synthesis of the GHIJK ring system and further experimental support for the originally assigned structure of maitotoxin. Angew. Chem. Int. Ed. 46:8875, 2007.
Nicolaou, K.C., Dethe, D.H., Chen, D.Y.-K. Total syntheses of amythiamicins A, B
and C. Chem. Commun. (Camb.) Issue 23:2632, 2008.
Nicolaou, K.C., Dethe, D.H., Leung, G.Y.C., Zou, B., Chen, D.Y.-K. Total synthesis
of thiopeptide antibiotics GE2270A, GE2270T, and GE2270C1. Chem. Asian J.
3:413, 2008.
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
103
Nicolaou, K.C., Wang, J., Tang, Y. Synthesis of the sporolide ring framework
through a cascade sequence involving an intramolecular [4+2] cycloaddition reaction of an o-quinone. Angew. Chem. Int. Ed. 47:1432, 2008.
Vale, C., Gómez-Limia, B., Nicolaou, K.C., Frederick, M.O., Vieytes, M.R.,
Botana, L.M. The c-Jun-N-terminal kinase is involved in the neurotoxic effect of
azaspiracid-1. Cell. Physiol. Biochem. 20:957, 2007.
Vilariño, N., Nicolaou, K.C., Frederick, M.O., Vieytes, M.R., Botana, L.M. Irreversible cytoskeletal disarrangement is independent of caspase activation during in
vitro azaspiracid toxicity in human neuroblastoma cells. Biochem. Pharmacol.
74:327, 2007.
Translational Chemistry
and Medicine
E. Roberts, G. Cherukupalli, C. Chiruta, O. Ghoneim,
M. Guerrero, S. Park, X. Peng, F. Pinacho-Crisostomo,
R. Poddutoori, K. Reynolds, M. Toussaint, M. Urbano,
S. Velaparthi, Y. Wang
F i g . 1 . Selected target molecules.
Nicolaou, K.C., Frederick, M.O., Burtoloso, A.C.B., Denton, R.M., Rivas, F., Cole,
K.P., Aversa, R.J., Gibe, R., Umezawa, T., Suzuki, T. Chemical synthesis of the
GHIJKLMNO ring system of maitotoxin. J. Am. Chem. Soc. 130:7466, 2008.
Nicolaou, K.C., Guduru, R., Sun, Y.-P., Banerji, B., Chen, D.Y.-K. Total synthesis
of the originally proposed and revised structures of palmerolide A. Angew. Chem.
Int. Ed. 46:5896, 2007.
Nicolaou, K.C., Krasovskiy, A., Trépanier, V.É., Chen, D.Y.-K. An expedient strategy for the synthesis of tryptamines and other heterocycles. Angew. Chem. Int. Ed.
47:4217, 2008.
Nicolaou, K.C., Li, H., Nold, A.L., Pappo, D., Lenzen, A. Total synthesis of
kinamycins C, F, and J. J. Am. Chem. Soc. 129:10356, 2007.
Nicolaou, K.C., Lister, T., Denton, R.M., Gelin, C.F. Cascade reactions involving
formal [2+2] thermal cycloadditions: total synthesis of artochamins F, H, I, and J.
Angew. Chem. Int. Ed. 46:7501, 2007.
Nicolaou, K.C., Lister, T., Denton, R.M., Gelin, C.F. Total synthesis of artochamins F,
H, I, and J through cascade reactions. Tetrahedron 64:4736, 2008.
Nicolaou, K.C., Majumder, U., Philippe Roche, S., Chen, D.Y.-K. Construction of
the “left-domain” of haplophytine. Angew. Chem. Int. Ed. 46:4715, 2007.
Nicolaou, K.C., Ortiz, A., Denton, R.M. Metathesis reactions in the synthesis of
complex molecules. Chem. Today 25:70, 2007.
Nicolaou, K.C., Pappo, D., Tsang, K.Y., Gibe, R., Chen, D.Y.-K. A chiral pool
based synthesis of platensimycin. Angew. Chem. Int. Ed. 47:944, 2008.
Nicolaou, K.C., Sun, Y.-P., Guduru, R., Banerji, B., Chen, D.Y.-K. Total synthesis
of the originally proposed and revised structures of palmerolide A and isomers
thereof. J. Am. Chem. Soc. 130:3633, 2008.
ntroduction of new medicines is crucial to preserve
human health. We are dedicated to the pursuit of
new and better therapies and continually challenge
the frontiers of drug discovery. As we take major scientific steps toward the future, we move to a more
knowledge-based drug discovery. Our goal is to generate high-quality clinical candidates as new medicines
in therapeutic areas such as epilepsy/seizures, neuropathic pain, autoimmune diseases (e.g., multiple sclerosis), and developmental disorders (e.g., autism).
I
T R E AT M E N T O F N E U R O L O G I C D I S E A S E S
Epilepsy is a disease in which a hyperexcited state
of the CNS is caused by an imbalance between inhibitory and excitatory neurotransmission. Current epilepsy
therapy focuses on the modulation of the classical
neurotransmitters glutamate and γ-aminobutyric acid.
The neuropeptide galanin antagonizes excitatory glutaminergic neurotransmission in the hippocampus, suggesting that galanin may have a role in seizure activity.
In collaboration with T. Bartfai and X. Lu, Molecular and Integrative Neurosciences Department, we have
identified new nonpeptidic ligands for the galanin receptors GalR1 and GalR2. This set of small, druglike molecules can displace the peptide galanin from its protein
binding site. Selectivity and potency of these initial molecular starting points are being optimized.
DUAL OPIOID AGONISTS–CHOLECYSTOKININ
Nicolaou, K.C., Tang, Y., Wang, J., Stepan, A.F., Li, A., Montero, A. Total synthesis and antibacterial properties of carbaplatensimycin. J. Am. Chem. Soc.
129:14850, 2007.
Nicolaou, K.C., Tria, G.S., Edmonds, D.J. Total synthesis of platencin. Angew.
Chem. Int. Ed. 47:1780, 2008.
A N TA G O N I S T S F O R T R E AT M E N T O F C H R O N I C A N D
N E U R O PAT H I C PA I N
Nociception, or the perception of pain, and its modulation depend on the interaction of many endogenous
104 CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
neurotransmitters in the spinal cord. The interaction
of endogenous peptides such as cholecystokinin with
exogenously administered opioids markedly alters activity in acute and chronic pain states. This interaction
may lead to the development of novel medications that
are more effective and safer than currently available
opioids alone.
Molecules with the property of being both opioid
agonists and cholecystokinin antagonists would be useful
in conditions in which the effectiveness of opioids is
reduced, as in the development of tolerance to opioid
pain relievers in chronic pain (e.g., in pain caused by
cancers) and in neuropathic pain conditions in which
opioids are ineffective. Thus, because of the prevention
(or reversal) of tolerance, additionally physical dependence on opioids might be diminished or inhibited. The
advantages of developing a single compound with dual
opioid agonist–cholecystokinin antagonist activity rather
than a combination of an opioid agonist taken with a
separate cholecystokinin antagonist are clear. Development of a single compound involves only a single set
of parameters, such as toxicology, pharmacokinetics,
and formulation, rather than 2 independent and often
unrelated sets of data.
In collaboration with F. Porreca and J. Lai, University of Arizona, Tucson, we are using a limited set of
molecular templates that have affinity across a wide
range of type 1 G protein–coupled receptors to develop
compounds with the required dual pharmacology. The
3 cloned opiate receptors (µ, δ, and κ) and the 2 cho-
I M M U N O M O D U L AT I N G C O M P O U N D S F O R T H E
lecystokinin receptors 1/(A) and 2/(B) are all members
of this subclass of G protein–coupled receptors (Fig. 1).
F i g . 1 . Opioid agonist–cholecystokinin antagonist hybrids.
