STRUCTURE-GUIDED, FRAGMENT-BASED LEAD GENERATION FOR ONCOLOGY TARGETS



Similar documents
How To Understand Protein-Protein Interaction And Inhibitors

De novo design in the cloud from mining big data to clinical candidate

Accelerating Lead Generation: Emerging Technologies and Strategies

We use Reaxys intensively for hit identification, hit-to-lead and lead optimization.

Corporate Presentation November, 2013

Combinatorial Chemistry and solid phase synthesis seminar and laboratory course

Eudendron: an Innovative Biotech Start-up

Data Visualization in Cheminformatics. Simon Xi Computational Sciences CoE Pfizer Cambridge

Call 2014: High throughput screening of therapeutic molecules and rare diseases

Lead generation and lead optimisation:

Crossing the drug development divide

How To Understand The Chemistry Of A 2D Structure

Integrating Medicinal Chemistry and Computational Chemistry: The Molecular Forecaster Approach

Use of Predictive ADME in Library Profiling and Lead Optimization

The Clinical Trials Process an educated patient s guide

Strategies and tactics for optimizing the Hit-to-Lead process and beyond A computational chemistry perspective

Exploiting the Pathogen box

Designed chemical libraries for hit/lead optimisation

exactly. The need for efficiency in developing effective new therapeutics has never been greater.

Pharmacology skills for drug discovery. Why is pharmacology important?

PHYSIOLOGY AND MAINTENANCE Vol. II - On The Determination of Enzyme Structure, Function, and Mechanism - Glumoff T.

Cheminformatics and its Role in the Modern Drug Discovery Process

Alterações empresariais sustentadas pelo conceito de engenharia do Produto Patrício Soares da Silva, MD, PhD

Computational Tools for Medicinal Chemists Increasing the Dimensions of Drug Discovery. Dr Robert Scoffin CEO

Ligand efficiency indices for effective drug discovery

Diabetes and Drug Development

FACT SHEET TESTETROL, A NOVEL ORALLY BIOACTIVE ANDROGEN

Drug design Drug repositioning Virtual screening

博 士 論 文 ( 要 約 ) A study on enzymatic synthesis of. stable cyclized peptides which. inhibit protein-protein interactions

Corporate Overview. Dr Robert Scoffin CEO. http;// STAND NUMBER: 27

Molecular Spectroscopy

Bachelor of Science in Applied Bioengineering

Course Curriculum for Master Degree in Clinical Pharmacy

The Open PHACTS Discovery Platform Semantic data integration for Medicinal Chemists

Scoring Functions and Docking. Keith Davies Treweren Consultants Ltd 26 October 2005

High-Throughput Screening at The University of Chicago Cellular Screening Center. Sam Bettis Technical Director

MYRIAD, HITACHI, ORACLE & FRIEDLI JOIN FORCES TO MAP THE ENTIRE HUMAN PROTEOME

Medicines for Neglected Diseases Workshop. Dennis Liotta, Ph.D. Director Emory Institute for Drug Discovery Atlanta, Georgia

Phase determination methods in macromolecular X- ray Crystallography

II Ill Ill1 II. /y3n-7 GAO. Testimony. Supercomputing in Industry

Biochemistry. Entrance Requirements. Requirements for Honours Programs. 148 Bishop s University 2015/2016

Consensus Scoring to Improve the Predictive Power of in-silico Screening for Drug Design

Florida Translational Research Program (FTRP)

Dr Alexander Henzing

pencil. Vocabulary: 1. Reactant 2. Product 3. Activation energy 4. Catalyst 5. substrate 6. Chemical reaction Keep your textbooks when you are done

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE S1A. Current Step 4 version

On the Impact of Virtual Screening to Genuine Lead Generation Campaigns

Cheminformatics and Pharmacophore Modeling, Together at Last

QSAR. The following lecture has drawn many examples from the online lectures by H. Kubinyi

Selvita Integrated drug discovery collaborations

Diversity screening versus focussed screening in drug discovery

Your partner in immunology

Strategic Consulting Services

Micromyx. Micromyx. A Microbiology Services Company. Lab Services Research - Consulting -

BROMOscan Quantitative Ligand Binding Assays

Chemistry/Biochemistry

M.Sc. in Nano Technology with specialisation in Nano Biotechnology

Apply with Resume to: Submenu Path Company/Careers/Current Openings/Job Type: Science

TECHNICAL INSIGHTS TECHNOLOGY ALERT

Academic Drug Discovery in the Center for Integrative Chemical Biology and Drug Discovery

Chapter 11: Molecular Structure of DNA and RNA

ICH Topic S 1 A The Need for Carcinogenicity Studies of Pharmaceuticals. Step 5

Data Warehouse Design for Pharmaceutical Drug Discovery Research

Bachelor of Science in Biochemistry and Molecular Biology

AAGPs TM Anti-Aging Glyco Peptides. Enhancing Cell, Tissue and Organ Integrity Molecular and biological attributes of lead AAGP molecule

COURSE TITLE COURSE DESCRIPTION

Name Date Class CHAPTER 1 REVIEW. Answer the following questions in the space provided.

