Jump to content

Structural Genomics Consortium: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
m →‎External links: add authority control
SZAckloo (talk | contribs)
Editing 'Structural Genomics Consortium' Wikipedia page. Dear Wikipedia Colleagues. I have proposed here a number of major changes. I accept that such major changes are subject to thorough review and I welcome your advice. The changes are aimed to share a more detailed description of how the SGC started and how it has grown all while championing open science.
Line 1: Line 1:
The '''Structural Genomics Consortium''' (SGC) is a public-private-partnership focusing on elucidating the functions and disease relevance of all proteins encoded by the human genome, with an emphasis on those that are relatively understudied<sup>1,2</sup>. As a core principle, the SGC places all its research output into the public domain without restriction and never files for patents, making the SGC one of the leading proponents of [[open science]]<sup>3-13</sup>.{{Short description|A nonprofit consortium of bioscience researchers to openly share protein structure data}}
{{Short description|A nonprofit consortium of bioscience researchers to openly share protein structure data}}
The '''Structural Genomics Consortium''' ('''SGC''') is a not-for-profit organization formed in 2004 to determine the three-dimensional structures of [[protein]]s of medical relevance, and place them in the [[Protein Data Bank]] without restriction on use. The SGC operates out of the Universities of [[University of Oxford|Oxford]] and [[University of Toronto|Toronto]]. Over the past five years, the SGC has accounted for ~25% of the global output of novel human protein structures each year, and ~40% of the annual global output of structures of proteins from human parasites. The SGC target proteins have relevance to human health and disease, such as [[diabetes]], [[cancer]] and infectious diseases such as [[malaria]].


=== Governance ===
The SGC is an 'open data' partnership. All research results are published with no restriction.<ref>{{cite journal | author = Perkmann Markus, Schildt Henri | year = 2015 | title = Open Data Partnerships between Firms and Universities: The Role of Boundary Organizations | journal = Research Policy | volume = 44 | issue = 5| pages = 1133–1143 | doi=10.1016/j.respol.2014.12.006| doi-access = free }}</ref> The SGC is also spearheading an "open access" chemistry partnership&nbsp;– a new model for pre-competitive drug discovery in which the public and private sectors collaborate to generate potent and selective pharmacological inhibitors of human proteins that regulate epigenetic signalling, and commit to make these reagents available without restriction on use.
Founded in 2003, and modelled after the [[DbSNP|Single Nucleotide Polymorphism Database (dbSNP) Consortium]], the SGC is a charitable company whose Members comprise organizations that contribute over $5,4M Euros to the SGC over a five-year period. The Board has one representative from each Member and an independent Chair, who serves one 5-year term. The current Chair is Anke Müller-Fahrnow (Germany), and previous Chairs have been [https://www.city.ac.uk/people/academics/michael-morgan Michael Morgan] (U.K.), [[Wayne Hendrickson]] (U.S.A.), [https://www.bioc.uzh.ch/index.php?id=205 Markus Gruetter] (Switzerland) and [https://www.weforum.org/people/tetsuyuki-maruyama Tetsuyuki Maruyama] (Japan). The founding and current CEO is [[Aled Edwards]] (Canada).  The founding Members of the SGC Company were the [[Canadian Institutes of Health Research]], [[Genome Canada]], the Ontario Research Fund, [[GlaxoSmithKline]] and [[Wellcome Trust]]. The current (August 2020) Members comprise AbbVie, Bayer Pharma AG, Boehringer Ingelheim, the Eshelman Institute for Innovation, Genentech, Genome Canada, Janssen, Merck KGaA, MSD (Merck, Sharpe and Dohme), Pfizer, Takeda, and Wellcome Trust.


SGC research activities take place in a coordinated network of university-affiliated laboratories – at [[Goethe University Frankfurt]], [[Karolinska Institute|Karolinska Institutet]], [[McGill University]] the Universities of [[University of North Carolina at Chapel Hill|North Carolina at Chapel Hill]] and [[University of Toronto| Toronto]]. The research activities are supported both by funds from the SGC Company as well as by grants secured by the scientists affiliated with the SGC programs.  At each university, the scientific teams are led by a Chief Scientist, who are [https://www.bmls.de/news/news-2018/18th_Jan.html Stefan Knapp] (Goethe University Frankfurt), [https://www.cmm.ki.se/michael-sundstrom-team Michael Sundstrom] (Karolinska Institutet), [https://www.mcgill.ca/neuro/edward-fon-md Ted Fon] (McGill University),  [https://pharmacy.unc.edu/directory/tmw20653/ Tim Willson] (University of North Carolina at Chapel Hill), and [[Cheryl Arrowsmith]] (University of Toronto). The SGC currently comprises ~200 scientists.
In 2011, the SGC launched a project to create high-quality recombinant antibodies to proteins implicated in epigenetic events in an effort to stimulate research on these proteins. The project is carried out in partnership with leading academic research groups in the field (Tony Kossiakoff and Shohei Koide at University of Chicago, Sachdev Sidhu at the University of Toronto) and the laboratory of Jack Greenblatt at the University of Toronto. These reagents will be made available without restriction on use.


== Major Scientific Programs at the SGC ==
The SGC is headed by [[Aled Edwards]] (CEO/Director). Operations at each site are managed by a Chief Scientist&nbsp;– [[Cheryl Arrowsmith]] in [[Toronto, Ontario]], [[Canada]] and Chas Bountra in [[Oxford]], [[UK]].
[[Open science|Structural biology]] of human proteins – The SGC has contributed over [https://www.thesgc.org/structures 2200 structures] of human proteins of potential relevance for drug discovery into the [https://www.rcsb.org/ public domain] since 2003, and this effort continues.  An increasing number of structures constitute complexes with synthetic small molecules, an effort that is aided by a strategic partnership with the [[Diamond Light Source|Diamond synchrotron in Oxfordshire]]<sup>14</sup>.  


=== Chemical biology of human proteins ===
== Publication overviews ==
The SGC chemical probe program is focused on members of protein families in areas of science of relevance for human biology and drug discovery, and include [[Epigenetics|epigenetic signaling]]<sup>15,16</sup>, [[Solute carrier family|solute transport]]<sup>17,18</sup>, protein proteostasis<sup>19-23</sup>, and [https://www.thesgc.org/chemical-probes/kinase protein phosphorylation]<sup>11,24,25</sup>. Scientific strengths include bioinformatics ([https://chromohub.thesgc.org/static/ChromoHub.html ChromoHub], [https://ubihub.thesgc.org/static/UbiHub.html UbiHub]) for target identification and selectivity assays, protein production and biochemistry, crystallography and structure determination, biophysics, and cell biology (including target engagement assays). The SGC has contributed ~100 [https://www.thesgc.org/chemical-probes chemical probes] <sup>9,26,27</sup> into the public domain over the past decade, and >25,000 samples of these probes have been distributed to the scientific community. The chemical probes conform to the now community-standard quality criteria created by the SGC and its collaborative network<sup>9,28-32</sup>. Notable chemical probes include PFI-1<sup>33</sup> and JQ1<sup>34</sup> for the BET family, UNC0642<sup>35</sup> for G9a/GLP, UNC1999<sup>36</sup> for EZH2/H1, and OICR-9429<sup>37</sup> for WDR5.
;{{Prose|section|date=November 2017}}
; "Discovery of a Selective, Substrate Competitive Inhibitor of the Lysine Methyltransferase SETD8"
: To date, SETD8 is the only methyltransferase that induces monomethylation of histone H4 lysine 20 (H4K20), which is involved with the regulation of biological processes such as DNA damage response. SETD8 also monomethylates [[proliferating cell nuclear antigen]] (PCNA) and promotes [[carcinogenesis]], but inhibitors are virtually unknown besides nahuoic acid A. In result of this study, the first substrate-competitive inhibitor was found (UNC0379).<ref>{{cite journal|last1=Ma|first1=A|title=Discovery of a Selective, Substrate-Competitive Inhibitor of the Lysine Methyltransferase SETD8|journal=J Med Chem|volume=57|issue=15|pages=6822–6833|pmid=25032507|doi=10.1021/jm500871s|date=Aug 2014|pmc=4136711}}</ref>


=== Seeding drug discovery ===
; "Copper is required for oncogenic BRAF signalling and tumorigenesis": When the [[BRAF (gene)|BRAF]] kinase is mutated, it induces an active state of cancer (e.g. melanomas, thyroid cancer, etc.). The study found that when Copper Transporter 1 (CTR1) levels are decreased, the BRAF-related signaling and tumorigenesis levels decrease, also. Next, when human cells are transformed by BRAF or resistant to its inhibition, copper chelators used to treat [[Wilson's disease]] can help lessen tumor growth. When combined, the results of this study indicate that copper [[chelation therapy]] could be a very strong candidate for treating cancers related to any BRAF mutation.<ref>{{cite journal|last1=Brady|first1=DC|title=Copper is required for oncogenic BRAF signalling and tumorigenesis|journal=Nature|volume=509|issue=7501|pages=492–496|pmid=24717435|doi=10.1038/nature13180|date=May 2014|pmc=4138975}}</ref>
Through open science the SGC catalyzes drug discovery programs with a focus on the under-studied proteome. JQ1<sup>34</sup>, was used to validate BET bromodomains as tractable drug targets in cancer. There have been 35 clinical trials on [https://clinicaltrials.gov/ct2/results?cond=&term=bet+bromodomain&cntry=&state=&city=&dist= bromodomain] containing proteins (including 20 on the BET family), and additional bromodomain chemical probes are enabling research on over 20 targets<sup>26</sup>. Most recently, a [http://triphaseco.com/trph-395/ drug discovery program] in blood cancers was initiated based on data obtained using the open access chemical probe OICR-9429<sup>37</sup> that was co-developed by OICR and SGC. This probe linked WDR5 (a component of the MLL1 complex) to tumor growth suppression in leukemia. The drug discovery program resulted in a lead compound that ultimately attracted a [https://www.nortonrosefulbright.com/en-ca/about/client-work/ec7660a1/toronto-advised-facit-and-propellon-on-a-partnership-between-triphase-and-celgene strategic collaboration worth $40M USD]. Currently (08/20) there are ~15 [https://clinicaltrials.gov/ clinical trials] on 5 (non-bromodomain) epigenetic targets each of which the SGC has developed chemical probes.