T R E AT M E N T O F M U LT I P L E S C L E R O S I S
Sphingosine 1-phosphate (S1P) is an endogenous
mediator that functions both as an intracellular messenger and as an extracellular signaling molecule. S1P is
involved in a number of processes, including vascular
stabilization, cardiac development, and cancer angiogenesis. Extracellularly, S1P elicits its biological effects
through a family of G protein–coupled receptors that
bind to the S1P1–S1P5 subtypes of S1P. Activation of
the receptor for S1P1 had effects in multiple sclerosis
and organ transplantation via the immunosuppression
evoked by the nonselective S1P agonist FTY720, which
is currently in clinical trials. It was speculated that the
effects on heart rate and lung function in clinical trails
with FTY720 were due to agonism of S1P3.
In collaboration with H. Rosen and his group, Department of Immunology ane Microbial Science, we have
identified novel small-molecule compounds that are
exquisitely selective for S1P1, are stable, are orally
active, and penetrate into the CNS (Table 1). These
compounds are expected to be useful in disease states
such as multiple sclerosis.
T a b l e 1 . Agonists of sphingosine 1-phosphates.
Compound Total polar
Molecular Calculated
surface area, Å2 weight, kD logP
EC 50, nM
Sphingosine 1phosphate 1
Sphingosine 1phosphate 3
CYM5313
64.4
349
4.5
0.52
823
CYM5326
64.4
351
4.4
0.2
529
CYM5327
64.4
349
4.5
3.0
Not applicable
CYM5332
55.7
381
4.5
4.1
5200
CYM5357
64.4
351
4.4
3.1
1800
CYM5358
81.5
365
3.3
1.7
Not applicable
CYM5380
72.6
354
3.6
0.8
774
CYM5389
72.6
340
3.5
4.2
606
CYM5390
72.6
340
3.5
7
1500
CYM5391
72.6
354
3.7
0.5
716
CYM5399
72.6
354
3.7
1.1
691
CYM5410
85.0
341
2.0
0.14
938
CYM5418
72.6
366
4.6
0.1
Not applicable
CYM5422
64.8
361
4.5
1.24
Not applicable
CYM5440
75.9
395
4.2
0.5
Not applicable
CYM5442
84.7
409.5
3.7
1.1
Not applicable
CYM5449
67.7
464
5.7
0.7
Not applicable
CYM5457
84.7
397
3.4
3.4
692
CYM5464
75.9
409
4.6
0.84
Not applicable
CYM5452
54.2
416
5.6
1.5
Not applicable
CYM5386
46.3
387
5.2
4.5
1300
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
105
I L - 6 A N TA G O N I S T S F O R T R E AT M E N T O F
INCREASING THE CHEMICAL AND GENETIC
I N F L A M M AT O R Y D I S E A S E S
POTENTIAL OF DNA
Clinical studies have provided strong evidence that
specific blockade of IL-6–regulated signaling pathways
is a validated approach for treatment of inflammatory
diseases. Currently, only monoclonal antibodies are
available to block the actions of IL-6. Using the humanized antibody tocilizumab to block the actions of IL-6
has been therapeutically effective in patients with rheumatoid arthritis, systemic juvenile idiopathic arthritis,
and Crohn’s disease. Tocilizumab is in phase 1 clinical
trials in the United States and in phase 2 clinical trials
in France for the treatment of multiple myeloma.
However, serious adverse effects with tocilizumab
have been reported, including a death and allergic pneumonitis. Increases in the levels of serum lipids, liver
function abnormalities, and reduction in white blood
cell count occurred in substantial numbers of patients.
In collaboration with Dr. Porreca, we have identified
small, orally active IL-6 receptor antagonists that are
being modified for affinity and druglike properties. Once
optimized, these antagonists should have fewer adverse
side effects than does tocilizumab.
We are interested in increasing the information
potential of DNA by expanding the genetic alphabet
with a third base pair composed of unnatural nucleobases. Using hydrophobicity, polarity, shape complementarity, and hydrogen bonding, we have developed
novel unnatural base pairs, including several that are
replicable in vitro. More recently, we screened more than
3600 unnatural nucleotides and identified a base pair
that, after optimization, is replicated with an efficiency
close to that of natural DNA synthesis.
Nature developed the natural genetic code, not only
by optimizing DNA and RNA but also by evolving the
polymerases that synthesize these nucleic acids. We
developed an activity-based selection system (Fig. 1)
PUBLICATIONS
Lum, C., Kahl, J., Kessler, L., Kucharski, J., Lundström, J., Miller, S., Nakanishi,
H., Pei, Y., Pryor, K., Roberts, E., Sebo, L., Sullivan, R., Urban, J., Wang, Z. 2,5Diaminopyrimidines and 3,5-disubstituted azapurines as inhibitors of glycogen synthase kinase-3 (GSK-3). Bioorg. Med. Chem. Lett. 18:3578, 2008.
Montalban, A.G., Boman, E., Chang, C.D., Ceide, S.C., Dahl, R., Dalesandro, D.,
Delaet, N.G., Erb, E., Ernst, J.T., Gibbs, A., Kahl, J., Kessler, L., Lundström, J.,
Miller, S., Nakanishi, H., Roberts, E., Saiah, E., Sullivan, R., Wang, Z., Larson,
C.J. The design and synthesis of novel α-ketoamide-based p38 MAP kinase inhibitors. Bioorg. Med. Chem. Lett. 18:1772, 2008.
F i g . 1 . Activity-based phage display selection system for evolv-
ing polymerases with novel activity. Infection of phage (B) with the
polymerase library (A) leads to production of phage particles that
Chemical, Biological, and
Biophysical Approaches to
Understanding Evolution
F.E. Romesberg, D.A. Bachovchin, P. Capek, J.K. Chin,
R.T. Cirz, M.E. Cremeens, N. Gingles, Y. Hari, D.A. Harris,
A. Horhota, G.T. Hwang, A.M. Leconte, E.T. Lis, S. Matsuda,
B.A. O’Neill, M. Patel, M.E. Powers, T.C. Roberts, Y.J. Seo,
P.A. Smith, M.C. Thielges, P. Weinkam, W. Yu, J. Zimmermann
he molecules of biology are unique because they
have been evolved for function. We use multidisciplinary methods in conjunction with chemical biological principles to develop unique approaches
to understanding evolution.
T
display 0–1 copies of the polymerase and 3–5 copies of the acidic
peptide. Phage particles are combined with DNA primer–template
(C) and incubated with the desired nucleoside triphosphates. Active
mutants are isolated (D) and characterized.
to evolve polymerases for any desired function. Using
this system, we have already evolved polymerases with
a variety of novel functions, including the synthesis of
DNA containing one of the unnatural base pairs. We are
optimizing these polymerases and evolving new ones.
REENGINEERING ANCIENT ANTIBIOTICS
Because of the potential for cross-resistance, a great
need exists for new antibiotics, especially ones that act
via novel mechanisms. Although medicinal chemists
have successfully reengineered already validated antibiotic scaffolds that were compromised by resistance, the
identification of novel synthetic (nonnatural) scaffolds
106 CHEMISTRY
2008
has been extraordinarily challenging. Thus, natural products that might be candidates for antibiotics, perhaps
even products that do not appear to still be active
because of cross-resistance, warrant a careful examination. Using modern tools of synthesis and chemical biology, we might be able to determine why the products
lost activity and perhaps use this knowledge to reengineer them to again be potent, broad-spectrum antibiotics.
Arylomycins are a series of biphenyl-linked macrocyclic lipopeptide natural products that inhibit the essential bacterial signal peptidase I (SPase) in vitro but have
low potency and a narrow spectrum as antibiotics. After
some initial interest, these natural products were abandoned by the pharmaceutical industry because of their
insufficient potency, which presumably was due to the
resistance that developed during their use in bacterial
warfare over eons of time. We recently reported the first
total synthesis of a member of this class of natural products: arylomycin A2.