MSC IN MEDICINAL CHEMISTRY

Lead optimization services

How To Make A Drug From A Peptide

Bringing Real World Practice into an Organic Chemistry Class. University of Wisconsin-Madison. The Institute on Learning Technology

Exelixis Showcases R&D Pipeline at JPMorgan Healthcare Conference

Zebrafish Chemical Screens

Proposal Submitted before WHO Expert Working Group on R&D Financing

Building innovative drug discovery alliances. Evotec Munich. Quantitative Proteomics to Support the Discovery & Development of Targeted Drugs

Drug Discovery in China

Perspectives on the Future of Graduate Education in Medicinal Chemistry and Pharmacognosy 1

CHEMISTRY. Real. Amazing. Program Goals and Learning Outcomes. Preparation for Graduate School. Requirements for the Chemistry Major (71-72 credits)

A career on the science park

The Techniques of Molecular Biology: Forensic DNA Fingerprinting

Cell Discovery 360: Explore more possibilities.

Ligand efficiency as a guide in fragment hit selection and optimization

SciFinder. Essential Content. Proven Results. Are You Ready for the Web? Österreichischer Bibliothekartag Innsbruck 2011

Contents. Page 1 of 21

Master of Philosophy (MPhil) and Doctor of Philosophy (PhD) Programs in Life Science

Luca Romagnoli, Ph.D. Business Development Manager

Leading Genomics. Diagnostic. Discove. Collab. harma. Shanghai Cambridge, MA Reykjavik

Salute in Horizon 2020: Giornata Nazionale di Lancio dei bandi

BBSRC TECHNOLOGY STRATEGY: TECHNOLOGIES NEEDED BY RESEARCH KNOWLEDGE PROVIDERS

If you need additional information please contact me at or esc8@,comcast.net).

Subject Area(s) Biology. Associated Unit Engineering Nature: DNA Visualization and Manipulation. Associated Lesson Imaging the DNA Structure

A Novel Bioconjugation Technology

Evalution the multiplexed analysis of biomarkers made fast and simple by MyCartis

High throughput screening, high content screening, primary and stem cells. new techniques now converging

SIPBS Portfolio Entry

M The Nucleus M The Cytoskeleton M Cell Structure and Dynamics

Tuberculosis OUR MISSION THE OPPORTUNITY

Bachelor of Science in Pharmaceutical Sciences (BSPS) Program Overview and Internship Requirements

CHEMISTRY. Faculty. Programs Offered. Bachelor of Science in Chemistry (certified by the American Chemical Society) Careers in Chemistry

Transcription:

STRUCTURE-GUIDED, FRAGMENT-BASED LEAD GENERATION FOR ONCOLOGY TARGETS Stephen K. Burley Structural GenomiX, Inc. 10505 Roselle Street, San Diego, CA 92121 sburley@stromix.com www.stromix.com Summary Structural GenomiX, Inc. (SGX) has combined high-throughput X-ray crystallography with a fragment-based approach to lead identification/lead optimization. FAST (Fragments of Active Structures) exploits crystallographic screening to detect, visualize, and identify small lead-like ligands (molecular weight 150-200, typical hit rates 1-5%) that bind productively to the target protein. Each member of the FAST fragment library is amenable to rapid chemical elaboration at two or three points to provide access to unrivalled chemical diversity using parallel organic synthesis. Initial lead optimization involves using knowledge of the structure of the target-fragment complex and advanced computational chemistry tools to guide synthesis of small focused linear (onedimensional) libraries. These linearly elaborated fragments are then evaluated with in vitro biochemical assays and crystallography. Thereafter, optimal variations at each point of chemical diversity are combined to synthesize focused combinatorial (two- or threedimensional) libraries that are again examined with assays and crystallography. (The theoretical chemical diversity of the fully elaborated FAST fragment library exceeds 160 million compounds.) These focused combinatorial libraries typically contain multiple novel compounds of low molecular weight (<350) that bind the target protein with low nm IC 50 values and display considerable selectivity. Finally, lead series are prioritized for further medicinal chemistry and compound advancement efforts using the results of cellular and animal model assays, in vitro and in vivo ADME and in vitro toxicology studies in concert with structural information. Application of the FAST fragment-based lead generation process to various oncology targets will be presented.

