Daniel Nomura

Last updated
Daniel K. Nomura
20230921BP DanNomura82 square800px.jpg
BornJuly 10th, 1981
Alma materUniversity of California, Berkeley; University of California, Berkeley
Scientific career
FieldsChemical Biology, Chemistry, Molecular and Cell Biology, Drug Discovery, Cancer Biology
Institutions
  • UC Berkeley
  • Innovative Genomics Institute
  • Novartis-Berkeley Translational Chemical Biology Institute

Daniel K. Nomura is an American chemical biologist and Professor of Chemical Biology and Molecular Therapeutics at the University of California, Berkeley, in the Departments of Chemistry and Molecular & Cell Biology. His work employs chemoproteomic approaches to develop small molecule therapeutics and therapeutic modalities against traditionally "undruggable" proteins. [1]

Contents

He is a member of the scientific advisory committee of the Mark Foundation for Cancer Research, a foundation set up by billionaire Alexander Knaster which funds early-stage cancer research. [2] He is also an Investment Advisory Partner for a16z, an Investment Advisor for Droia Ventures, and an iPartner with The Column Group. [3]

Research and education

Nomura received his BA in Molecular and Cell Biology in 2003 and PhD in Molecular Toxicology in 2008 from UC Berkeley, studying under the mentorship of John Casida. He went on to perform his postdoctoral research with Ben Cravatt at Scripps Research in La Jolla, California. There he discovered a role for monoacylglycerol lipase in generating oncogenic signaling lipids that promote cancer [4] and in proinflammatory cascades that impact neurodegenerative disorders. [5] In 2011, Nomura started his independent research group at UC Berkeley. His work focuses on implementing chemoproteomic platforms to develop small molecule therapeutics against traditionally "undruggable" proteins. These approaches have led to the discovery of novel inhibitors and new ligands that expand the scope of proteolysis targeting chimeras (PROTACS), [6] which are bifunctional molecules that harness the cells ubiquitin-proteasome system to degrade targets of interest. Notable recent discoveries include an inhibitor of mTORC1, [7] a molecular glue between UBR7 and p53 that activates p53 tumor suppressor activity, [8] a covalent inhibitor against MYC, [9] novel covalent recruiters against E3 ubiquitin ligases such as RNF114, RNF4, and FEM1B for targeted protein degradation applications, [10] [11] [12] the Deubiquitinase Targeting Chimera (DUBTAC) platform for targeted protein stabilization, [13] and a chemical rational design strategy for developing molecular glue degraders. [14] In 2017, Nomura became the Director of the Novartis-Berkeley Translational Chemical Biology Institute, [15] [16] which aims to develop chemical technologies and therapeutics against undruggable targets. Nomura is also co-founder of Frontier Medicines and Vicinitas Therapeutics. [17] [18]

Awards

Related Research Articles

<span class="mw-page-title-main">Post-translational modification</span> Biological processes

Post-translational modification (PTM) is the covalent process of changing proteins following protein biosynthesis. PTMs may involve enzymes or occur spontaneously. Proteins are created by ribosomes translating mRNA into polypeptide chains, which may then change to form the mature protein product. PTMs are important components in cell signalling, as for example when prohormones are converted to hormones.

Benjamin Franklin Cravatt III is a professor in the Department of Chemistry at The Scripps Research Institute in La Jolla, California. Considered a co-inventor of activity-based proteomics and a substantial contributor to research on the endocannabinoid system, he is a prominent figure in the nascent field of chemical biology. Cravatt was elected to the National Academy of Sciences in 2014, and the American Academy of Arts and Sciences in 2016. He is Gilula Chair of Chemical Biology, a Cope Scholar, and a Searle Scholar.