With large quantities of arylomycin A2 in hand, we
evaluated it against a wide variety of bacteria and discovered that it is extremely potent against the important
human pathogen Staphylococcus epidermidis. By using
UV light to create mutants of S epidermidis, we discovered that resistance evolved via the introduction of a
proline residue into a conserved region of the SPase substrate-binding site. Sequence analysis of other bacterial
SPases revealed that all bacteria with natural resistance
to arylomycin A2 already had the "resistance-conferring"
proline, and we found that when this proline is removed
genetically, important pathogens such as Escherichia
coli and Staphylococcus aureus become sensitive to
arylomycin A2. Sequence analysis also indicated additional pathogens predicted to be sensitive, including the
gram-positive pathogens Streptococcus pyogenes and
Streptococcus pneumoniae and the gram-negative pathogens Helicobacter pylori and Chlamydia trachomatis.
These data suggest that if the arylomycins can be
reengineered to bind SPase regardless of the resistanceconferring proline, they will again be potent, broadspectrum antibiotics. Currently, we are characterizing
the mechanism of arylomycin resistance to determine
how to reengineer these natural products for potency
and for the design, synthesis, and characterization of
potentially active derivatives.
EVOLUTION OF PROTEIN DYNAMICS
Molecular recognition underlies almost all of a protein’s biological functions. Nowhere is the evolution of
molecular recognition more impressive than within the
THE SCRIPPS RESEARCH INSTITUTE
immune response; antibodies are evolved within a matter of days to selectively recognize almost any foreign
molecule. Antibodies are also remarkable because different intermediates can be isolated during their evolution. We use ultrafast nonlinear optical and nuclear
magnetic resonance spectroscopy to characterize these
intermediates. We have generated a comprehensive view
over all timescales, from femtoseconds to seconds, of
how antibodies are evolved for molecular recognition.
Finally, the products of evolution are molecules with
unique vibrational dynamics. The study of vibrational
dynamics in proteins and nucleic acids has been limited by spectral complexity, but selective deuteration
of a protein or a nucleic acid results in a carbon-deuterium oscillator that absorbs light in an otherwise
transparent region of the infrared spectrum. The synthesis of selectively deuterated proteins has provided
us with a residue-specific probe of flexibility, function,
and folding. Previously, we focused on the biological
redox activities of cytochrome c. More recently, we have
focused on the protein recognition module SH3 and the
enzyme dihydrofolate reductase.
PUBLICATIONS
Cirz, R.T., Jones, M.B., Gingles, N.A., Minogue, T.D., Jarrahi, B., Peterson, S.N.,
Romesberg, F.E. The complete and SOS-mediated response of Staphylococcus
aureus to the antibiotic ciprofloxacin. J. Bacteriol. 189:531, 2007.
Heideker, J., Lis, E.T., Romesberg, F.E. Phosphatases, DNA damage checkpoints
and checkpoint deactivation. Cell Cycle 6:3058, 2007.
Hwang, G.T., Leconte, A.M., Romesberg, F.E. Polymerase recognition and stability
of fluoro-substituted pyridone nucleobase analogues. Chembiochem 8:1606,
2007.
Matsuda, S., Fillo, J.D., Henry, A.A., Rai, P., Wilkens, S.J., Dwyer, T.J., Geierstanger, B.H., Wemmer, D.E., Schultz, P.G., Spraggon, G., Romesberg, F.E.
Efforts toward expansion of the genetic alphabet: structure and replication of
unnatural base pairs. J. Am. Chem. Soc. 129:10466, 2007.
Roberts, T.C., Smith, P.A., Cirz, R.T., Romesberg, F.E. Structural and biological
analysis of synthetic arylomycin A2. J. Am. Chem. Soc. 129:15830, 2007.
Smith, P.A., Romesberg, F.E. Combating bacteria and drug resistance by inhibiting
mechanisms of persistence and adaptation. Nat. Chem. Biol. 3:549, 2007.
CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
107
Synthesis of Natural Products,
Development of Synthetic
Methods, and Medicinal
Chemistry
W.R. Roush, R. Bates, D. Bykowski, M. Chen, E. Darout,
A. DeBaillie, J. Dunetz, G. Halvorsen, M. Handa, J. Hicks,
T. Hopkins, C.-W. Huh, F. Li, A. Legg, R. Lira, L. Martinez,
C. Nguyen, G. Nora, R. Pragani, R. Rahaim, J. Roth,
H. Sun, M. Tortosa, J. Whitaker, S. Winbush
ur research has 2 major themes. One is the total
synthesis of structurally complex, biologically
active natural products such as those shown
in Figure 1. In each of these syntheses, we emphasize
the discovery, development, and/or illustration of new
reactions and methods for achieving high levels of stereochemical control. These efforts are pursued in parallel
with reaction design, stereochemical studies, and the
development of new synthetic methods. We are particularly interested in stereochemical aspects of intramolecular and transannular Diels-Alder reactions, development
of methods for the diastereoselective and enantioselective reactions of allylmetal compounds with carbonyl
compounds, and nucleophilic phosphine-catalyzed
organic reactions.
Recent research has included stereochemical studies
of transannular Diels-Alder reactions used in total syntheses of spinosyn A and superstolide A and development of new versions of the double allylboration reactions
of aldehydes with γ-boryl-substituted allylboranes for
stereocontrolled synthesis of 1,5-ene-diols, which are
being used in several ongoing syntheses, including those
of tetrafibricin, apoptolidin A, and peloruside. In addition,
we have synthesized highly substituted tetrahydrofurans
via [3+2]-annulation reactions of highly functionalized
allylsilanes; this chemistry was recently applied to total
syntheses of 10-hydroxytrilobacin and 3 stereoisomers.
We have also developed phosphine-mediated organocatalytic reactions, and we recently completed the total
synthesis of tedanolide.
Our second area of major interest involves problems
in bioorganic chemistry and medicinal chemistry. One
long-term project is the design and synthesis of inhibitors of cysteine proteases isolated from tropical parasites, such as Trypanosoma cruzi, the causative agent
of Chagas’ disease, and Plasmodium falciparum, the
O
F i g . 1 . Structures of recently synthesized natural products.
most virulent of the malaria parasites. This research is
performed in collaboration with colleagues at the University of California, San Francisco. In collaboration with
S. Reed, University of California, San Diego, we have
developed a cysteine protease inhibitor with remarkable
ability to prevent Entamoeba histolytica from invading
human intestinal tissue. Optimization of this inhibitor
108 CHEMISTRY
2008
for in vivo applications is in progress. New projects
involve discovery of small molecules that affect cancer
and other disease-related biochemical targets (e.g.,
nuclear hormone receptors), studies of structure-activity
relationships, and optimization of the pharmacologic
profile of certain natural products.
PUBLICATIONS
Chen, Y.-T., Lira, R., Hansell, E., McKerrow, J.H., Roush, W.R. Synthesis of
macrocyclic trypanosomal cysteine protease inhibitors. Bioorg. Med. Chem. Lett.
18:5860, 2008.
Dunetz, J., Roush, W.R. Concerning the synthesis of the tedanolide C(13)-C(23)
fragment via an anti-aldol reaction. Org. Lett. 10:2059, 2008.
Handa, M., Scheidt, K.A., Bossart, M., Zheng, N., Roush, W.R. Studies on the
synthesis of apoptolidin A, I: synthesis of the C(1)-C(11) fragment. J. Org. Chem.
73:1031, 2008.
Handa, M., Smith, W.J. III, Roush, W.R. Studies on the synthesis of apoptolidin
A, II: synthesis of the disaccharide unit. J. Org. Chem. 73:1036, 2008.
Hicks, J.C., Huh, C.W., Legg, A.D., Roush, W.R. Concerning the selective protection of (Z)-1,4-syn-ene-diols and (E)-1,5-anti-ene-diols as allylic triethylsilyl ethers.
Org. Lett. 9:5621, 2007.