Structure based approaches have attracted the interest of the pharmaceutical and biotechnology industries for more than two decades. Until the advent of high-brightness synchrotron sources and cryogenic sample preservation methods in the 1990s, however, protein structures were only attainable after lengthy and resource-intensive efforts, greatly limiting their contributions to drug discovery. More recently, Nienaber and colleagues at Abbott Laboratories demonstrated the feasibility of using X-ray crystallography to screen for drug discovery starting points (Nienaber et al., 2000). Despite its promise, crystallographic screening has not been adopted widely because the approach depends critically on (i) the availability of high quality crystals of the target protein early in a drug discovery campaign, (ii) the ability to produce large numbers of target-small molecule complex structures in a time frame compatible with the needs of the medicinal chemist, and (iii) a reliable, efficient strategy for exploiting structural information to discover and optimize drug candidates. Today, high-throughput X-ray crystallography, advanced computational tools, and parallel organic synthesis have been combined to meet these challenges and provide new opportunities for lead discovery and optimization. By way of historical introduction, it is important to understand how efforts to introduce X-ray crystallography, computational design, and parallel organic synthesis combined with high-throughput screening into main-stream drug discovery fared. More than a decade ago X-ray crystallography was taken up by most of the large pharmaceutical companies, because of its promise to deliver rationally-designed drugs. Regrettably, this anticipated revolution in drug discovery failed to materialize. Earlier incarnations of the X-ray method usually proved to be too slow to keep up with the pace of medicinal chemistry. In addition, computational design strategies based on the structure of the protein target by itself did not in fact permit rational design of small molecule inhibitors of biochemical and biological function. Two other would be revolutionary methods came along later, both of which also failed to live up to expectations. The advent of combinatorial chemistry did allow chemists to make large numbers of molecules quickly, but these compounds rarely displayed the drug like properties needed to make it through pre-clinical testing and into patients. Highthroughput screening, which was developed to take advantage of the chemical bounty provided by combinatorial approaches, also failed to deliver useful lead compounds for many targets. In response to pressure to reduce discovery timelines for new pharmaceuticals and increase the rate at which new chemical entities are discovered and brought to market, Structural GenomiX, Inc. (SGX) has developed a novel technology called Fragments of Active Structures (FAST ) that serves as an engine for high-quality lead generation. With the benefit of technical advances that have reduced many of their historical liabilities, this new approach to drug discovery exploits the strengths of X-ray crystallographic screening, structure based lead optimization, computational design, and combinatorial chemistry. For crystallographic screening to deliver maximum value to the medicinal chemist, fragment hits must be suitable to rapid, iterative "hit-to-lead" optimization. The FAST process is based on a library of small, lead-like fragments that are fully enabled for chemical modification. With access to industry leading structure

determination capabilities, SGX quickly produces potent and selective small molecule inhibitors of drug targets by leveraging medicinal chemistry resources with structural information and insights from computational chemistry. FAST Fragment Hit Discovery The SGX fragment-based approach begins with crystallographic screening of a target protein against the FAST library of 1,000 small lead-like fragments (molecular weight 150-200). Each fragment has been tailored to include multiple points of diversity, or chemical handles, which are amenable to rapid parallel synthesis. The entire library is then screened within one to two days using a dedicated, state-of-the-art synchrotron beamline, SGX-CAT (Figure 1). Crystallographic screening entails soaking of preformed protein crystals with mixtures of ten structurally dissimilar fragments, followed by X-ray analysis of the soaked crystals. This approach yields hit rates of 1-5%, which is significantly higher than the hit rates of 0.010-0.025% typical for traditional highthroughput screening (HTS). The resulting structures provide direct looks at each one of the 10-50 lead-like fragments that bind to the crystalline target protein. The presence of bromine atoms in more than half of the SGX fragments facilitates the crystallogric screening process, using anomalous scattering signals to definitively establish the orientation of the bound fragment. Crystallographic screening hits are evaluated in terms of mode of binding and measured IC 50 (typically 1mM to 500µM) to identify the most promising subset of candidates for lead optimization against the target. Elaborating FAST Hits With detailed insights from the structure of each high-priority fragment bound to the target protein, small linear (one-dimensional) libraries are designed through modifications at each site of chemical diversity. The presence of bromine atoms in more than half of the SGX fragments provides a second important process advantage, enabling carbon-carbon formation via Suzuki coupling or the Heck reaction. By applying an advanced computational filter, chemical modifications predicted to make undesirable steric clashes with the target are excluded, thereby eliminating application of expensive synthetic resources to chemical modifications that are unlikely to increase potency and selectivity. Each one-dimensional library is synthesized and then evaluated using in vitro biochemical assays. Compounds with improved IC 50 values relative to the starting point or parent fragment (typically 10-100µM) are then examined by X-ray crystallography to visualize the effect of each chemical modification on mode of binding and room to move in terms of further synthetic chemistry. The most promising substitutions at each chemical handle are combined in small combinatorial (two- or three-dimensional) libraries of modifications at each site of chemical diversity. Again, advanced SGX computational tools are used to limit synthesis efforts to only those combinations of chemical modifications likely to improve potency and selectivity. Each combinatorial library is synthesized and then evaluated using in vitro biochemical assays and X-ray crystallography. At this stage, the best of these multiply elaborated fragments have IC 50 values of 10-100nM and often display considerable selectivity against closely related proteins. Most targets yield at least two structurally distinct lead series that can be evaluated for activity in cells and animal