The [https://www.thesgc.org/tep Target Enabling Package] (TEP) programme is built upon the recognition that genetic data is proving to be a powerful tool for target validation. TEPs provide a critical mass of reagents and knowledge on a protein target to allow rapid biochemical and chemical exploration and characterisation of proteins with genetic linkage to key disease areas. This dataset aims to catalyse new biology and disease understanding to target/ drug discovery. The SGC has [https://www.thesgc.org/tep opened target nominations to the public].
; "LRIG2 mutations cause urofacial syndrome": [[Urofacial syndrome]] (UFS) is linked with a major risk of kidney failure and unique facial expression when crying, smiling, or laughing. Those afflicted with UFS have a mutated LRIG2, which is a protein that is heavily involved with tumorigenesis and neural cell signaling. Previous studies showed that UFS can also be caused by mutations in encoding [[heparanase]]-2 (HPSE2), and after studying nerve fascicles within muscle bundles in the human fetal bladder, it is confirmed that both are involved in the neural development of the urinary tract.<ref>{{cite journal|last1=Stuart|first1=HM|title=LRIG2 mutations cause urofacial syndrome|journal=Am J Hum Genet|date=2013|volume=92|issue=2|pages=259–264|pmid=23313374|doi=10.1016/j.ajhg.2012.12.002|pmc=3567269}}</ref>


To provide relevant data for target validation the SGC started the [https://ultra-dd.org/ Unrestricted Leveraging of Targets for Research Advancement and Drug Discovery] (ULTRA-DD) program with funding from the European Commission’s [https://www.imi.europa.eu/ Innovative Medicines Initiative] (IMI). The goal is to identify and validate under-explored novel targets in auto-immune and inflammatory diseases creating and profiling target-directed chemical and antibody probes in the highest quality patient-cell derived assays, providing biomarker and [https://ultra-dd.org/tissue-platforms/cell-assay-datasets phenotypic read-outs] in a more disease relevant context<sup>38</sup>.
; "Human-Chromatin-Related Protein Interactions Identify a Demethylase Complex Required for Chromosome Segregation": The study resulted in a comprehensive protein-protein interaction map (chromatin-related) while also identifying 164 chromatin-modifying complexes. Also, they found that KDM8 and RCCD1 form a histone [[demethylase]] complex, which plays a very big role in the chromosome's stability and division.<ref>{{cite journal|last1=Edyta|first1=Marcon|title=Human-Chromatin-Related Protein Interactions Identify a Demethylase Complex Required for Chromosome Segregation|journal=Cell Reports|date=26 June 2014|volume=8|issue=1|pages=297–310|doi=10.1016/j.celrep.2014.05.050|pmid=24981860|doi-access=free}}</ref>


In the realm of non-human proteins, the SGC led [https://www.thesgc.org/sddc Structure-guided Drug Discovery Coalition] (SDDC) comprising the [https://www.ssgcid.org/ Seattle Structural Genomics Center for Infectious Disease] (SSGCID), the [https://www.anl.gov/bio/midwest-center-for-structural-genomics Midwest Center for Structural Genomics], the [https://csgid.org/ Center for Structural Genomics of Infectious Diseases] (CSGID), academic researchers in North America and Europe, and drug discovery teams from academia and industry has so far resulted in 7 early drug leads for tuberculosis (TB), malaria, and cryptosporidiosis. The SDDC receives funding from participating academic initiatives and the [[Bill & Melinda Gates Foundation]].
; "Protein Interferon-Stimulated Gene 15 Conjugation Delays but Does Not Overcome Coronavirus Proliferation in a Model of Fulminant Hepatitis": The study revolved around the pathway in the human immune system dealing with a [[coronavirus]] diagnosis. The results had a great indication of the ISG15 pathway being of great significance. Though there are likely numerous pathways or targets, the study provided great insight into the subject, nonetheless.<ref>{{cite journal|last1=Ma|first1=Xue-Zhong|title=Protein Interferon-Stimulated Gene 15 Conjugation Delays but Does Not Overcome Coronavirus Proliferation in a Model of Fulminant Hepatitis|journal=Journal of Virology|date=19 March 2014|volume=88|issue=11|pages=6195–6204|doi=10.1128/jvi.03801-13|pmid=24648452|pmc=4093886}}</ref>


More recently, the SGC, the University of North Carolina at Chapel Hill and the [https://unceii.org/ Eshelman Institute for Innovation], announced the launch of a global organization [https://www.readdi.org/ Rapidly Emerging Antiviral Drug Development Initiative] (READDI™) and Canada-based [https://vimiopen.org/ Viral Interruption to Medicines Initiative] (VIMI™). REDDI™ is modelled after a successful model for non-profit drug research and development [https://dndi.org/ Drugs for Neglected Diseases Initiative] (DNDi). READDI™ and VIMI™ non-profit, open science initiatives will focus on developing therapeutics for all viruses that have pandemic potential.
; "Novel Approaches for Targeting Kinases: Allosteric Inhibition, Allosteric Activation and Pseudokinases": Deregulation of protein kinases are very involved with a plethora of different diseases, and the study involved the finding of kinase inhibitors that have good pharmacokinetic/pharmacodynamic properties. It went over the different pathways involved and the possibilities for the future medical properties.<ref>{{cite journal|last1=Cowan-Jacob|first1=Sandra|title=Novel Approaches for Targeting Kinases: Allosteric Inhibition, Allosteric Activation and Pseudokinases|journal=Future Science|volume=6|issue=5|pages=541–561|doi=10.4155/fmc.13.216|pmid=24649957|year=2014}}</ref>


=== Open Science ===
; "The Roles of Jumonji-Type Oxygenases in Human Disease": The publication went over the human Jmj-type enzymes' involvement with diseases dealing with development, metabolism, and cancer. It went over the important biological processes and pathways that are heavily affected by any altering of the characteristic functions of the Jmj-type enzyme class.<ref>{{cite journal|last1=Johansson|first1=Catrine|title=The Roles of Jumonji-Type Oxygenases in Human Disease|journal=Epigenomics|volume=6|issue=1|pages=89–120|doi=10.2217/epi.13.79|pmid=24579949|pmc=4233403|year=2014}}</ref>
Since 2004, [https://www.nature.com/articles/nbt0509-409.pdf?origin=ppub open science] has been at the core of the SGC, and this was codified in the SGC Open Science Principles, which were developed by Edwards and Richard Gold, of McGill University. The [https://www.thesgc.org/click-trust-v2 principles]<sup>3-5,39</sup>, supported by the SGC host institutions, state that no SGC scientist will agree to file for a patent on any of their research activities, including medicinal chemistry. Freely available research reagents and technologies discovered and developed by the SGC, including cDNA clones ([http://www.addgene.org/sgc/ Addgene]), [https://www.thesgc.org/chemical-probes/ chemical probes], [https://www.thesgc.org/structures 3D structures], experimental protocols, are being used by thousands of laboratories around the world.


SGC scientists have been profiled extensively for their [https://www.rand.org/pubs/research_reports/RR512.html contributions to open science] and have been pioneering even more rapid dissemination of their science using [https://openlabnotebooks.org/ Open Lab Notebooks]<sup>8</sup>. This is particularly impactive to rare diseases, patient groups and disease foundations. For example [https://m4kpharma.com/ diffuse intrinsic pontine glioma] (DIPG), [https://openlabnotebooks.org/category/disease-foundation/fop/ Fibrodysplasia ossificans progressiva], [https://labscribbles.com/ Huntington’s disease]<sup>7,40</sup>, [https://www.michaeljfox.org/ Parkinson’s disease], and [https://themarkfoundation.org/ Chordoma].
; "Structural Basis for Histone Mimicry and Hijacking of Host Proteins by Influenza Virus Protein NS1": Diseases can arise from pathogens that mimic host proteins. The NS1 protein that is involved with [[influenza A]] that is well known for using mimicry to compete with host proteins. The study concluded that the mimicry was imperfect in its copying, but it still must have underlying involvement with influenza A.<ref>{{cite journal|last1=Qin|first1=Su|title=Structural Basis for Histone Mimicry and Hijacking of Host Proteins by Influenza Virus Protein NS1|journal=Nature Communications|date=21 December 2013|volume=5|issue=3952|pages=3952|doi=10.1038/ncomms4952|pmid=24853335|doi-access=free}}</ref>


=== Open Drug Discovery ===
; "Mutant Prolactin Receptor and Familial Hyperprolactinemia": The study dealt with three sisters with [[hyperprolactinemia]] and how their heredity is involved. The study concluded that the mutations that causes this disease leads to prolactin insensitivity and in turn a germline familial hyperprolactinemia.<ref>{{cite journal|last1=Newey|first1=Paul|title=Mutant Prolactin Receptor and Familial Hyperprolactinemia|journal=The New England Journal of Medicine|date=21 November 2013|pages=2012–2020|doi=10.1056/NEJMoa1307557|pmid=24195502|volume=369|issue=21|pmc=4209110}}</ref>
For 15 years, the SGC has maintained that the early stages of drug discovery, from new therapeutic hypothesis to proof-of-concept in patients should be for public good and should take place in the open<sup>3-8,10</sup>. The concept is being reduced to practice within a series of spin-off “no patent, open science” pharma companies<sup>6</sup>. These for-profit companies include [https://m4kpharma.com/ M4K Pharma] (Medicines for Kids), [https://www.utoronto.ca/news/u-t-researcher-s-open-science-drug-discovery-model-expands-neurodegenerative-diseases M4ND Pharma] (Medicines for Neurological Diseases) and M4ID Pharma (Medicines for Infectious Diseases).


All the M4 companies are wholly owned by a Canadian charity, whose mandate is to share scientific knowledge and ensure affordable access to all medicines. M4K Pharma has the most advanced open drug discovery program. Supported with funding from the [[Ontario Institute for Cancer Research]], the [https://www.thebraintumourcharity.org/ Brain Tumour Charity], [https://www.criver.com/ Charles River Laboratories] and [https://www.criver.com/ Reaction Biology], and with contributions from scientists at the Universities of McGill, North Carolina, Oxford, Pennsylvania, and Toronto and in the [https://www.sjdhospitalbarcelona.org/en Sant Joan de Déu hospital], the [[University Health Network]] hospitals, the [[The Hospital for Sick Children (Toronto)|Hospital for Sick Children]], and [https://www.icr.ac.uk/ The Institute of Cancer Research], M4K Pharma is developing a selective inhibitor of ALK2<sup>13</sup> for DIPG, a uniformly fatal pediatric brain tumour.
; "Why is Epigenetics Important in Understanding the Pathogenesis of Inflammatory Musculoskeletal Diseases?": The publication went over the underlying effects [[epigenetics]] has on the pathogenesis of musculoskeletal diseases. It mostly went over how the epigenetic machinery is extremely important in homeostasis and natural development. The overview went over the therapeutic medicine relevant to [[rheumatoid arthritis]] and its development. The researchers extend this claim by saying there are epigenetic implications with other autoimmune diseases and cancer.<ref>{{cite journal|last1=Oppermann|first1=Udo|title=Why Epigenetics Important in Understanding the Pathogenesis of Inflammatory Musculoskeletal Diseases?|journal=[[Arthritis Research & Therapy]]|date=3 April 2013|volume=15|issue=209|pages=209|doi=10.1186/ar4186|pmid=23566317|pmc=3672786}}</ref>