Hicks, J.D., Roush, W.R. Synthesis of the C(26)-C(42) and C(43)-C(67) pyrancontaining fragments of amphidinol 3 via a common pyran intermediate. Org. Lett.
10:681, 2008.
Lira, R., Roush, W.R. Enantio- and diastereoselective synthesis of syn-β-hydroxyallylsilanes via a chiral (Z)-γ-silylallylboronate. Org. Lett. 9:4315, 2007.
Methot, J.L., Roush, W.R. Applications of tricoordinated phosphorus compounds in
organic catalysis. In: Organophosphorus Compounds. Trost, B.M. (Ed.). Thieme
Chemistry, New York, in press. Vol. 42 in Science of Synthesis.
Roth, J., Madoux, F., Hodder, P., Roush, W.R. Synthesis of small molecule inhibitors of the orphan nuclear receptor steroidogenic factor-1 (NR5A1) based on isoquinolinone scaffolds. Bioorg. Med. Chem. Lett. 18:2628, 2008.
Roush, W.R. Total synthesis of biologically active natural products. J. Am. Chem.
Soc. 130:6654, 2008.
Tortosa, M., Yakelis, N.A., Roush, W.R. Total synthesis of (+)-superstolide A. J.
Am. Chem. Soc. 130:2722, 2008.
Winbush, S.M., Mergott, D.J., Roush, W.R. Total synthesis of (–)-spinosyn A:
examination of structural features that govern the stereoselectivity of the key
transannular Diels-Alder reaction. J. Org. Chem. 73:1818, 2008.
Biological Chemistry
P.G. Schultz, E. Brustad, P. Chen, C. Dambacher, D. Groff,
J. Grünewald, J. Guo, B. Hutchins, S. Kazane, H. Lee,
J.-S. Lee, C. Liu, C. Lyssiotis, C. Melancon, J. Mills, R. Perera,
F. Peters, S. Schiller, M. Sever, L. Supekova, T. Young
lthough chemists are remarkably adept at the
synthesis of molecular structure, they are far
less sophisticated in designing and synthesizing
molecules with defined biological or chemical functions.
Nature, on the other hand, has produced an array of
molecules with remarkably complex functions, ranging
from photosynthesis and signal transduction to molec-
A
THE SCRIPPS RESEARCH INSTITUTE
ular recognition and catalysis. Our aim is to combine
the synthetic strategies and biological processes of
Nature with the tools and principles of chemistry to
create new molecules with novel chemical and biological functions. By studying the properties of the
resulting molecules, we can gain new insights into
the molecular mechanisms of complex biological and
chemical systems.
For example, we have shown that the tremendous
combinatorial diversity of the immune response can be
chemically reprogrammed to generate selective enzymelike catalysts. We have developed antibodies that catalyze a wide array of chemical and biological reactions,
from acyl transfer to redox reactions. Characterization
of the structure and mechanisms of these catalytic
antibodies has led to important new insights into the
mechanisms of biological catalysis. In addition, the
detailed characterization of the properties and structures of germ-line and affinity-matured antibodies has
revealed fundamental new aspects of the evolution of
binding and catalytic function, in particular, the role
of structural plasticity in the immune response. Most
recently, we have focused on in vitro evolution methods
that involve the development of novel chemical screens
and selections for identifying metalloantibodies with
proteolytic activity.
In addition, we are extending this combinatorial
approach to many other problems, including the generation of novel cellular reporters, the ab initio evolution of novel protein domains, and the synthesis of
structure-based combinatorial libraries of small heterocycles. The libraries of small heterocycles are being
used in conjunction with novel cellular and organismal
screens to identify molecules that modulate the activity of important proteins involved in such cellular processes as differentiation, proliferation, and signaling.
Indeed, we have identified molecules that control adult
and embryonic stem cell differentiation and stem cell
self-renewal and that reprogram lineage-committed cells
to alternative cell fates. We are using x-ray crystallographic and biochemical studies, together with mRNA
profiling technology and genetic complementation, to
characterize the mode of action of these compounds and
to study their effects on cellular processes and in animal models of regeneration. More recently, we extended
such studies to a variety of genetic and neglected diseases (e.g., malaria, type 1 diabetes, spinal muscular
atrophy, sickle cell anemia). We are also developing
and applying modern genomics tools (e.g., cell-based
phenotypic screens of arrayed genomic cDNA and short
CHEMISTRY
2008
interfering RNA libraries) and proteomics tools (mass
spectrometric phosphoprotein profiling) to a variety of
significant biomedical problems in cancer biology, neurodegenerative disease, and virology. In addition, we are
investigating the role and regulation of noncoding RNAs.
We have also developed a general biosynthetic
method that makes it possible to site specifically incorporate unnatural amino acids into proteins in vitro and
in vivo. Using this method, we effectively expanded the
genetic code of living organisms by adding new components to the existing biosynthetic machinery. We
have genetically encoded amino acids with novel spectroscopic and chemical properties (e.g., metal-binding,
sulfated, fluorescent, photocross-linking, and photoisomerizable) in response to unique 3- and 4-base codons.
These amino acids are being used to explore protein
structure and function both in vitro and in vivo, create
novel therapeutic agents and biomaterials, and evolve
proteins with novel properties. This approach has been
developed for Escherichia coli, yeast, and mammalian
cells, and we are now extending it to multicellular organisms. Our results have removed a billion-year constraint
imposed by the genetic code on the ability to chemically
manipulate the structures of proteins during translation.
PUBLICATIONS
Galkin, A.V., Melnick, J.S., Kim, S., Hood, T.L., Li, N., Li, L., Xia, G., Steensma,
R., Chopiuk, G., Jiang, J., Wan, Y., Ding, P., Liu, F., Sun, F., Schultz, P.G., Gray,
N.S., Warmuth, M. Identification of NVP-TAE684: a potent, selective. and efficacious inhibitor of NPM-ALK [published correction appears in Proc. Natl. Acad. Sci.
U. S. A. 104:2025, 2007]. Proc. Natl. Acad. Sci. U. S. A. 104:270, 2007.
Liu, Y., Kern, J.T., Walker, J. R. Johnson, J., Schultz, P.G., Luesch, H. A genomic
screen for activators of the antioxidant response element. Proc. Natl. Acad. Sci.
U. S. A. 104:5205, 2007.
Gumireddy, K., Sun, F., Klein-Szanto, A.,J., Gibbins, J.M., Saunders, A., Schultz,
P.G., Huang, Q. In vivo selection for metastasis promoting genes in the mouse.
Proc. Natl. Acad. Sci. U. S. A. 104:6696, 2007.
Liu, W., Alfonta, L., Mack, A.V., Schultz, P.G. Structural basis for the recognition
of para-benzoyl-L-phenylalanine by evolved aminoacyl-tRNA synthetases. Angew.
Chem. Int. Ed. 46:6073, 2007.
Xie, J., Supekova, L., Schultz, P.G. A genetically encoded metabolically stable analogue of phosphotyrosine in Escherichia coli. ACS Chem. Biol. 2:474, 2007.
THE SCRIPPS RESEARCH INSTITUTE
Click Chemistry and
Biological Activity
K.B. Sharpless, J. Culhane, J. Fotsing, S. Grecian,
N. Grimster, J. Hein, T. Horneff, J. Kalisiak, K. Korthals,
S.-W. Kwok, S. Pitram, J. Raushel, B. Stump, J. Tripp,
C. Valdez, T. Weide
he driving forces in our research are the discovery and understanding of chemical reactivity, the
harbingers of new discoveries in chemistry. Our
goal is to develop chemical transformations that enable
scientists to rapidly synthesize diverse compounds with
desired properties; after all, it is the function of molecules that matters. The nature of the building blocks
and the speed with which synthesis, screening for the
desired function, and lead optimization can be performed
are determining factors in the search for new compounds, whether the new entities are drugs, better plastics, or dyes. The greater the variety of scaffolds and
functional groups that can be used in the rapid construction of candidate compounds, the more likely it is that
new and useful function will be discovered. Because
of the enormous number of compounds to explore (the
number of small druglike molecules may be as high
as 1064), the size of a given collection becomes much
less important than the ability to rapidly probe the collection for a desired activity.