<span class="mw-page-title-main">SWI/SNF</span> Subfamily of ATP-dependent chromatin remodeling complexes

In molecular biology, SWI/SNF, is a subfamily of ATP-dependent chromatin remodeling complexes, which is found in eukaryotes. In other words, it is a group of proteins that associate to remodel the way DNA is packaged. This complex is composed of several proteins – products of the SWI and SNF genes, as well as other polypeptides. It possesses a DNA-stimulated ATPase activity that can destabilize histone-DNA interactions in reconstituted nucleosomes in an ATP-dependent manner, though the exact nature of this structural change is unknown. The SWI/SNF subfamily provides crucial nucleosome rearrangement, which is seen as ejection and/or sliding. The movement of nucleosomes provides easier access to the chromatin, allowing genes to be activated or repressed.

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. The SGC places all its research output into the public domain without restriction and does not file for patents and continues to promote open science. Two recent publications revisit the case for open science. 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 Members comprise Bayer Pharma AG, Bristol Myers Squibb, Boehringer Ingelheim, the Eshelman Institute for Innovation, Genentech, Genome Canada, Janssen, Merck KGaA, Pfizer, and Takeda.

<span class="mw-page-title-main">Fatty-acid amide hydrolase 1</span> Mammalian protein found in Homo sapiens

Fatty-acid amide hydrolase 1 or FAAH-1(EC 3.5.1.99, oleamide hydrolase, anandamide amidohydrolase) is a member of the serine hydrolase family of enzymes. It was first shown to break down anandamide (AEA), an N-acylethanolamine (NAE) in 1993. In humans, it is encoded by the gene FAAH. FAAH also regulate the contents of NAE's in Dictyostelium discoideum, as they modulate their NAE levels in vivo through the use of a semispecific FAAH inhibitor.

<span class="mw-page-title-main">ADP-ribosylation</span> Addition of one or more ADP-ribose moieties to a protein.

ADP-ribosylation is the addition of one or more ADP-ribose moieties to a protein. It is a reversible post-translational modification that is involved in many cellular processes, including cell signaling, DNA repair, gene regulation and apoptosis. Improper ADP-ribosylation has been implicated in some forms of cancer. It is also the basis for the toxicity of bacterial compounds such as cholera toxin, diphtheria toxin, and others.

Druggability is a term used in drug discovery to describe a biological target that is known to or is predicted to bind with high affinity to a drug. Furthermore, by definition, the binding of the drug to a druggable target must alter the function of the target with a therapeutic benefit to the patient. The concept of druggability is most often restricted to small molecules but also has been extended to include biologic medical products such as therapeutic monoclonal antibodies.

<span class="mw-page-title-main">Stapled peptide</span>

A stapled peptide is a short peptide, typically in an alpha-helical conformation, that is constrained by a synthetic brace ("staple"). The staple is formed by a covalent linkage between two amino acid side-chains, forming a peptide macrocycle. Staples, generally speaking, refer to a covalent linkage of two previously independent entities. Peptides with multiple, tandem staples are sometimes referred to as stitched peptides. Among other applications, peptide stapling is notably used to enhance the pharmacologic performance of peptides.

<span class="mw-page-title-main">Targeted covalent inhibitors</span>

Targeted covalent inhibitors (TCIs) or Targeted covalent drugs are rationally designed inhibitors that bind and then bond to their target proteins. These inhibitors possess a bond-forming functional group of low chemical reactivity that, following binding to the target protein, is positioned to react rapidly with a proximate nucleophilic residue at the target site to form a bond.

Chemoproteomics entails a broad array of techniques used to identify and interrogate protein-small molecule interactions. Chemoproteomics complements phenotypic drug discovery, a paradigm that aims to discover lead compounds on the basis of alleviating a disease phenotype, as opposed to target-based drug discovery, in which lead compounds are designed to interact with predetermined disease-driving biological targets. As phenotypic drug discovery assays do not provide confirmation of a compound's mechanism of action, chemoproteomics provides valuable follow-up strategies to narrow down potential targets and eventually validate a molecule's mechanism of action. Chemoproteomics also attempts to address the inherent challenge of drug promiscuity in small molecule drug discovery by analyzing protein-small molecule interactions on a proteome-wide scale. A major goal of chemoproteomics is to characterize the interactome of drug candidates to gain insight into mechanisms of off-target toxicity and polypharmacology.