== Notable achievements ==
== Scientific Themes ==
[[Kinase|'''Kinases''']] have been the focus of many drug discovery programs and 50 drugs have been approved by the FDA for treatment of cancer, inflammation, and fibrosis<sup>41</sup>. But based on the literature only ~20% of the kinome has been studied. To expand the druggable kinome<sup>24</sup> the laboratories in Frankfurt, North Carolina and Oxford have so far contributed to one third of the deposited structures of kinases, and in collaboration with pharmaceutical companies and academia co-developed 11 [https://www.thesgc.org/chemical-probes/kinase chemical probes] (and matching negative controls), and version 1.0 of 187 well-characterized [https://www.sgc-unc.org/kcgs chemogenomic] inhibitors (aka KCGS) for 215 targets<sup>11,25</sup>.
{{Unreferenced section|date=November 2017}}
* Since 2008, SGC has developed selective and cell-permeable inhibitors of protein function (chemical probes) involved in [[epigenetic]] control such as G9a [[methyltransferase]] and Lysine [[demethylase]]
* Donated more than 1500 high-resolution structures of proteins that were relevant to human medicine to online databases
* Solved the structure of the first human [[ABC transporter]] called the mitochondrial ABC transporter ABCB10
* Released 63 human protein kinases to the public domain that never had been released before publicly


[[Epigenetics|'''Epigenetic proteins''']] regulate chromatin condensation or participate in DNA-templated processes and have been classified as “writers,” “erasers,” and “readers”<sup>15</sup>. Laboratories in Oxford and Toronto have (in the last 10 years) co-developed with pharmaceutical and academic partners [https://www.thesgc.org/chemical-probes/ 57 chemical probes and 44 negative controls].
== Partners ==
The SGC has numerous notable partners, such as [[AbbVie]], [[Bayer HealthCare Pharmaceuticals]], and [[Pfizer]]. Recently, these organizations together have committed more than US$65 million to the consortium to sustain operation from 2011 to 2015.


[[Ubiquitin#Ubiquitylation|'''Ubiquitin signaling proteins''']] can be similarly grouped into “writers,” “erasers,” and “readers”. By August 2015 the SGC had already solved (and deposited) the structures of over 65 protein domains involved in ubiquitin signaling<sup>42-44</sup>. Building on this, the SGC has been ramping up its activities in this protein family over the last 5 years<sup>20-23,45</sup>.
==Highlight publications (2008–2012)==

*{{cite journal |doi=10.1038/nchembio.599 |title=A chemical probe selectively inhibits G9a and GLP methyltransferase activity in cells |year=2011 |last1=Vedadi |first1=Masoud |last2=Barsyte-Lovejoy |first2=Dalia |last3=Liu |first3=Feng |last4=Rival-Gervier |first4=Sylvie |last5=Allali-Hassani |first5=Abdellah |last6=Labrie |first6=Viviane |last7=Wigle |first7=Tim J |last8=Dimaggio |first8=Peter A |last9=Wasney |first9=Gregory A |last10=Siarheyeva |first10=Alena |last11=Dong |first11=Aiping |last12=Tempel |first12=Wolfram |last13=Wang |first13=Sun-Chong |last14=Chen |first14=Xin |last15=Chau |first15=Irene |last16=Mangano |first16=Thomas J |last17=Huang |first17=Xi-Ping |last18=Simpson |first18=Catherine D |last19=Pattenden |first19=Samantha G |last20=Norris |first20=Jacqueline L |last21=Kireev |first21=Dmitri B |last22=Tripathy |first22=Ashutosh |last23=Edwards |first23=Aled |last24=Roth |first24=Bryan L |last25=Janzen |first25=William P |last26=Garcia |first26=Benjamin A |last27=Petronis |first27=Arturas |last28=Ellis |first28=James |last29=Brown |first29=Peter J |last30=Frye |first30=Stephen V |journal=Nature Chemical Biology |volume=7 |issue=8 |pages=566–74 |pmid=21743462 |pmc=3184254|display-authors=8 }}
[[Integral membrane protein|'''Integral membrane proteins''']] which are permanently attached to the cell membrane include the [[Solute carrier family|solute carrier (SLC) proteins]]. The SLCs are largely unexplored therapeutically and in fact as many as 30% of all SLCs are considered ‘orphaned’ because their substrate specificity and biological function are unknown. In 2019 a public-private partnership comprising 13 partners, including the SGC, formed the [https://re-solute.eu/ The RESOLUTE Consortium]<sup>18</sup> with funding from the [https://www.imi.europa.eu/ IMI]. RESOLUTE’s strategic goal is to foster appreciation and intensity of research on SLCs to establish the family as a tractable class for R&D.   
*{{cite journal |doi=10.1038/nmeth.1607 |title=A roadmap to generate renewable protein binders to the human proteome |year=2011 |last1=Colwill |first1=Karen |last2=Persson |first2=Helena |last3=Jarvik |first3=Nicholas E |last4=Wyrzucki |first4=Arkadiusz |last5=Wojcik |first5=John |last6=Koide |first6=Akiko |last7=Kossiakoff |first7=Anthony A |last8=Koide |first8=Shohei |last9=Sidhu |first9=Sachdev |last10=Dyson |first10=Michael R |last11=Pershad |first11=Kritika |last12=Pavlovic |first12=John D |last13=Karatt-Vellatt |first13=Aneesh |last14=Schofield |first14=Darren J |last15=Kay |first15=Brian K |last16=McCafferty |first16=John |last17=Mersmann |first17=Michael |last18=Meier |first18=Doris |last19=Mersmann |first19=Jana |last20=Helmsing |first20=Saskia |last21=Hust |first21=Michael |last22=Dübel |first22=Stefan |last23=Berkowicz |first23=Susie |last24=Freemantle |first24=Alexia |last25=Spiegel |first25=Michael |last26=Sawyer |first26=Alan |last27=Layton |first27=Daniel |last28=Nice |first28=Edouard |last29=Dai |first29=Anna |last30=Rocks |first30=Oliver |journal=Nature Methods |volume=8 |issue=7 |pages=551–8 |pmid=21572409|display-authors=8 }}

*{{cite journal |doi=10.1038/nature09504 |title=Selective inhibition of BET bromodomains |year=2010 |last1=Filippakopoulos |first1=Panagis |last2=Qi |first2=Jun |last3=Picaud |first3=Sarah |last4=Shen |first4=Yao |last5=Smith |first5=William B. |last6=Fedorov |first6=Oleg |last7=Morse |first7=Elizabeth M. |last8=Keates |first8=Tracey |last9=Hickman |first9=Tyler T. |last10=Felletar |first10=Ildiko |last11=Philpott |first11=Martin |last12=Munro |first12=Shonagh |last13=McKeown |first13=Michael R. |last14=Wang |first14=Yuchuan |last15=Christie |first15=Amanda L. |last16=West |first16=Nathan |last17=Cameron |first17=Michael J. |last18=Schwartz |first18=Brian |last19=Heightman |first19=Tom D. |last20=La Thangue |first20=Nicholas |last21=French |first21=Christopher A. |last22=Wiest |first22=Olaf |last23=Kung |first23=Andrew L. |last24=Knapp |first24=Stefan |last25=Bradner |first25=James E. |journal=Nature |volume=468 |issue=7327 |pages=1067–73 |pmid=20871596 |pmc=3010259|display-authors=8 }}
== History ==
*{{cite journal |doi=10.1016/j.cell.2011.05.039 |title=An Allosteric Inhibitor of the Human Cdc34 Ubiquitin-Conjugating Enzyme |year=2011 |last1=Ceccarelli |first1=Derek F. |last2=Tang |first2=Xiaojing |last3=Pelletier |first3=Benoit |last4=Orlicky |first4=Stephen |last5=Xie |first5=Weilin |last6=Plantevin |first6=Veronique |last7=Neculai |first7=Dante |last8=Chou |first8=Yang-Chieh |last9=Ogunjimi |first9=Abiodun |last10=Al-Hakim |first10=Abdallah |last11=Varelas |first11=Xaralabos |last12=Koszela |first12=Joanna |last13=Wasney |first13=Gregory A. |last14=Vedadi |first14=Masoud |last15=Dhe-Paganon |first15=Sirano |last16=Cox |first16=Sarah |last17=Xu |first17=Shuichan |last18=Lopez-Girona |first18=Antonia |last19=Mercurio |first19=Frank |last20=Wrana |first20=Jeff |last21=Durocher |first21=Daniel |last22=Meloche |first22=Sylvain |last23=Webb |first23=David R. |last24=Tyers |first24=Mike |last25=Sicheri |first25=Frank |journal=Cell |volume=145 |issue=7 |pages=1075–87 |pmid=21683433|display-authors=8 |doi-access=free }}

*{{cite journal |doi=10.1016/j.cell.2008.11.038 |title=Large-Scale Structural Analysis of the Classical Human Protein Tyrosine Phosphatome |year=2009 |last1=Barr |first1=Alastair J. |last2=Ugochukwu |first2=Emilie |last3=Lee |first3=Wen Hwa |last4=King |first4=Oliver N.F. |last5=Filippakopoulos |first5=Panagis |last6=Alfano |first6=Ivan |last7=Savitsky |first7=Pavel |last8=Burgess-Brown |first8=Nicola A. |last9=Müller |first9=Susanne |last10=Knapp |first10=Stefan |journal=Cell |volume=136 |issue=2 |pages=352–63 |pmid=19167335 |pmc=2638020|display-authors=8 }}
=== '''The Concept''' ===
*{{cite journal |doi=10.1038/nature07273 |title=Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1 |year=2008 |last1=Avvakumov |first1=George V. |last2=Walker |first2=John R. |last3=Xue |first3=Sheng |last4=Li |first4=Yanjun |last5=Duan |first5=Shili |last6=Bronner |first6=Christian |last7=Arrowsmith |first7=Cheryl H. |last8=Dhe-Paganon |first8=Sirano |journal=Nature |volume=455 |issue=7214 |pages=822–5 |pmid=18772889}}
In 2000, a group of companies and Wellcome conceptualized forming a Structural Genomics Consortium (SGC) to focus on determining the three-dimensional structures of human proteins<sup>1</sup>. The aim of the consortium was to place all structural information and supporting reagents into the public domain without restriction. The effort was designed to complement the other structural genomics programs in the world, which at the time focused largely on microbial proteins and aimed either identify new protein folds, or to achieve broad structural coverage of an organism ([[Structural genomics|https://en.wikipedia.org/wiki/Structural_genomics]]).
*{{cite journal |doi=10.1016/j.cell.2008.07.047 |title=Structural Coupling of SH2-Kinase Domains Links Fes and Abl Substrate Recognition and Kinase Activation |year=2008 |last1=Filippakopoulos |first1=Panagis |last2=Kofler |first2=Michael |last3=Hantschel |first3=Oliver |last4=Gish |first4=Gerald D. |last5=Grebien |first5=Florian |last6=Salah |first6=Eidarus |last7=Neudecker |first7=Philipp |last8=Kay |first8=Lewis E. |last9=Turk |first9=Benjamin E. |last10=Superti-Furga |first10=Giulio |last11=Pawson |first11=Tony |last12=Knapp |first12=Stefan |journal=Cell |volume=134 |issue=5 |pages=793–803 |pmid=18775312 |pmc=2572732|display-authors=8 }}