Several years ago, we proposed a minimalistic
approach to synthesis that relies solely on the best
reactions for assembly of new molecules. Inspired by
the natural synthesis of the myriad functional molecules (nucleic acids, proteins, and carbohydrates) from
just a handful of building blocks, we devised a fast,
reliable, and highly modular style of organic synthesis,
which we termed click chemistry. Click reactions fulfill
the most stringent criteria of usefulness and convenience
(Fig. 1); they are highly energetically driven, and the
T
Wang, J., Schiller, S., Schultz, P.G. A biosynthetic route to dehydroalanine-containing proteins. Angew. Chem. Int. Ed. 46:6849, 2007.
Xie, J., Liu, W., Schultz, P.G. A genetically encoded bidentate, metal-binding
amino acid. Angew. Chem. Int. Ed. 46:9239, 2007.
Supekova, L., Supek, F., Lee, J., Chen, S., Gray, N., Pezacki, J., Schlapbach, A.,
Schultz, P.G. Identification of human kinases involved in hepatitis C virus replication by small interference RNA library screening. J. Biol. Chem. 283:29, 2008.
Lemke, E.A., Summerer, D., Geierstanger, B.H., Brittain, S.M., Schultz, P.G. Control of protein phosphorylation with a genetically encoded photocaged amino acid.
Nat. Chem. Biol. 3:769, 2007.
Guo, J., Wang, J., Anderson, J.C., Schultz, P.G. Addition of an α-hydroxy acid to
the genetic code of bacteria. Angew. Chem. Int. Ed. 47:722, 2008.
109
F i g . 1 . Click chemistry: molecular diversity from a handful of
near-perfect reactions.
110 CHEMISTRY
majority of them form carbon-heteroatom bonds. The
reactions produce only the expected products and work
regardless of which functional groups are present in the
starting materials. Naturally, the number of reactions that
meet these criteria is limited, but we contend that a
wide variety of interesting and useful molecules can
be easily made by using click chemistry and that the
chances for achieving desirable biological activity with
such compounds are at least as good as chances with
the traditional target-guided approach.
Recently, we realized that olefins are probably the
most attractive starting molecules available to synthetic
organic chemists. Olefins are readily accessible in large
quantities and in many varieties, and processes for their
selective oxidation provide convenient access to electrophilic intermediates such as epoxides, aziridines, aziridinium ions, and cyclic sulfates. These electrophilic
intermediates are ideal for introduction of reactive “hot
spots,” such as azides and acetylenes, that can be
used for the assembly of final structures via 1,3-dipolar cycloadditions.
The 1,3-dipolar cycloaddition of azides and alkynes,
most extensively studied by R. Huisgen in the 1960s,
and the copper- and ruthenium-catalyzed variants we
developed with V.V. Fokin, Department of Chemistry,
take a prominent place in click reactions. These transformations enable reliable assembly of complex molecules by means of the 1,2,3-triazole heterocycle.
Although both alkynes and azides are highly energetic, they are quite unreactive to a broad range of
reagents, solvents, and other common functional groups.
This inertness allows clean sequential transformations
of broad scope without the need for protecting groups,
even if the reactions are performed in aqueous solvent
in the presence of atmospheric oxygen. The 1,2,3-triazoles have advantageous properties of high chemical
stability (in general, they are inert to severe hydrolytic,
oxidizing, and reducing conditions, even at high temperatures), strong dipole moment, presence of aromatic
groups, and the ability to accept hydrogen bonds. Thus,
they can interact productively in several ways with
biological molecules. For example, 1,2,3-triazoles can
replace the amide bond in peptides, preventing proteolytic degradation of the peptides.
Our focus on this powerful and underappreciated
class of azoles led us back to the simple parent triazole
(C2H 3N 3), which in solution is a rapidly equilibrating
mixture of 2 tautomers (Fig. 2). The physical properties
of the NH-triazole struck us as highly unusual and are, in
2008
THE SCRIPPS RESEARCH INSTITUTE
F i g . 2 . Michael additions of NH-triazole.
fact, much like those of water. These properties include
its weak acid-base character, high proton conductivity,
and a liquid range spanning nearly 200 degrees. In addition, the NH-1,2,3-triazole is stable: it is insensitive to
impact, friction, rapid heating, and even detonation.
We studied the Michael reaction of NH-triazole with
α,β-unsaturated ketones. The 1H-1,2,3-triazolyl-ketones
were selectively generated when the triazole was combined with a variety of enones under solvent-free conditions. The use of aprotic solvents with a catalytic
base gave the corresponding 2H-regioisomers. Together,
these 2 protocols provide direct access to either the N1or N2-substituted 1,3-triazolyl ketone regioisomers.
PUBLICATIONS
Finn, M.G., Kolb, H.C., Fokin, V.V., Sharpless, K.B. Concept and applications of
click chemistry from the standpoint of advocates. Kagaku to Kogyo 60:976, 2007.
Hawker, C.J., Fokin, V.V., Finn, M.G., Sharpless, K.B. Bringing efficiency to materials synthesis: the philosophy of click chemistry. Aust. J. Chem. 60:381, 2007.
Kalisiak, J., Sharpless, K.B., Fokin, V.V. Efficient synthesis of 2-substituted-1,2,3triazoles. Org. Lett. 10:3171, 2008.
Kwok, S.-W., Hein, J.E., Fokin, V.V., Sharpless, K.B. Regioselective synthesis of
either 1H- or 2H-1,2,3-triazoles via Michael addition to α,β-unsaturated ketones.
Heterocycles 76:1141, 2008.
Liu, Y., Díaz, D.D., Accurso, A.A., Sharpless, K.B., Fokin, V.V., Finn, M.G. Click
chemistry in materials synthesis, III: metal-adhesive polymers from Cu(I)-catalyzed
azide-alkyne cycloaddition. J. Polym. Sci. A Polym. Chem. 45:5182, 2007.
Radić, Z., Manetsch, R., Fournier, D., Sharpless, K.B., Taylor, P. Probing gorge
dimensions of cholinesterases by freeze-frame click chemistry. Chem. Biol. Interact.
175:161, 2008.
Sugawara, A., Sunazuka, T., Hirose, T., Nagai, K., Yamaguchi, Y., Hanaki, H.,
Sharpless, K.B., Omura, S. Design and synthesis via click chemistry of 8,9-anhydroerythromycin A 6,9-hemiketal analogues with anti-MRSA and -VRE activity.
Bioorg. Med. Chem. Lett. 17:6340, 2007.
Van der Eycken, E., Sharpless, K.B. Click chemistry. QSAR Comb. Sci. 26:1115,
2007.
CHEMISTRY
2008
Vestberg, R., Malkoch, M., Kade, M., Wu, P., Fokin, V.V., Sharpless, K.B., Drockenmuller, E., Hawker, C.J. Role of architecture and molecular weight in the formation of tailor-made ultrathin multilayers using dendritic macromolecules and click
chemistry. J. Polym. Sci. A Polym. Chem. 45:2835, 2007.
Yoo, E.J., Ahlquist, M., Bae, I., Sharpless, K.B., Fokin, V.V., Chang, S. Mechanistic
studies on the Cu-catalyzed three-component reactions of sulfonyl azides, 1-alkynes
and amines, alcohols, or water: dichotomy via a common pathway. J. Org. Chem.
73:5520, 2008.