models of disease. During the process of lead optimization via FAST structure-guided lead engineering, particular attention is paid to keeping the mass of the inhibitor as low as possible. Typical molecular weights of doubly and triply elaborated fragments are <350, which represent an important factor in determining the bioavailability of a drug candidate. Similar attention is paid to other determinants of bioavailability, such as solubility, lipophilicity, the number of freely rotatable bonds, and the number of hydrogen bond donors and acceptors (Wenlock et al., 2002). Chemical moieties with known liabilities in terms of toxicity and undesirable ADME properties were excluded from both the FAST library and the reagents used for parallel synthesis of the focused linear and combinatorial libraries around each high-priority fragment hit. Although the SGX fragment library consists of 1,000 compounds, the combinatorial nature of the FAST process ensures that the library has unrivalled potential in terms of chemical diversity and typically yields novel compounds after only two rounds of chemical synthesis. The number of elaborations possible at each site of chemical diversity ranges from 400 to 40,000. Thus, the worst case scenario of only 400 possible modifications at each one of only two chemical handles gives a minimum chemical diversity of 400*400=160,000 distinct compounds/fragment or 160 million distinct compounds from the FAST library. Less conservative estimates of the potential chemical diversity of the SGX fragment library give the number of distinct compounds in the hundreds of billions. The combination of high-throughput X-ray crystallography and advanced computational tools allows SGX to synthesize only the small fraction of this enormous bounty of potential compounds that is likely to yield compounds with desirable target binding properties. Throughout the process, fragment modifications generating previously patented compounds are excluded and computational filters are used to ensure that expensive synthetic resources are used to explore only novel regions of chemical shape space. The Changing Face of Lead Discovery The FAST process is a highly efficient and adaptable lead discovery strategy for using large numbers of X-ray structures of lead-like fragments bound to important disease targets. These structures serve as the building blocks for novel drug candidates by guiding further modifications of initial screening hits. The method exploits highthroughput X-ray crystallography to identify and visualize synthetically enabled fragments bound productively to kinases and other enzymes. Thereafter, knowledge of how each fragment binds to its target is combined with advanced computational tools to maximally leverage the efforts of parallel organic synthesis to produce engineered leads that are potent, reasonably selective, and have drug-like physiochemical properties. The speed with which a FAST campaign can be prosecuted (3-6 months) and the ability of the process to access an enormous scope of diverse and novel compounds makes it both competitive with and complementary to HTS (Table 1). FAST may also provide a useful means of approaching targets, such as proteases, for which HTS often fails to yield acceptable medicinal chemistry starting points. None of this would be possible without

dedicated, full time access to a high-brightness synchrotron beamline, an optimally designed fragment library, and proprietary computational chemistry software and hardware developed and retained by SGX. References Nienaber, V.L., Richardson, P.L., Klighofer, V. Bouska, J.J., Giranda, V.L, and Greer, J. (2000) Discovering novel ligands for macromolecules using X-ray crystallographic screening. Nature Biotechnology 18, 1105-1108. Wenlock, M.C., Austin, R.P., Barton, P., Davis, A.M., and Leeson, P.D. (2002) A comparison of physiochemical property profiles of development and marketed oral drugs. J. Med. Chem. 46, 1250-1256. Stephen K. Burley, M.D., D.Phil., F.R.S.C., is the Chief Scientific Officer and Senior Vice President of Research at SGX (San Diego) and Adjunct Professor at the Rockefeller University.

Table 1. Comparison of FAST and high-throughput screening (HTS) combined with lead optimization. FAST HTS/Lead Optimization Screening Library Size >160MM compounds 2MM compounds Hit Rate 1-5% 0.01-0.025% Lead IC 50 /Lead MW <100nM/ 350 MW <100nM/ 500 MW Novelty Potential High Limited by HTS library Time Required 3-6 months 8-15 months Drug Like Properties Built in at the outset Addressed later Target Limitations Crystallographically Many targets (e.g., enabled targets required (i.e., not applicable to proteases) fail to produce useful starting points for GPCRs and ion channels, lead optimization. at present).