*{{cite journal |doi=10.1038/nmeth.f.202 |title=Protein production and purification |year=2008 |last1=Gräslund |first1=Susanne |last2=Nordlund |first2=Pär |last3=Weigelt |first3=Johan |last4=Bray |first4=James |last5=Gileadi |first5=Opher |last6=Knapp |first6=Stefan |last7=Oppermann |first7=Udo |last8=Arrowsmith |first8=Cheryl |last9=Hui |first9=Raymond |last10=Ming |first10=Jinrong |last11=Dhe-Paganon |first11=Sirano |last12=Park |first12=Hee-won |last13=Savchenko |first13=Alexei |last14=Yee |first14=Adelinda |last15=Edwards |first15=Aled |last16=Vincentelli |first16=Renaud |last17=Cambillau |first17=Christian |last18=Kim |first18=Rosalind |last19=Kim |first19=Sung-Hou |last20=Rao |first20=Zihe |last21=Shi |first21=Yunyu |last22=Terwilliger |first22=Thomas C |last23=Kim |first23=Chang-Yub |last24=Hung |first24=Li-Wei |last25=Waldo |first25=Geoffrey S |last26=Peleg |first26=Yoav |last27=Albeck |first27=Shira |last28=Unger |first28=Tamar |last29=Dym |first29=Orly |last30=Prilusky |first30=Jaime |journal=Nature Methods |volume=5 |issue=2 |pages=135–46 |pmid=18235434 |pmc=3178102|display-authors=8 }}
==== '''Phase I (2004-2007)''' ====
In July 2004, the SGC scientific program was launched, with activities at the Universities of Oxford and Toronto, and with a goal to contribute >350 human protein structures into the public domain by mid-2007. To be counted toward these goals, the proteins had to derive from a pre-defined list and the protein structures were required to meet pre-defined quality criteria. The quality of protein structures was and continues to be adjudicated by a committee of independent academic scientists. Michael Morgan was the Chair of the SGC Board, and the scientific activities were led by Cheryl Arrowsmith (Toronto) and Michael Sundstrom (Oxford). In mid 2005 [https://www.vinnova.se/en/ VINNOVA], [[Knut and Alice Wallenberg Foundation]] and [[Swedish Foundation for Strategic Research|The Foundation for Strategic Research (SSF)]] established the Swedish research node of the SGC. Experimental activities were launched at the Karolinska Institutet in Stockholm, led by Pär Nordlund and Johan Weigelt. Together, the three SGC laboratories contributed 392 human protein structures into the public domain by July 2007. A pilot program in the structural biology of proteins in the malaria parasite was also initiated<sup>46</sup>.

==== '''Phase II (2007-2011)''' ====
The new SGC mandate was to determine the structures of 650 proteins by mid 2011.  In this phase the SGC focused considerable activities in the areas of ubiquitination, protein phosphorylation, small G-proteins and epigenetics, and also initiated an effort in the structural biology of integral membrane proteins. In this phase, the SGC determined the structures of 665 human proteins from its Target List.

In 2009, with support from Wellcome and GSK, the SGC launched a program to develop freely-available [[Chemical probe|chemical probes]] to proteins involved in epigenetic signalling. This effort was predicated on the notion that there were many human proteins that received too little attention by the research community and the observation that research on previously lesser-studied human proteins increased upon the availability of a chemical probe<sup>2,4</sup>. To ensure that the chemical probes were of the highest quality, each was subject to two levels of review prior to their dissemination to the public. The first level was internal, through a Joint Management Committee comprising representatives from each member organization. The second was provided by a group of independent experts selected from academia.  This level of oversight was key to developing reagents of sufficient quality to support reproducible research<sup>47-49</sup>, and led to the creation of the [[Chemical Probes Portal]], which is a community-driven effort to provide expert insight into the quality and use of published chemical probes.

The SGC Memberships expanded to include Merck, Sharpe and Dohme, and Novartis. [[Wayne Hendrickson]] served as the Chair of the SGC Board.

==== '''Phase III (2011-2015)''' ====
The SGC mandate diversified to include goals of protein structures (200, including 5 structure of human integral membrane proteins) and chemical probes (30). Many of the chemical probes’ programs were undertaken in partnership with scientists in the pharmaceutical companies, which made the commitment to contribute the collaborative chemical probe into the public domain, without restriction.

In Phase III, the SGC, along with the SSGCID (<nowiki>https://www.ssgcid.org/</nowiki>) and the CSGID (<nowiki>https://csgid.org/</nowiki>), also launched the [https://www.thesgc.org/sddc Structure-guided Drug Discovery Coalition]. The SDDC, which continues to be funded by the [[Bill & Melinda Gates Foundation|Bill and Melinda Gates Foundation]] and led by [https://www.thesgc.org/sddc Chris Walpole], was formed to develop early drug leads for malaria and tuberculosis. To date, the SDDC and its partners have delivered 7 early-leads into the TB, malaria, and cryptosporidiosis drug discovery pipelines.

The SGC Memberships expanded to include AbbVie, Bayer AG, Boehringer Ingelheim, Eli Lilly and Janssen.  Markus Gruetter became the Chair of the SGC Board. Merck, Sharpe and Dohme left the consortium, as did the Canadian Institutes for Health Research.

==== '''Phase IV (2015-2020)''' ====
This phase included goals to generate new protein structures, new chemical probes and renewable and well-characterized antibodies to human proteins. Importantly the SGC initiated a concerted effort to develop disease-relevant, cell-based assays using (primary) cells or tissue from patients. This phase saw the launch of research activities at Goethe University in Frankfurt, at McGill University, and at the Universities of Campinas and North Carolina, and participation in [https://ultra-dd.org/ ULTRADD] and [https://re-solute.eu/ RESOLUTE]<sup>17,18</sup> within [https://www.imi.europa.eu/ IMI].

The SGC Memberships expanded to include Merck KGaA and the Eshelman Institute for Innovation.  Merck, Sharpe and Dohme re-joined while GSK and Eli Lilly left the SGC.  Tetsuyuki Maruyama became the Chair of the Board.  

= The Future - Target 2035 =
'''Target 2035''' is an [[open science]] initiative with the goal of creating chemical<sup>11,20,26,27</sup> and/or biological<sup>12,50</sup> tools for the entire proteome by 2035<sup>51</sup>. Seed projects currently underway include the SGC’s epigenetics chemical probe program<sup>52,53</sup>, the NIH’s Illuminating the Druggable Genome initiative for under-explored kinases, [[G protein-coupled receptor|GPCR’s]] and ion channels<sup>54-56</sup>, and [https://www.imi.europa.eu/ IMI]’s [https://re-solute.eu/ RESOLUTE] project on human SLCs<sup>17,18</sup>. These teams are linked to SGC’s global collaborative network<sup>2,9,29,38,49,50</sup> and Target 2035 is intended to draw from these experiences to bring the ‘dark genome’ into the light thus creating the opportunity to develop new therapeutics.