Chemistry, Biology, and
Inflammatory Disease
P. Wentworth, Jr., D. Angrish, J. Dambacher, V. Dubrovskaya,
R.K. Grover, J. Nieva, M. Puga, B.D. Song, M.M.R. Peram,
J.K. Rogel, S.R. Troseth, H. Wang, A.D. Wentworth
ur research is interdisciplinary and involves
aspects of bioorganic, biophysical, physical
organic, synthetic, and analytical chemistry coupled with biochemical techniques, cell-based assays,
and animal models. We are interested in uncovering
new mechanisms of disease in major conditions such
as atherosclerosis, neurodegenerative diseases, ischemiareperfusion injury, macular degeneration, cancer, and
infectious diseases.
O
THE SCRIPPS RESEARCH INSTITUTE
111
his group, Department of Chemistry, we extended this
model and showed that these cholesterol seco-sterols
also trigger the misfolding of amyloid β-peptide 1-40 ,
leading to formation of fibrils similar to those observed
in patients with Alzheimer ’s disease. Using mutated
synthetic sequences of amyloid β-peptide1-40, we found
that the accelerated aggregation of this protein only
occurs when only lysine 16, not lysine 28 or the N-terminal amino group of aspartic acid 1, of the sequence
is modified. More recently, in studies of inflammatory
aldehyde–initiated misfolding of antibody light chains
(Bence-Jones proteins), we found that different aldehydes
can trigger different forms of aggregation in different
proteins. Thus, we have shown that the cholesterol
seco-sterols atheronal-A and atheronal-B accelerate an
amorphous form of aggregation, whereas 4-hydroxynonenal induced an amyloid form of aggregation of both λ
and κ light chains (Fig. 1).
A N T I B O D Y - C ATA LY Z E D WAT E R O X I D AT I O N PAT H WAY
Our discovery that all antibody molecules can catalyze the reaction between singlet oxygen and water to
give hydrogen peroxide is causing a revision of the idea
that antibodies are only an adapter molecule within
the immune system, linking recognition and killing of
foreign pathogens. We are exploring both the chemical
and biological aspects of this pathway, and new insights
into how the pathway plays a role in immune defense
and inflammatory damage are emerging.
We are searching for the active site for the antibody-catalyzed water oxidation pathway within the antibody structure. We have cloned and expressed soluble
individual domains (VHVL, CH1CL, VH, VL, CH1, CL) of
the murine Fab 4C6. All of the domains can generate
hydrogen peroxide when presented with singlet dioxygen, suggesting that the driving force is related to the
immunoglobulin fold of the whole antibody.
I N F L A M M AT O R Y A L D E H D Y E S A N D P R O T E I N
MISFOLDING
We have shown that the inflammation-derived cholesterol seco-sterols atheronal-A and atheronal-B trigger
a deformation in the secondary structure of the normally
folded low-density lipoprotein apoB-100 into a proamyloidogenic form. In collaboration with J.W. Kelly and
F i g . 1 . Electron micrograph of fibrillar aggregation of antibody
light chains induced by cholesterol seco-sterol and 4-hydroxynonenal (shown in white).
Epidemiologic and clinical evidence point to an
increased risk of cancer when linked with chronic inflammation, in a process thought to involve the establishment
of a local inflammatory microenvironment conducive to
the development of neoplasia. However, because of the
complex interrelationships between the 2 conditions, the
precise molecular and cellular mechanisms that underpin
this relationship remain largely unresolved.
We found that the inflammation-derived cholesterol
5,6-seco-sterol aldehydes atheronal-A and atheronal-B
cause a loss of function of wild-type tumor suppressor
protein p53, the so-called guardian of the genome, in
a process that involves p53 misfolding and amyloidogen-
112 CHEMISTRY
2008
esis. Atheronal-A and atheronal-B, but not the aldehydes 4-hydroxynonenal and 4-hydroxyhexenal derived
from polyunsaturated fatty acids, induce misfolding of
wild-type p53 into an amyloidogenic form that binds
thioflavin T and Congo red dye but cannot bind to a
consensus DNA sequence (Fig. 2). Treatment of lung car-
F i g . 2 . Optical microscopy images (100X) obtained with normal
(upper) and cross-polarized (lower) light of aggregates generated by
incubation of hexahistidine-tagged native p53 with atheronal-A and
stained with Congo red.
cinoma cells expressing wild-type p53 with atheronal-A
and atheronal-B leads to dysfunctional p53, as determined by analysis of extracted nuclear protein and transcription activation of p21.
Our results reveal a hitherto unknown chemical link
between inflammation and cancer and expand the already
pivotal role of p53 dysfunction in the risk for cancer. The
increasing generality and specificity of aldehyde-initiated
protein misfolding suggests that inflammatory aldehydes
and their posttranslational modification of amyloidogenic peptides may be the chemical link between the
known associations of inflammation, oxidative damage,
and various misfolding diseases.
THE SCRIPPS RESEARCH INSTITUTE
species), African trypanosomiasis (sleeping sickness,
Trypanosoma brucei), and American trypanosomiasis
(Chagas’ disease, Tr ypanosoma cruzi) have limited
effectiveness, thereby increasing drug resistance and
inherent toxic effects of the drugs. Thus, an elucidation of new parasite-specific biological targets for therapeutic agents is needed. In this regard, the discovery
that DNA from members of the order Kinetoplastida,
but not other eukaryotes, contains an unusual modified base, β- D -glucosyl(hydroxymethyl)uracil, called
base J, was a breakthrough. Extracts of several kinetoplastids contain a J-binding protein (JBP) that specifically binds to J-containing duplex DNA. JBP-1 is
essential in Leishmania.
As a drug target, JBP has merit. The protein shares
little homology with other proteins in the Protein Data
Bank, and it has a unique ligand, J-DNA containing
telomeric stretches of double-stranded DNA, that does
not occur in other eukaryotes. However, a preliminary
high-throughput screen, focused on disrupting binding
between JBP-1 and J-DNA, with a library of compounds
consisting of all the major drug pharmacophoric groups
has revealed no compounds of interest.
In parallel, we have studied the molecular recognition that underlies JBP-1 recognition of glycosylated
DNA. In collaboration with D.P. Millar and D.A. Case,
Department of Molecular Biology, we found that JBP-1
interacts with the J-containing DNA only when a critical conformation of the glucose within the major groove
is established. More recently, we discovered that low
micromolar concentrations of the DNA intercalators
daunorubicin and mitoxantrone disrupt the binding of
JBP-1 with duplex DNA containing J-DNA. Modeling
suggests that DNA binding of the intercalators leads
to distortion, which leads to disruption of the edge-on
conformation of the glucose within the major groove
of the DNA.
PUBLICATIONS
Grover, R.K., Wentworth, P., Jr. Emerging therapies for kinetoplastid diseases.
Prog. Infect. Dis., in press.
Nieva, J., Shafton, A., Altobell, L.J. III, Tripurenani, S., Rogel, J.K., Wentworth,
A.D., Lerner, R.A., Wentworth, P., Jr. Inflammatory aldehydes accelerate antibody
light chain amyloid and amorphous aggregation. Biochemistry 47:7695, 2008.
P R O T E I N 1 A N D G LY C O S Y L AT E D D N A
Scanlan, C.N., Ritchie, G.E., Baruah, K., Crispin, M.D., Harvey, D.J., Singer,
B.B., Lucka, L., Wormald, M.R., Wentworth, P., Jr., Zitzmann, N., Rudd, P.M.,
Burton, D.R., Dwek, R.A. Inhibition of mammalian glycan biosynthesis produces
non-self antigens for a broadly neutralising, HIV-1 specific antibody. J. Mol. Biol.
372:16, 2007.
Current treatments of parasitic infections such as
leishmaniasis (cutaneous or visceral, Leishmania
Scheinost, J.C., Boldt, G.E., Wentworth, P., Jr. Protein misfolding diseases. In:
Encyclopedia of Chemical Biology, Wiley Blackwell, New York, in press.