==References==
==References==
{{Reflist}}
{{reflist}}
<ref>{{cite journal
|vauthors=Williamson AR
|title=Creating a structural genomics consortium
|journal=Nat. Struct. Biol.
|volume=7 Suppl
|issue=
|pages=953
|date=November 2000
|pmid=11103997
|doi=10.1038/80726
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Edwards AM, Isserlin R, Bader GD, Frye SV, Willson TM, Yu FH
|title=Too many roads not taken
|journal=Nature
|volume=470
|issue=7333
|pages=163–5
|date=February 2011
|pmid=21307913
|doi=10.1038/470163a
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Edwards A
|title=Open-source science to enable drug discovery
|journal=Drug Discov. Today
|volume=13
|issue=17-18
|pages=731–3
|date=September 2008
|pmid=18790412
|doi=10.1016/j.drudis.2008.04.011
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Edwards AM, Bountra C, Kerr DJ, Willson TM
|title=Open access chemical and clinical probes to support drug discovery
|journal=Nat. Chem. Biol.
|volume=5
|issue=7
|pages=436–40
|date=July 2009
|pmid=19536100
|doi=10.1038/nchembio0709-436
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Masum H, et al.
|title=Ten simple rules for cultivating open science and collaborative R&D
|journal=PLoS Comput. Biol.
|volume=9
|issue=9
|pages=e1003244
|date=2013
|pmid=24086123
|pmc=3784487
|doi=10.1371/journal.pcbi.1003244
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Morgan MR, Roberts OG, Edwards AM
|title=Ideation and implementation of an open science drug discovery business model - M4K Pharma
|journal=Wellcome Open Res
|volume=3
|issue=
|pages=154
|date=2018
|pmid=30705971
|pmc=6346698
|doi=10.12688/wellcomeopenres.14947.1
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Harding RJ
|title=Open notebook science can maximize impact for rare disease projects
|journal=PLoS Biol.
|volume=17
|issue=1
|pages=e3000120
|date=January 2019
|pmid=30689629
|pmc=6366684
|doi=10.1371/journal.pbio.3000120
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Schapira M, Harding RJ
|title=Open laboratory notebooks: good for science, good for society, good for scientists
|journal=F1000Res
|volume=8
|issue=
|pages=87
|date=2019
|pmid=31448096
|pmc=6694453
|doi=10.12688/f1000research.17710.2
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Müller S, et al.
|title=Donated chemical probes for open science
|journal=Elife
|volume=7
|issue=
|pages=
|date=April 2018
|pmid=29676732
|pmc=5910019
|doi=10.7554/eLife.34311
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Drewry DH, Wells CI, Zuercher WJ, Willson TM
|title=A Perspective on Extreme Open Science: Companies Sharing Compounds without Restriction
|journal=SLAS Discov
|volume=24
|issue=5
|pages=505–514
|date=June 2019
|pmid=31034310
|pmc=6624833
|doi=10.1177/2472555219838210
|url=
}}</ref>
<ref>{{cite journal |last1=Wells |first1=Carrow I |last2=et |first2=al. |title=The Kinase Chemogenomic Set (KCGS): An open science resource for kinase vulnerability identification |date=2019 |doi=https://doi.org/10.1101/2019.12.22.886523}}</ref>
<ref>{{cite journal
|vauthors=Laflamme C, et al.
|title=Implementation of an antibody characterization procedure and application to the major ALS/FTD disease gene C9ORF72
|journal=Elife
|volume=8
|issue=
|pages=
|date=October 2019
|pmid=31612854
|pmc=6794092
|doi=10.7554/eLife.48363
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Ensan D, et al.
|title=Targeting ALK2: An Open Science Approach to Developing Therapeutics for the Treatment of Diffuse Intrinsic Pontine Glioma
|journal=J. Med. Chem.
|volume=63
|issue=9
|pages=4978–4996
|date=May 2020
|pmid=32369358
|doi=10.1021/acs.jmedchem.0c00395
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Collins PM, Douangamath A, Talon R, Dias A, Brandao-Neto J, Krojer T, von Delft F
|title=Achieving a Good Crystal System for Crystallographic X-Ray Fragment Screening
|journal=Meth. Enzymol.
|volume=610
|issue=
|pages=251–264
|date=2018
|pmid=30390801
|doi=10.1016/bs.mie.2018.09.027
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M
|title=Epigenetic protein families: a new frontier for drug discovery
|journal=Nat Rev Drug Discov
|volume=11
|issue=5
|pages=384–400
|date=April 2012
|pmid=22498752
|doi=10.1038/nrd3674
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Huston A, Arrowsmith CH, Knapp S, Schapira M
|title=Probing the epigenome
|journal=Nat. Chem. Biol.
|volume=11
|issue=8
|pages=542–5
|date=August 2015
|pmid=26196765
|doi=10.1038/nchembio.1871
|url=
}}</ref>
<ref>{{cite journal
|vauthors=César-Razquin A, et al.
|title=A Call for Systematic Research on Solute Carriers
|journal=Cell
|volume=162
|issue=3
|pages=478–87
|date=July 2015
|pmid=26232220
|doi=10.1016/j.cell.2015.07.022
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Superti-Furga G, et al.
|title=The RESOLUTE consortium: unlocking SLC transporters for drug discovery
|journal=Nat Rev Drug Discov
|volume=19
|issue=7
|pages=429–430
|date=July 2020
|pmid=32265506
|doi=10.1038/d41573-020-00056-6
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Liu L, Damerell DR, Koukouflis L, Tong Y, Marsden BD, Schapira M
|title=UbiHub: a data hub for the explorers of ubiquitination pathways
|journal=Bioinformatics
|volume=35
|issue=16
|pages=2882–2884
|date=August 2019
|pmid=30601939
|pmc=6691330
|doi=10.1093/bioinformatics/bty1067
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Schapira M, Calabrese MF, Bullock AN, Crews CM
|title=Targeted protein degradation: expanding the toolbox
|journal=Nat Rev Drug Discov
|volume=18
|issue=12
|pages=949–963
|date=December 2019
|pmid=31666732
|doi=10.1038/s41573-019-0047-y
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Harding RJ, et al.
|title=Small Molecule Antagonists of the Interaction between the Histone Deacetylase 6 Zinc-Finger Domain and Ubiquitin
|journal=J. Med. Chem.
|volume=60
|issue=21
|pages=9090–9096
|date=November 2017
|pmid=29019676
|doi=10.1021/acs.jmedchem.7b00933
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Ferreira de Freitas R, et al.
|title=Identification and Structure-Activity Relationship of HDAC6 Zinc-Finger Ubiquitin Binding Domain Inhibitors
|journal=J. Med. Chem.
|volume=61
|issue=10
|pages=4517–4527
|date=May 2018
|pmid=29741882
|doi=10.1021/acs.jmedchem.8b00258
|url=
}}}</ref>
<ref>{{cite journal
|vauthors=Mann MK, et al.
|title=Discovery of Small Molecule Antagonists of the USP5 Zinc Finger Ubiquitin-Binding Domain
|journal=J. Med. Chem.
|volume=62
|issue=22
|pages=10144–10155
|date=November 2019
|pmid=31663737
|doi=10.1021/acs.jmedchem.9b00988
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Knapp S, et al.
|title=A public-private partnership to unlock the untargeted kinome
|journal=Nat. Chem. Biol.
|volume=9
|issue=1
|pages=3–6
|date=January 2013
|pmid=23238671
|doi=10.1038/nchembio.1113
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Drewry DH, et al.
|title=Progress towards a public chemogenomic set for protein kinases and a call for contributions
|journal=PLoS ONE
|volume=12
|issue=8
|pages=e0181585
|date=2017
|pmid=28767711
|pmc=5540273
|doi=10.1371/journal.pone.0181585
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Wu Q, et al.
|title=A chemical toolbox for the study of bromodomains and epigenetic signaling
|journal=Nat Commun
|volume=10
|issue=1
|pages=1915
|date=April 2019
|pmid=31015424
|pmc=6478789
|doi=10.1038/s41467-019-09672-2
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Scheer S, et al.
|title=A chemical biology toolbox to study protein methyltransferases and epigenetic signaling
|journal=Nat Commun
|volume=10
|issue=1
|pages=19
|date=January 2019
|pmid=30604761
|pmc=6318333
|doi=10.1038/s41467-018-07905-4
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Frye SV
|title=The art of the chemical probe
|journal=Nat. Chem. Biol.
|volume=6
|issue=3
|pages=159–161
|date=March 2010
|pmid=20154659
|doi=10.1038/nchembio.296
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Arrowsmith CH, et al.
|title=The promise and peril of chemical probes
|journal=Nat. Chem. Biol.
|volume=11
|issue=8
|pages=536–41
|date=August 2015
|pmid=26196764
|pmc=4706458
|doi=10.1038/nchembio.1867
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Blagg J, Workman P
|title=Choose and Use Your Chemical Probe Wisely to Explore Cancer Biology
|journal=Cancer Cell
|volume=32
|issue=2
|pages=268–270
|date=August 2017
|pmid=28810148
|pmc=5559281
|doi=10.1016/j.ccell.2017.07.010
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Antolin AA, Tym JE, Komianou A, Collins I, Workman P, Al-Lazikani B
|title=Objective, Quantitative, Data-Driven Assessment of Chemical Probes
|journal=Cell Chem Biol
|volume=25
|issue=2
|pages=194–205.e5
|date=February 2018
|pmid=29249694
|pmc=5814752
|doi=10.1016/j.chembiol.2017.11.004
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Antolin AA, Workman P, Al-Lazikani B
|title=Public resources for chemical probes: the journey so far and the road ahead
|journal=Future Med Chem
|volume=
|issue=
|pages=
|date=November 2019
|pmid=31778323
|doi=10.4155/fmc-2019-0231
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Picaud S, et al.
|title=PFI-1, a highly selective protein interaction inhibitor, targeting BET Bromodomains
|journal=Cancer Res.
|volume=73
|issue=11
|pages=3336–46
|date=June 2013
|pmid=23576556
|pmc=3673830
|doi=10.1158/0008-5472.CAN-12-3292
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Filippakopoulos P, et al.
|title=Selective inhibition of BET bromodomains
|journal=Nature
|volume=468
|issue=7327
|pages=1067–73
|date=December 2010
|pmid=20871596
|pmc=3010259
|doi=10.1038/nature09504
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Liu F, et al.
|title=Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP
|journal=J. Med. Chem.
|volume=56
|issue=21
|pages=8931–42
|date=November 2013
|pmid=24102134
|pmc=3880643
|doi=10.1021/jm401480r
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Konze KD, et al.
|title=An orally bioavailable chemical probe of the Lysine Methyltransferases EZH2 and EZH1
|journal=ACS Chem. Biol.
|volume=8
|issue=6
|pages=1324–34
|date=2013
|pmid=23614352
|pmc=3773059
|doi=10.1021/cb400133j
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Grebien F, et al.
|title=Pharmacological targeting of the Wdr5-MLL interaction in C/EBPα N-terminal leukemia
|journal=Nat. Chem. Biol.
|volume=11
|issue=8
|pages=571–578
|date=August 2015
|pmid=26167872
|pmc=4511833
|doi=10.1038/nchembio.1859
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Edwards AM, et al.
|title=Preclinical target validation using patient-derived cells
|journal=Nat Rev Drug Discov
|volume=14
|issue=3
|pages=149–50
|date=March 2015
|pmid=25722227
|doi=10.1038/nrd4565
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Edwards A, et al.
|title=A trust approach for sharing research reagents
|journal=Sci Transl Med
|volume=9
|issue=392
|pages=
|date=May 2017
|pmid=28566431
|doi=10.1126/scitranslmed.aai9055
|url=
}}</ref>
<ref>{{cite web
|url=http://blogs.nature.com/scientificdata/2016/10/19/an-open-approach-to-huntingtons-disease-research/
|title=An open approach to Huntington’s disease research : Scientific Data
|format=
|work=
|accessdate=
}}</ref>
<ref>{{cite journal
|vauthors=Roskoski R
|title=Properties of FDA-approved small molecule protein kinase inhibitors
|journal=Pharmacol. Res.
|volume=144
|issue=
|pages=19–50
|date=June 2019
|pmid=30877063
|doi=10.1016/j.phrs.2019.03.006
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Bezsonova I, Walker JR, Bacik JP, Duan S, Dhe-Paganon S, Arrowsmith CH
|title=Ring1B contains a ubiquitin-like docking module for interaction with Cbx proteins
|journal=Biochemistry
|volume=48
|issue=44
|pages=10542–8
|date=November 2009
|pmid=19791798
|doi=10.1021/bi901131u
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Panier S, Ichijima Y, Fradet-Turcotte A, Leung CC, Kaustov L, Arrowsmith CH, Durocher D
|title=Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks
|journal=Mol. Cell
|volume=47
|issue=3
|pages=383–95
|date=August 2012
|pmid=22742833
|doi=10.1016/j.molcel.2012.05.045
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Sheng Y, et al.
|title=A human ubiquitin conjugating enzyme (E2)-HECT E3 ligase structure-function screen
|journal=Mol. Cell Proteomics
|volume=11
|issue=8
|pages=329–41
|date=August 2012
|pmid=22496338
|pmc=3412965
|doi=10.1074/mcp.O111.013706
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Kitevski-LeBlanc J, et al.
|title=The RNF168 paralog RNF169 defines a new class of ubiquitylated histone reader involved in the response to DNA damage
|journal=Elife
|volume=6
|issue=
|pages=
|date=April 2017
|pmid=28406400
|pmc=5426901
|doi=10.7554/eLife.23872
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Vedadi M, et al.
|title=Genome-scale protein expression and structural biology of Plasmodium falciparum and related Apicomplexan organisms
|journal=Mol. Biochem. Parasitol.
|volume=151
|issue=1
|pages=100–10
|date=January 2007
|pmid=17125854
|doi=10.1016/j.molbiopara.2006.10.011
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Frye SV, Arkin MR, Arrowsmith CH, Conn PJ, Glicksman MA, Hull-Ryde EA, Slusher BS
|title=Tackling reproducibility in academic preclinical drug discovery
|journal=Nat Rev Drug Discov
|volume=14
|issue=11
|pages=733–4
|date=November 2015
|pmid=26388229
|doi=10.1038/nrd4737
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Baker M
|title=1,500 scientists lift the lid on reproducibility
|journal=Nature
|volume=533
|issue=7604
|pages=452–4
|date=May 2016
|pmid=27225100
|doi=10.1038/533452a
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Edwards A
|title=Reproducibility: Team up with industry
|journal=Nature
|volume=531
|issue=7594
|pages=299–301
|date=March 2016
|pmid=26983524
|doi=10.1038/531299a
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Marcon E, et al.
|title=Assessment of a method to characterize antibody selectivity and specificity for use in immunoprecipitation
|journal=Nat. Methods
|volume=12
|issue=8
|pages=725–31
|date=August 2015
|pmid=26121405
|doi=10.1038/nmeth.3472
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Carter AJ, Kraemer O, Zwick M, Mueller-Fahrnow A, Arrowsmith CH, Edwards AM
|title=Target 2035: probing the human proteome
|journal=Drug Discov. Today
|volume=24
|issue=11
|pages=2111–2115
|date=November 2019
|pmid=31278990
|doi=10.1016/j.drudis.2019.06.020
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Song R, Wang ZD, Schapira M
|title=Disease Association and Druggability of WD40 Repeat Proteins
|journal=J. Proteome Res.
|volume=16
|issue=10
|pages=3766–3773
|date=October 2017
|pmid=28956604
|doi=10.1021/acs.jproteome.7b00451
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Schapira M, Tyers M, Torrent M, Arrowsmith CH
|title=WD40 repeat domain proteins: a novel target class?
|journal=Nat Rev Drug Discov
|volume=16
|issue=11
|pages=773–786
|date=November 2017
|pmid=29026209
|pmc=5975957
|doi=10.1038/nrd.2017.179
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Roth BL, Irwin JJ, Shoichet BK
|title=Discovery of new GPCR ligands to illuminate new biology
|journal=Nat. Chem. Biol.
|volume=13
|issue=11
|pages=1143–1151
|date=November 2017
|pmid=29045379
|pmc=5835362
|doi=10.1038/nchembio.2490
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Wacker D, Stevens RC, Roth BL
|title=How Ligands Illuminate GPCR Molecular Pharmacology
|journal=Cell
|volume=170
|issue=3
|pages=414–427
|date=July 2017
|pmid=28753422
|pmc=5560499
|doi=10.1016/j.cell.2017.07.009
|url=
}}</ref>
<ref>{{cite journal
|vauthors=Roth BL
|title=How structure informs and transforms chemogenetics
|journal=Curr. Opin. Struct. Biol.
|volume=57
|issue=
|pages=9–16
|date=August 2019
|pmid=30818201
|doi=10.1016/j.sbi.2019.01.016
|url=
}}</ref>


==External links==
==External links==
*[https://www.thesgc.org/openaccess/funders Partner List]
*[http://www.thesgc.org/ Global SGC website]
*[http://www.thesgc.org/ Global SGC website]
*[https://www.thesgc.org/profile/unicamp/parruda SGC Campinas]
*[https://www.sgc-unc.org/ SGC UNC]
*[https://www.sgc-frankfurt.de/ SGC Frankfurt]
*[https://www.thesgc.org/karolinska SGC Karolinska]
*[http://www.thesgc.org/scientists/groups/toronto SGC Toronto]
*[http://www.thesgc.org/scientists/groups/toronto SGC Toronto]
*[http://www.thesgc.org/scientists/groups/oxford SGC Oxford]
*[http://www.thesgc.org/scientists/groups/oxford SGC Oxford]
*Chemical probe resources: [[Chemical Probes Portal]], [https://probeminer.icr.ac.uk/ Probe Miner], [https://www.thesgc.org/chemical-probes SGC Chemical Probes], [https://www.thesgc.org/donated-chemical-probes SGC Donated Chemical Probes] Program
*[http://www.thesgc.org/about/key_achievements Notable Achievements]
*Chemogenomics: [https://www.sgc-unc.org/kcgs Kinase Chemogenomic Set v1.0]
*[http://www.thesgc.org/about/partners Partner List]


{{authority control}}
{{authority control}}

Revision as of 18:35, 31 August 2020

The Structural Genomics Consortium (SGC) is a public-private-partnership focusing on elucidating the functions and disease relevance of all proteins encoded by the human genome, with an emphasis on those that are relatively understudied1,2. As a core principle, the SGC places all its research output into the public domain without restriction and never files for patents, making the SGC one of the leading proponents of open science3-13.

Governance

Founded in 2003, and modelled after the Single Nucleotide Polymorphism Database (dbSNP) Consortium, the SGC is a charitable company whose Members comprise organizations that contribute over $5,4M Euros to the SGC over a five-year period. The Board has one representative from each Member and an independent Chair, who serves one 5-year term. The current Chair is Anke Müller-Fahrnow (Germany), and previous Chairs have been Michael Morgan (U.K.), Wayne Hendrickson (U.S.A.), Markus Gruetter (Switzerland) and Tetsuyuki Maruyama (Japan). The founding and current CEO is Aled Edwards (Canada).  The founding Members of the SGC Company were the Canadian Institutes of Health Research, Genome Canada, the Ontario Research Fund, GlaxoSmithKline and Wellcome Trust. The current (August 2020) Members comprise AbbVie, Bayer Pharma AG, Boehringer Ingelheim, the Eshelman Institute for Innovation, Genentech, Genome Canada, Janssen, Merck KGaA, MSD (Merck, Sharpe and Dohme), Pfizer, Takeda, and Wellcome Trust.

SGC research activities take place in a coordinated network of university-affiliated laboratories – at Goethe University Frankfurt, Karolinska Institutet, McGill University the Universities of North Carolina at Chapel Hill and  Toronto. The research activities are supported both by funds from the SGC Company as well as by grants secured by the scientists affiliated with the SGC programs.  At each university, the scientific teams are led by a Chief Scientist, who are Stefan Knapp (Goethe University Frankfurt), Michael Sundstrom (Karolinska Institutet), Ted Fon (McGill University),  Tim Willson (University of North Carolina at Chapel Hill), and Cheryl Arrowsmith (University of Toronto). The SGC currently comprises ~200 scientists.

Major Scientific Programs at the SGC

Structural biology of human proteins – The SGC has contributed over 2200 structures of human proteins of potential relevance for drug discovery into the public domain since 2003, and this effort continues.  An increasing number of structures constitute complexes with synthetic small molecules, an effort that is aided by a strategic partnership with the Diamond synchrotron in Oxfordshire14.  

Chemical biology of human proteins

The SGC chemical probe program is focused on members of protein families in areas of science of relevance for human biology and drug discovery, and include epigenetic signaling15,16, solute transport17,18, protein proteostasis19-23, and protein phosphorylation11,24,25. Scientific strengths include bioinformatics (ChromoHub, UbiHub) for target identification and selectivity assays, protein production and biochemistry, crystallography and structure determination, biophysics, and cell biology (including target engagement assays). The SGC has contributed ~100 chemical probes 9,26,27 into the public domain over the past decade, and >25,000 samples of these probes have been distributed to the scientific community. The chemical probes conform to the now community-standard quality criteria created by the SGC and its collaborative network9,28-32. Notable chemical probes include PFI-133 and JQ134 for the BET family, UNC064235 for G9a/GLP, UNC199936 for EZH2/H1, and OICR-942937 for WDR5.

Seeding drug discovery

Through open science the SGC catalyzes drug discovery programs with a focus on the under-studied proteome. JQ134, was used to validate BET bromodomains as tractable drug targets in cancer. There have been 35 clinical trials on bromodomain containing proteins (including 20 on the BET family), and additional bromodomain chemical probes are enabling research on over 20 targets26. Most recently, a drug discovery program in blood cancers was initiated based on data obtained using the open access chemical probe OICR-942937 that was co-developed by OICR and SGC. This probe linked WDR5 (a component of the MLL1 complex) to tumor growth suppression in leukemia. The drug discovery program resulted in a lead compound that ultimately attracted a strategic collaboration worth $40M USD. Currently (08/20) there are ~15 clinical trials on 5 (non-bromodomain) epigenetic targets each of which the SGC has developed chemical probes.

The Target Enabling Package (TEP) programme is built upon the recognition that genetic data is proving to be a powerful tool for target validation. TEPs provide a critical mass of reagents and knowledge on a protein target to allow rapid biochemical and chemical exploration and characterisation of proteins with genetic linkage to key disease areas. This dataset aims to catalyse new biology and disease understanding to target/ drug discovery. The SGC has opened target nominations to the public.

To provide relevant data for target validation the SGC started the Unrestricted Leveraging of Targets for Research Advancement and Drug Discovery (ULTRA-DD) program with funding from the European Commission’s Innovative Medicines Initiative (IMI). The goal is to identify and validate under-explored novel targets in auto-immune and inflammatory diseases creating and profiling target-directed chemical and antibody probes in the highest quality patient-cell derived assays, providing biomarker and phenotypic read-outs in a more disease relevant context38.

In the realm of non-human proteins, the SGC led Structure-guided Drug Discovery Coalition (SDDC) comprising the Seattle Structural Genomics Center for Infectious Disease (SSGCID), the Midwest Center for Structural Genomics, the Center for Structural Genomics of Infectious Diseases (CSGID), academic researchers in North America and Europe, and drug discovery teams from academia and industry has so far resulted in 7 early drug leads for tuberculosis (TB), malaria, and cryptosporidiosis. The SDDC receives funding from participating academic initiatives and the Bill & Melinda Gates Foundation.

More recently, the SGC, the University of North Carolina at Chapel Hill and the Eshelman Institute for Innovation, announced the launch of a global organization Rapidly Emerging Antiviral Drug Development Initiative (READDI™) and Canada-based Viral Interruption to Medicines Initiative (VIMI™). REDDI™ is modelled after a successful model for non-profit drug research and development Drugs for Neglected Diseases Initiative (DNDi). READDI™ and VIMI™ non-profit, open science initiatives will focus on developing therapeutics for all viruses that have pandemic potential.

Open Science

Since 2004, open science has been at the core of the SGC, and this was codified in the SGC Open Science Principles, which were developed by Edwards and Richard Gold, of McGill University. The principles3-5,39, supported by the SGC host institutions, state that no SGC scientist will agree to file for a patent on any of their research activities, including medicinal chemistry. Freely available research reagents and technologies discovered and developed by the SGC, including cDNA clones (Addgene), chemical probes, 3D structures, experimental protocols, are being used by thousands of laboratories around the world.

SGC scientists have been profiled extensively for their contributions to open science and have been pioneering even more rapid dissemination of their science using Open Lab Notebooks8. This is particularly impactive to rare diseases, patient groups and disease foundations. For example diffuse intrinsic pontine glioma (DIPG), Fibrodysplasia ossificans progressiva, Huntington’s disease7,40, Parkinson’s disease, and Chordoma.

Open Drug Discovery

For 15 years, the SGC has maintained that the early stages of drug discovery, from new therapeutic hypothesis to proof-of-concept in patients should be for public good and should take place in the open3-8,10. The concept is being reduced to practice within a series of spin-off “no patent, open science” pharma companies6. These for-profit companies include M4K Pharma (Medicines for Kids), M4ND Pharma (Medicines for Neurological Diseases) and M4ID Pharma (Medicines for Infectious Diseases).