INTERACTION BETWEEN PROTOZOAN J-BINDING
CHEMISTRY
2008
Scheinost, J.C., Wang, H., Boldt, G.E., Offer, J., Wentworth, P., Jr. Cholesterol secosterol-induced aggregation of methylated amyloid-β peptides, insights into aldehydeinitiated fibrillization of amyloid-β. Angew. Chem. Int. Ed. 47:3919, 2008.
Temperini, C., Cecchi, A., Boyle, N.A., Scozzafava, A., Cabeza, J.E., Wentworth,
P., Jr., Blackburn, G.M., Supuran, C.T. Carbonic anhydrase inhibitors. Interaction
of 2-N,N-dimethylamino-1,3,4-thiadiazole-5-methylsulfonamide with 12 mammalian isoforms: kinetic and x-ray crystallographic studies. Bioorg. Med. Chem. Lett.
18:999, 2008.
Wentworth, P., Jr., Witter, D. Antibody-catalyzed water-oxidation pathway. Pure
Appl. Chem. 80:1849, 2008.
Bioorganic and Synthetic
Chemistry
C.-H. Wong, C. Bennett, S. Dean, S. Ficht, Y. Fu,
W. Greenberg, R. Guy, S. Hanson, Z. Hong, D.-R. Hwang,
M. Imamura, K. Kishikawa, J.-C. Lee, P.-H. Liang, L. Liu,
T. Polat, S.-K. Wang, Y.-Y. Yang
e develop new chemical and enzymatic strategies for synthesis of bioactive small molecules and biomolecules. We use the methods
to probe carbohydrate-mediated recognition events important in cancer, bacterial infections, and viral infections,
including HIV disease and influenza.
W
THE SCRIPPS RESEARCH INSTITUTE
113
these enzymes to catalyze new reactions and synthesize
new molecules of pharmaceutical relevance.
C A R B O H Y D R AT E - M E D I AT E D R E C O G N I T I O N I N
DISEASE
We are using our synthetic methods to discover
inhibitors and therapeutic agents in several diseases
related to carbohydrates. Current targets include bacterial transglycosidase, sulfatases, and glycoprocessing
enzymes involved in the biosynthesis of carbohydrates
that mediate cancer metastasis, inflammation, and viral
infection. Enzymatically synthesized iminocyclitols are
being investigated as treatments for osteoarthritis and
Gaucher disease. Inspired by the broadly neutralizing
anti-HIV antibody 2G12, which recognizes a dense
array of oligomannose displayed on HIV gp120, we
are designing dendrimeric oligomannose structures
for development of an HIV vaccine. In collaboration with
D.R. Burton, Department of Immunology, we are testing the immunogenicity of these constructs. We have
designed glycolipid ligands for CD1, which activate
natural killer T cells and are a promising new immunotherapeutic approach for treatment of bacterial and viral
infections and cancer. They may also be useful as
adjuvants in vaccine development.
SYNTHETIC METHODS
We have developed new methods for sugar-assisted
ligation of glycopeptides for synthesis of homogenous
glycoproteins. We have used the methods in conjunction with enzymatic glycosylation techniques to assemble
complex glycopeptides by chemical synthesis, and we
are optimizing the techniques to achieve the total synthesis of therapeutic glycoproteins. Glycoproteins are
expressed in vivo as complex mixtures of glycoforms,
a situation that hinders efforts to study the role of glycosylation in protein folding, stability, and function. By
synthesizing pure glycoforms, we can characterize in
molecular detail the effects of glycans on protein function.
Using chemical techniques such as programmable
1-pot oligosaccharide synthesis, as well as enzymatic
synthesis, we create glycoarrays on glass slides for highthroughput quantitative analysis of protein-carbohydrate
interactions. These arrays are being used to study the
binding specificity of carbohydrate-binding receptors and
antibodies. We have applied aldolases, glycosyltransferases, glycosidases, and other enzymes to develop
practical new methods of synthesizing molecules such
as iminocyclitols, which are inhibitors of glycosidases
and other enzymes, and glycopeptides, and other glycoconjugates. Using directed evolution, we are evolving
G LY C O P R O T E O M I C S A N D M O L E C U L A R G LY C O B I O L O G Y
Using metabolic oligosaccharide engineering, we
have developed methods for incorporating tagged sugars into glycans expressed on mammalian cells. The
engineered glycans can be labeled with a variety of
molecules by using click chemistry. One application is
glycan-specific fluorescent labeling, which is used for
fluorescent imaging to compare glycosylation patterns
of different cells, such as normal vs cancer cells or
cancer cells vs cancer stem cells. We found that protein fucosylation and sialylation are both elevated in
cancer cell lines.
A second application of this chemistry is GIDmap,
a new method for glycoproteomic analysis (Fig. 1).
Whole cells are fed with tagged sugars, and after biochemical incorporation of the sugars into cellular glycoproteins, click chemistry is used to attach a handle
for purification of tagged proteins. Mass spectrometric
proteomic methods are then used to identify proteins
that are differentially glycosylated. We are using GIDmap
to identify proteins that are aberrantly glycosylated in
different stages of cancer. These cancer-associated glycoproteins may be useful as biomarkers for diagnostics
or as targets for therapeutic intervention.
114 CHEMISTRY
2008
THE SCRIPPS RESEARCH INSTITUTE
Wang, S.-K., Liang, P.-H., Astronomo, R.D., Hsu, T.-L., Hsieh, S.-L., Burton,
D.R., Wong, C.-H. Targeting the carbohydrates on HIV-1: interaction of oligomannose dendrons with human monoclonal antibody 2G12 and DC-SIGN. Proc. Natl.
Acad. Sci. U. S. A. 105:3690, 2008.
Whalen, L.J., Greenberg, W.A., Mitchell, M.L., Wong, C.-H. Iminosugar-based glycosyltransferase inhibitors. In: Iminosugars: From Synthesis to Therapeutic Applications. Compain, P., Martin, O.R. (Eds.). Wiley-VCH, Hoboken, NJ, 2007, p. 153.
Wu, D., Fujio, M., Wong, C.-H. Glycolipids as immunostimulating agents. Bioorg.
Med. Chem. 16:1073, 2008.
Carbon-Hydrogen Activation,
Catalytic Reactions, and
Organometallic and
Synthetic Methods
J.-Q. Yu, K.M. Engle, R. Giri, T.-S. Mei, B.-F. Shi,
D.-H. Wang, M. Wasa, X.-S. Wang, Y.-H. Zhang
E N A N T I O S E L E C T I V E C A R B O N - H Y D R O G E N A C T I VAT I O N
C ATA LY Z E D B Y PA L L A D I U M ( I I ) – A M I N O A C I D
F i g . 1 . GIDmap glycoproteomic analysis via metabolic oligosac-
COMPLEXES
charide engineering.
lthough cleavage of inert carbon-hydrogen bonds
by transition metals has been extensively studied,
exploitation of this reactivity for regioselective
and enantioselective catalytic reactions of synthetically
useful chemical substances is still at its infant stage.
The 2 major challenges are the development of practical catalysis and the modulation of regioselectivity and
stereoselectivity by external ligands. We recently made
a number of discoveries that offer promising solutions
to these problems (Fig. 1).
PUBLICATIONS
Bennett, C.S., Dean, S.M., Payne, R.J., Ficht, S., Brik, A., Wong, C.-H. Sugarassisted glycopeptide ligation with complex oligosaccharides: scope and limitations.
J. Am. Chem. Soc. 130:11945, 2008.
Ficht, S., Payne, R.J., Guy, R.T., Wong, C.-H. Solid-phase synthesis of peptide
and glycopeptide thioesters through side-chain-anchoring strategies. Chem. Eur. J.
14:3620, 2008.
Giffin, M.J., Heaslet, H., Brik, A., Lin, Y.-C., Cauvi, G., Wong, C.-H., McRee,
D.E., Elder, J.H., Stout, C.D., Torbett, B.E. A copper(I)-catalyzed 1,2,3-triazole
azide-alkyne click compound is a potent inhibitor of a multidrug-resistant HIV-1
protease variant. J. Med. Chem. 51:6263, 2008.