All the M4 companies are wholly owned by a Canadian charity, whose mandate is to share scientific knowledge and ensure affordable access to all medicines. M4K Pharma has the most advanced open drug discovery program. Supported with funding from the Ontario Institute for Cancer Research, the Brain Tumour Charity, Charles River Laboratories and Reaction Biology, and with contributions from scientists at the Universities of McGill, North Carolina, Oxford, Pennsylvania, and Toronto and in the Sant Joan de Déu hospital, the University Health Network hospitals, the Hospital for Sick Children, and The Institute of Cancer Research, M4K Pharma is developing a selective inhibitor of ALK213 for DIPG, a uniformly fatal pediatric brain tumour.

Scientific Themes

Kinases have been the focus of many drug discovery programs and 50 drugs have been approved by the FDA for treatment of cancer, inflammation, and fibrosis41. But based on the literature only ~20% of the kinome has been studied. To expand the druggable kinome24 the laboratories in Frankfurt, North Carolina and Oxford have so far contributed to one third of the deposited structures of kinases, and in collaboration with pharmaceutical companies and academia co-developed 11 chemical probes (and matching negative controls), and version 1.0 of 187 well-characterized chemogenomic inhibitors (aka KCGS) for 215 targets11,25.

Epigenetic proteins regulate chromatin condensation or participate in DNA-templated processes and have been classified as “writers,” “erasers,” and “readers”15. Laboratories in Oxford and Toronto have (in the last 10 years) co-developed with pharmaceutical and academic partners 57 chemical probes and 44 negative controls.

Ubiquitin signaling proteins can be similarly grouped into “writers,” “erasers,” and “readers”. By August 2015 the SGC had already solved (and deposited) the structures of over 65 protein domains involved in ubiquitin signaling42-44. Building on this, the SGC has been ramping up its activities in this protein family over the last 5 years20-23,45.

Integral membrane proteins which are permanently attached to the cell membrane include the solute carrier (SLC) proteins. The SLCs are largely unexplored therapeutically and in fact as many as 30% of all SLCs are considered ‘orphaned’ because their substrate specificity and biological function are unknown. In 2019 a public-private partnership comprising 13 partners, including the SGC, formed the The RESOLUTE Consortium18 with funding from the IMI. RESOLUTE’s strategic goal is to foster appreciation and intensity of research on SLCs to establish the family as a tractable class for R&D.   

History

The Concept

In 2000, a group of companies and Wellcome conceptualized forming a Structural Genomics Consortium (SGC) to focus on determining the three-dimensional structures of human proteins1. The aim of the consortium was to place all structural information and supporting reagents into the public domain without restriction. The effort was designed to complement the other structural genomics programs in the world, which at the time focused largely on microbial proteins and aimed either identify new protein folds, or to achieve broad structural coverage of an organism (https://en.wikipedia.org/wiki/Structural_genomics).

Phase I (2004-2007)

In July 2004, the SGC scientific program was launched, with activities at the Universities of Oxford and Toronto, and with a goal to contribute >350 human protein structures into the public domain by mid-2007. To be counted toward these goals, the proteins had to derive from a pre-defined list and the protein structures were required to meet pre-defined quality criteria. The quality of protein structures was and continues to be adjudicated by a committee of independent academic scientists. Michael Morgan was the Chair of the SGC Board, and the scientific activities were led by Cheryl Arrowsmith (Toronto) and Michael Sundstrom (Oxford). In mid 2005 VINNOVA, Knut and Alice Wallenberg Foundation and The Foundation for Strategic Research (SSF) established the Swedish research node of the SGC. Experimental activities were launched at the Karolinska Institutet in Stockholm, led by Pär Nordlund and Johan Weigelt. Together, the three SGC laboratories contributed 392 human protein structures into the public domain by July 2007. A pilot program in the structural biology of proteins in the malaria parasite was also initiated46.

Phase II (2007-2011)

The new SGC mandate was to determine the structures of 650 proteins by mid 2011.  In this phase the SGC focused considerable activities in the areas of ubiquitination, protein phosphorylation, small G-proteins and epigenetics, and also initiated an effort in the structural biology of integral membrane proteins. In this phase, the SGC determined the structures of 665 human proteins from its Target List.

In 2009, with support from Wellcome and GSK, the SGC launched a program to develop freely-available chemical probes to proteins involved in epigenetic signalling. This effort was predicated on the notion that there were many human proteins that received too little attention by the research community and the observation that research on previously lesser-studied human proteins increased upon the availability of a chemical probe2,4. To ensure that the chemical probes were of the highest quality, each was subject to two levels of review prior to their dissemination to the public. The first level was internal, through a Joint Management Committee comprising representatives from each member organization. The second was provided by a group of independent experts selected from academia.  This level of oversight was key to developing reagents of sufficient quality to support reproducible research47-49, and led to the creation of the Chemical Probes Portal, which is a community-driven effort to provide expert insight into the quality and use of published chemical probes.

The SGC Memberships expanded to include Merck, Sharpe and Dohme, and Novartis. Wayne Hendrickson served as the Chair of the SGC Board.

Phase III (2011-2015)

The SGC mandate diversified to include goals of protein structures (200, including 5 structure of human integral membrane proteins) and chemical probes (30). Many of the chemical probes’ programs were undertaken in partnership with scientists in the pharmaceutical companies, which made the commitment to contribute the collaborative chemical probe into the public domain, without restriction.

In Phase III, the SGC, along with the SSGCID (https://www.ssgcid.org/) and the CSGID (https://csgid.org/), also launched the Structure-guided Drug Discovery Coalition. The SDDC, which continues to be funded by the Bill and Melinda Gates Foundation and led by Chris Walpole, was formed to develop early drug leads for malaria and tuberculosis. To date, the SDDC and its partners have delivered 7 early-leads into the TB, malaria, and cryptosporidiosis drug discovery pipelines.

The SGC Memberships expanded to include AbbVie, Bayer AG, Boehringer Ingelheim, Eli Lilly and Janssen.  Markus Gruetter became the Chair of the SGC Board. Merck, Sharpe and Dohme left the consortium, as did the Canadian Institutes for Health Research.

Phase IV (2015-2020)

This phase included goals to generate new protein structures, new chemical probes and renewable and well-characterized antibodies to human proteins. Importantly the SGC initiated a concerted effort to develop disease-relevant, cell-based assays using (primary) cells or tissue from patients. This phase saw the launch of research activities at Goethe University in Frankfurt, at McGill University, and at the Universities of Campinas and North Carolina, and participation in ULTRADD and RESOLUTE17,18 within IMI.

The SGC Memberships expanded to include Merck KGaA and the Eshelman Institute for Innovation.  Merck, Sharpe and Dohme re-joined while GSK and Eli Lilly left the SGC.  Tetsuyuki Maruyama became the Chair of the Board.  

The Future - Target 2035

Target 2035 is an open science initiative with the goal of creating chemical11,20,26,27 and/or biological12,50 tools for the entire proteome by 203551. Seed projects currently underway include the SGC’s epigenetics chemical probe program52,53, the NIH’s Illuminating the Druggable Genome initiative for under-explored kinases, GPCR’s and ion channels54-56, and IMI’s RESOLUTE project on human SLCs17,18. These teams are linked to SGC’s global collaborative network2,9,29,38,49,50 and Target 2035 is intended to draw from these experiences to bring the ‘dark genome’ into the light thus creating the opportunity to develop new therapeutics.

References

[1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56]

External links

  1. ^ Williamson AR (November 2000). "Creating a structural genomics consortium". Nat. Struct. Biol. 7 Suppl: 953. doi:10.1038/80726. PMID 11103997.
  2. ^ Edwards AM, Isserlin R, Bader GD, Frye SV, Willson TM, Yu FH (February 2011). "Too many roads not taken". Nature. 470 (7333): 163–5. doi:10.1038/470163a. PMID 21307913.
  3. ^ Edwards A (September 2008). "Open-source science to enable drug discovery". Drug Discov. Today. 13 (17–18): 731–3. doi:10.1016/j.drudis.2008.04.011. PMID 18790412.
  4. ^ Edwards AM, Bountra C, Kerr DJ, Willson TM (July 2009). "Open access chemical and clinical probes to support drug discovery". Nat. Chem. Biol. 5 (7): 436–40. doi:10.1038/nchembio0709-436. PMID 19536100.
  5. ^ Masum H, et al. (2013). "Ten simple rules for cultivating open science and collaborative R&D". PLoS Comput. Biol. 9 (9): e1003244. doi:10.1371/journal.pcbi.1003244. PMC 3784487. PMID 24086123.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  6. ^ Morgan MR, Roberts OG, Edwards AM (2018). "Ideation and implementation of an open science drug discovery business model - M4K Pharma". Wellcome Open Res. 3: 154. doi:10.12688/wellcomeopenres.14947.1. PMC 6346698. PMID 30705971.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  7. ^ Harding RJ (January 2019). "Open notebook science can maximize impact for rare disease projects". PLoS Biol. 17 (1): e3000120. doi:10.1371/journal.pbio.3000120. PMC 6366684. PMID 30689629.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  8. ^ Schapira M, Harding RJ (2019). "Open laboratory notebooks: good for science, good for society, good for scientists". F1000Res. 8: 87. doi:10.12688/f1000research.17710.2. PMC 6694453. PMID 31448096.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  9. ^ Müller S, et al. (April 2018). "Donated chemical probes for open science". Elife. 7. doi:10.7554/eLife.34311. PMC 5910019. PMID 29676732.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  10. ^ Drewry DH, Wells CI, Zuercher WJ, Willson TM (June 2019). "A Perspective on Extreme Open Science: Companies Sharing Compounds without Restriction". SLAS Discov. 24 (5): 505–514. doi:10.1177/2472555219838210. PMC 6624833. PMID 31034310.
  11. ^ Wells, Carrow I; et, al. (2019). "The Kinase Chemogenomic Set (KCGS): An open science resource for kinase vulnerability identification". doi:https://doi.org/10.1101/2019.12.22.886523. {{cite journal}}: Check |doi= value (help); Cite journal requires |journal= (help); External link in |doi= (help)
  12. ^ Laflamme C, et al. (October 2019). "Implementation of an antibody characterization procedure and application to the major ALS/FTD disease gene C9ORF72". Elife. 8. doi:10.7554/eLife.48363. PMC 6794092. PMID 31612854.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  13. ^ Ensan D, et al. (May 2020). "Targeting ALK2: An Open Science Approach to Developing Therapeutics for the Treatment of Diffuse Intrinsic Pontine Glioma". J. Med. Chem. 63 (9): 4978–4996. doi:10.1021/acs.jmedchem.0c00395. PMID 32369358.
  14. ^ Collins PM, Douangamath A, Talon R, Dias A, Brandao-Neto J, Krojer T, von Delft F (2018). "Achieving a Good Crystal System for Crystallographic X-Ray Fragment Screening". Meth. Enzymol. 610: 251–264. doi:10.1016/bs.mie.2018.09.027. PMID 30390801.
  15. ^ Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M (April 2012). "Epigenetic protein families: a new frontier for drug discovery". Nat Rev Drug Discov. 11 (5): 384–400. doi:10.1038/nrd3674. PMID 22498752.
  16. ^ Huston A, Arrowsmith CH, Knapp S, Schapira M (August 2015). "Probing the epigenome". Nat. Chem. Biol. 11 (8): 542–5. doi:10.1038/nchembio.1871. PMID 26196765.
  17. ^ César-Razquin A, et al. (July 2015). "A Call for Systematic Research on Solute Carriers". Cell. 162 (3): 478–87. doi:10.1016/j.cell.2015.07.022. PMID 26232220.
  18. ^ Superti-Furga G, et al. (July 2020). "The RESOLUTE consortium: unlocking SLC transporters for drug discovery". Nat Rev Drug Discov. 19 (7): 429–430. doi:10.1038/d41573-020-00056-6. PMID 32265506.
  19. ^ Liu L, Damerell DR, Koukouflis L, Tong Y, Marsden BD, Schapira M (August 2019). "UbiHub: a data hub for the explorers of ubiquitination pathways". Bioinformatics. 35 (16): 2882–2884. doi:10.1093/bioinformatics/bty1067. PMC 6691330. PMID 30601939.
  20. ^ Schapira M, Calabrese MF, Bullock AN, Crews CM (December 2019). "Targeted protein degradation: expanding the toolbox". Nat Rev Drug Discov. 18 (12): 949–963. doi:10.1038/s41573-019-0047-y. PMID 31666732.
  21. ^ Harding RJ, et al. (November 2017). "Small Molecule Antagonists of the Interaction between the Histone Deacetylase 6 Zinc-Finger Domain and Ubiquitin". J. Med. Chem. 60 (21): 9090–9096. doi:10.1021/acs.jmedchem.7b00933. PMID 29019676.
  22. ^ Ferreira de Freitas R, et al. (May 2018). "Identification and Structure-Activity Relationship of HDAC6 Zinc-Finger Ubiquitin Binding Domain Inhibitors". J. Med. Chem. 61 (10): 4517–4527. doi:10.1021/acs.jmedchem.8b00258. PMID 29741882.}
  23. ^ Mann MK, et al. (November 2019). "Discovery of Small Molecule Antagonists of the USP5 Zinc Finger Ubiquitin-Binding Domain". J. Med. Chem. 62 (22): 10144–10155. doi:10.1021/acs.jmedchem.9b00988. PMID 31663737.
  24. ^ Knapp S, et al. (January 2013). "A public-private partnership to unlock the untargeted kinome". Nat. Chem. Biol. 9 (1): 3–6. doi:10.1038/nchembio.1113. PMID 23238671.
  25. ^ Drewry DH, et al. (2017). "Progress towards a public chemogenomic set for protein kinases and a call for contributions". PLoS ONE. 12 (8): e0181585. doi:10.1371/journal.pone.0181585. PMC 5540273. PMID 28767711.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  26. ^ Wu Q, et al. (April 2019). "A chemical toolbox for the study of bromodomains and epigenetic signaling". Nat Commun. 10 (1): 1915. doi:10.1038/s41467-019-09672-2. PMC 6478789. PMID 31015424.
  27. ^ Scheer S, et al. (January 2019). "A chemical biology toolbox to study protein methyltransferases and epigenetic signaling". Nat Commun. 10 (1): 19. doi:10.1038/s41467-018-07905-4. PMC 6318333. PMID 30604761.
  28. ^ Frye SV (March 2010). "The art of the chemical probe". Nat. Chem. Biol. 6 (3): 159–161. doi:10.1038/nchembio.296. PMID 20154659.
  29. ^ Arrowsmith CH, et al. (August 2015). "The promise and peril of chemical probes". Nat. Chem. Biol. 11 (8): 536–41. doi:10.1038/nchembio.1867. PMC 4706458. PMID 26196764.
  30. ^ Blagg J, Workman P (August 2017). "Choose and Use Your Chemical Probe Wisely to Explore Cancer Biology". Cancer Cell. 32 (2): 268–270. doi:10.1016/j.ccell.2017.07.010. PMC 5559281. PMID 28810148.
  31. ^ Antolin AA, Tym JE, Komianou A, Collins I, Workman P, Al-Lazikani B (February 2018). "Objective, Quantitative, Data-Driven Assessment of Chemical Probes". Cell Chem Biol. 25 (2): 194–205.e5. doi:10.1016/j.chembiol.2017.11.004. PMC 5814752. PMID 29249694.
  32. ^ Antolin AA, Workman P, Al-Lazikani B (November 2019). "Public resources for chemical probes: the journey so far and the road ahead". Future Med Chem. doi:10.4155/fmc-2019-0231. PMID 31778323.
  33. ^ Picaud S, et al. (June 2013). "PFI-1, a highly selective protein interaction inhibitor, targeting BET Bromodomains". Cancer Res. 73 (11): 3336–46. doi:10.1158/0008-5472.CAN-12-3292. PMC 3673830. PMID 23576556.
  34. ^ Filippakopoulos P, et al. (December 2010). "Selective inhibition of BET bromodomains". Nature. 468 (7327): 1067–73. doi:10.1038/nature09504. PMC 3010259. PMID 20871596.
  35. ^ Liu F, et al. (November 2013). "Discovery of an in vivo chemical probe of the lysine methyltransferases G9a and GLP". J. Med. Chem. 56 (21): 8931–42. doi:10.1021/jm401480r. PMC 3880643. PMID 24102134.
  36. ^ Konze KD, et al. (2013). "An orally bioavailable chemical probe of the Lysine Methyltransferases EZH2 and EZH1". ACS Chem. Biol. 8 (6): 1324–34. doi:10.1021/cb400133j. PMC 3773059. PMID 23614352.
  37. ^ Grebien F, et al. (August 2015). "Pharmacological targeting of the Wdr5-MLL interaction in C/EBPα N-terminal leukemia". Nat. Chem. Biol. 11 (8): 571–578. doi:10.1038/nchembio.1859. PMC 4511833. PMID 26167872.
  38. ^ Edwards AM, et al. (March 2015). "Preclinical target validation using patient-derived cells". Nat Rev Drug Discov. 14 (3): 149–50. doi:10.1038/nrd4565. PMID 25722227.
  39. ^ Edwards A, et al. (May 2017). "A trust approach for sharing research reagents". Sci Transl Med. 9 (392). doi:10.1126/scitranslmed.aai9055. PMID 28566431.
  40. ^ "An open approach to Huntington's disease research : Scientific Data".
  41. ^ Roskoski R (June 2019). "Properties of FDA-approved small molecule protein kinase inhibitors". Pharmacol. Res. 144: 19–50. doi:10.1016/j.phrs.2019.03.006. PMID 30877063.
  42. ^ Bezsonova I, Walker JR, Bacik JP, Duan S, Dhe-Paganon S, Arrowsmith CH (November 2009). "Ring1B contains a ubiquitin-like docking module for interaction with Cbx proteins". Biochemistry. 48 (44): 10542–8. doi:10.1021/bi901131u. PMID 19791798.
  43. ^ Panier S, Ichijima Y, Fradet-Turcotte A, Leung CC, Kaustov L, Arrowsmith CH, Durocher D (August 2012). "Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks". Mol. Cell. 47 (3): 383–95. doi:10.1016/j.molcel.2012.05.045. PMID 22742833.
  44. ^ Sheng Y, et al. (August 2012). "A human ubiquitin conjugating enzyme (E2)-HECT E3 ligase structure-function screen". Mol. Cell Proteomics. 11 (8): 329–41. doi:10.1074/mcp.O111.013706. PMC 3412965. PMID 22496338.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  45. ^ Kitevski-LeBlanc J, et al. (April 2017). "The RNF168 paralog RNF169 defines a new class of ubiquitylated histone reader involved in the response to DNA damage". Elife. 6. doi:10.7554/eLife.23872. PMC 5426901. PMID 28406400.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  46. ^ Vedadi M, et al. (January 2007). "Genome-scale protein expression and structural biology of Plasmodium falciparum and related Apicomplexan organisms". Mol. Biochem. Parasitol. 151 (1): 100–10. doi:10.1016/j.molbiopara.2006.10.011. PMID 17125854.
  47. ^ Frye SV, Arkin MR, Arrowsmith CH, Conn PJ, Glicksman MA, Hull-Ryde EA, Slusher BS (November 2015). "Tackling reproducibility in academic preclinical drug discovery". Nat Rev Drug Discov. 14 (11): 733–4. doi:10.1038/nrd4737. PMID 26388229.
  48. ^ Baker M (May 2016). "1,500 scientists lift the lid on reproducibility". Nature. 533 (7604): 452–4. doi:10.1038/533452a. PMID 27225100.
  49. ^ Edwards A (March 2016). "Reproducibility: Team up with industry". Nature. 531 (7594): 299–301. doi:10.1038/531299a. PMID 26983524.
  50. ^ Marcon E, et al. (August 2015). "Assessment of a method to characterize antibody selectivity and specificity for use in immunoprecipitation". Nat. Methods. 12 (8): 725–31. doi:10.1038/nmeth.3472. PMID 26121405.
  51. ^ Carter AJ, Kraemer O, Zwick M, Mueller-Fahrnow A, Arrowsmith CH, Edwards AM (November 2019). "Target 2035: probing the human proteome". Drug Discov. Today. 24 (11): 2111–2115. doi:10.1016/j.drudis.2019.06.020. PMID 31278990.
  52. ^ Song R, Wang ZD, Schapira M (October 2017). "Disease Association and Druggability of WD40 Repeat Proteins". J. Proteome Res. 16 (10): 3766–3773. doi:10.1021/acs.jproteome.7b00451. PMID 28956604.
  53. ^ Schapira M, Tyers M, Torrent M, Arrowsmith CH (November 2017). "WD40 repeat domain proteins: a novel target class?". Nat Rev Drug Discov. 16 (11): 773–786. doi:10.1038/nrd.2017.179. PMC 5975957. PMID 29026209.
  54. ^ Roth BL, Irwin JJ, Shoichet BK (November 2017). "Discovery of new GPCR ligands to illuminate new biology". Nat. Chem. Biol. 13 (11): 1143–1151. doi:10.1038/nchembio.2490. PMC 5835362. PMID 29045379.
  55. ^ Wacker D, Stevens RC, Roth BL (July 2017). "How Ligands Illuminate GPCR Molecular Pharmacology". Cell. 170 (3): 414–427. doi:10.1016/j.cell.2017.07.009. PMC 5560499. PMID 28753422.
  56. ^ Roth BL (August 2019). "How structure informs and transforms chemogenetics". Curr. Opin. Struct. Biol. 57: 9–16. doi:10.1016/j.sbi.2019.01.016. PMID 30818201.