A
Hanson, S.R., Greenberg, W.A., Wong C.-H. Probing glycans with the copper(I)catalyzed [3+2] azide-alkyne cycloaddition. QSAR Comb. Sci. 26:1243, 2007.
Kinjo, Y., Pei, B., Bufali, S., Raju, R., Richardson, S.K., Imamura, M., Fujio, M.,
Wu, D., Khurana, A., Kawahara, K., Wong, C.-H., Howell, A.R., Seeberger, P.H.,
Kronenberg, M. Natural Sphingomonas glycolipids vary greatly in their ability to
activate natural killer T cells. Chem. Biol. 15:654, 2008.
Liang, P.-H., Imamura, M., Li, X., Wu, D., Fujio, M., Guy, R., Wu, B.-C., Tsuji,
M., Wong, C.-H. Quantitative microarray analysis of intact glycolipid-CD1d interaction and correlation with cell-based cytokine production. J. Am. Chem. Soc.
130:12348, 2008.
Liang, P.-H., Wu, C.-Y., Greenberg, W.A., Wong, C.-H. Glycan arrays: biological
and medical applications. Curr. Opin. Chem. Biol. 12:86, 2008.
Northen, T.R., Lee, J.-C., Hoang, L., Raymond, J., Hwang, D.-R., Yannone, S.M.,
Wong, C.-H., Siuzdak, G. A nanostructure-initiator mass spectrometry-based
enzyme activity assay. Proc. Natl. Acad. Sci. U. S. A. 105:3678, 2008.
Payne, R.J., Ficht, S., Greenberg, W.A., Wong, C.-H. Cysteine-free peptide and glycopeptide ligation by direct aminolysis. Angew. Chem. Int. Ed. 47:4411, 2008.
Sugiyama, M., Hong, Z., Liang, P.-H., Whalen, L.J., Greenberg, W.A., Wong, C.H. D-Fructose-6-phosphate aldolase-catalyzed one-pot synthesis of iminocyclitols.
J. Am. Chem. Soc. 129:14811, 2007.
F i g . 1 . Enantioselective carbon-hydrogen activation/carbon-car-
bon coupling reactions.
We discovered the first palladium(II)/palladium(0)
catalytic system to couple both sp2 and sp3 carbon-hydrogen bonds with organotin and organoboron reagents. We
also established conditions to use air as the stoichiometric oxidant. We further discovered that mono-N-protected
amino acids are suitable ligands for enantioselective
carbon-hydrogen activation reactions. We think that
the α-chirality of amino acids is relayed to the monoprotected nitrogen center that coordinates with the
CHEMISTRY
2008
metal center and controls stereoselectivity at the carbon-hydrogen activation step. We are extending this
enzymelike chiral recognition to sp 3 carbon centers
attached to 2 prochiral carbon-hydrogen bonds (CH 2
groups) and are expanding the functional groups to
broadly useful carboxyl and amine groups.
THE SCRIPPS RESEARCH INSTITUTE
115
hydrogen bond, yielding a novel class of analogs in gram
quantities for biological studies (Fig. 3).
REAGENT-CONTROLLED MONOSELECTIVE
C A R B O N - H Y D R O G E N A C T I VAT I O N : C O N S T R U C T I O N
OF DEMANDING 1,2,3-SUBSTITUTED ARENES
To improve the practicality of carbon-hydrogen
activation reactions, we have focused on inventing new
approaches to activate carbon-hydrogen bonds in
abundant substrates containing broadly useful functional groups such carboxyl, amino, and hydroxyl groups.
We recently discovered that table salt promotes carbonhydrogen activation in arene and aliphatic carboxylic
acids. This reactivity led to the development of arylation
and halogenation of inert carbon-hydrogen bonds in carboxylic acids (Fig. 2). The replacement of table salt with
F i g . 3 . Carbon-hydrogen functionalization of natural products.
V E R S AT I L E H E T E R O C Y C L E S Y N T H E S I S F R O M
ARYLETHYLAMINES VIA CARBON-HYDROGEN
A C T I VAT I O N
Heterocycle synthesis is a core technology in medicinal chemistry. Using various amino groups to direct
carbon-hydrogen activation, we are developing novel
synthetic disconnections through amination of carbonhydrogen bonds. These reactions are either complementary to current methods or allow rapid access to
unique heterocyclic structures from readily available
chemicals (Fig. 4).
F i g . 2 . Reagent-controlled selective halogenation of ortho-car-
bon-hydrogen bonds.
bulkier tetraalkylammonium chloride salts markedly
improved the monoselectivity of ortho-carbon-hydrogen
functionalization. These reactions offer a solution to the
well-known challenge in accessing 1,2,3-substituted
arenes in medicinal chemistry and synthesis.
F U N C T I O N A L I Z AT I O N O F B I O L O G I C A L LY A C T I V E
N AT U R A L P R O D U C T S V I A C A R B O N - H Y D R O G E N
A C T I VAT I O N
Using carbon-hydrogen activation/carbon-carbon
coupling reactions, we hope to rapidly access diversified structures that are analogous to biologically active
compounds yet difficult to synthesize by using conventional methods. Dehydroabietic acid is a natural product identified as an efficient opener of BK ion channels.
Compounds with such activity could lead to useful
treatments for diseases such as acute stroke, epilepsy,
and asthma. Typically, diversification of such structures
is difficult because of the lack of reactive sites on
these molecules other than the carboxylic acid moiety,
which is essential for biological activity of the molecule. Masking the carboxylic acid as the hydroxamic
acid allows for functionalization at the methyl carbon-
F i g . 4 . Heterocycle synthesis via carbon-hydrogen activation.
PUBLICATIONS
Giri, R., Maugel, N.L., Foxman, B.M., Yu, J.Q. Dehydrogenation of inert alkyl
groups via remote C-H activation: converting a propyl group into a π-allylic complex. Organometallics 27:1667, 2008.
Giri, R., Maugel, N.L., Li, J.J., Wang, D.H., Breazzano, S.P., Saunders, L.B., Yu,
J.Q. Palladium-catalyzed methylation and arylation of sp2 and sp3 C-H bonds in
simple carboxylic acids. J. Am. Chem. Soc. 129:3510, 2007.
Giri, R., Yu, J.Q. Iodine monoacetate as a reagent. In: Encyclopedia of Reagents
for Organic Synthesis, 2nd ed. Paquette, L.A., et al. (Eds.). Wiley Blackwell, Hoboken, NJ, in press.
Huang, Y.Q., Shen, Z.L., Okamura, T.A., Wang, Y., Wang, X.F., Sun, W.Y., Yu,
J.Q., Ueyama, N. Silver(I) complexes with oxazoline-containing tripodal ligands:
structure variation via counter anions and reaction conditions. Dalton Trans. Issue
2:204, 2008.
Li, J.J., Giri, R., Yu, J.Q. Remote C-H bond functionalization reveals the distancedependent isotope effect. Tetrahedron 64:6979, 2008.
Mei, T.S., Giri, R., Maugel, N., Yu, J.Q. PdII-catalyzed monoselective ortho halogenation of C-H bonds assisted by counter cations: a complimentary method to
directed ortho lithiation. Angew. Chem. Int. Ed. 47:5215, 2008.
Shi, B.F., Maugel, N., Zhang, Y.H., Yu, J.Q. PdII-catalyzed enantioselective activation of C(sp2)-H and C(sp3)-H bonds using monoprotected amino acids as chiral
ligands. Angew. Chem. Int. Ed. 47:4882, 2008.
Wang, D.H., Wasa, M., Giri, R., Yu, J.Q. Pd(II)-catalyzed cross-coupling of sp3 CH bonds with sp2 and sp3 boronic acids using air as the oxidant. J. Am. Chem.
Soc. 130:7190, 2008.
Download