Craig M. Crews is an American scientist at Yale University known for his contributions to chemical biology. He is known for his contributions to the field of induced proximity through his work in creating heterobifunctional molecules that hijack cellular processes by inducing the interaction of two proteins inside a living cell. His initial work focused on the discovery of PROteolysis-TArgeting Chimeras (PROTACs) to trigger degradation of disease-causing proteins, a process known as targeted protein degradation (TPD), and he has since developed new versions of -TACs to leverage other cellular processes and protein families to treat disease.

A proteolysis targeting chimera (PROTAC) is a heterobifunctional molecule composed of two active domains and a linker, capable of removing specific unwanted proteins. Rather than acting as a conventional enzyme inhibitor, a PROTAC works by inducing selective intracellular proteolysis. PROTACs consist of two covalently linked protein-binding molecules: one capable of engaging an E3 ubiquitin ligase, and another that binds to a target protein meant for degradation. Recruitment of the E3 ligase to the target protein results in ubiquitination and subsequent degradation of the target protein via the proteasome. Because PROTACs need only to bind their targets with high selectivity, there are currently many efforts to retool previously ineffective inhibitor molecules as PROTACs for next-generation drugs.

Susan Marqusee is the Eveland Warren Endowed Chair Professor of Biochemistry, Biophysics, and Structural Biology at the University of California, Berkeley, and the Berkeley campus director of the California Institute for Quantitative Biosciences. Her research concerns the structure and dynamics of protein molecules. She received her A.B. in Physics and Chemistry from Cornell University in 1982, and her Ph.D. (Biochemistry) and M.D. degrees from Stanford University in 1990, where she trained with Robert Baldwin on the intrinsic helical properties of amino acids in model peptides.

Nathanael S. Gray is a Krishnan-Shah Family Professor of chemical and systems biology at Stanford University and director of cancer therapeutics programme at Stanford University School of Medicine. Previously he was a Nancy Lurie Marks Professor of biological chemistry and molecular pharmacology at Harvard Medical School and professor of cancer biology at Dana–Farber Cancer Institute. Gray is also co-founder, science advisory board member (SAB) and equity holder in C4 Therapeutics, Gatekeeper, Syros, Petra, B2S, Aduro, Jengu, Allorion, Inception Therapeutics, and Soltego. C4 Therapeutics, which offered IPO in 2020, was founded based on the ground-breaking research of Jay Bradner, current president of Novartis Institutes for BioMedical Research (NIBR), and of Nathanael S. Gray, while he was professor at Harvard Medical School. Before moving to Stanford University, Nathanael S. Gray created Center for Protein Degradation at Harvard Medical School with $80 million agreement with Deerfield Management venture capital firm. In 2020, Gray Lab permanently moved to Stanford University, that was stated by Stuart Schreiber, co-founder of Broad Institute as "Stanford's huge gain".

Molecular glue refers to a class of chemical compounds or molecules that play a crucial role in binding and stabilizing protein-protein interactions in biological systems. These molecules act as "glue" by enhancing the affinity between proteins, ultimately influencing various cellular processes. Molecular glue compounds have gained significant attention in the fields of drug discovery, chemical biology, and fundamental research due to their potential to modulate protein interactions, and thus, impact various cellular pathways. They have unlocked avenues in medicine previously thought to be “undruggable.”

<span class="mw-page-title-main">Nicolas H. Thomä</span> German structural and chemical biologist

Nicolas H. Thomä is a German researcher, full professor at the EPFL School of Life Sciences and Director of the Paternot Chair for Cancer Research in Lausanne, Switzerland. He is a biochemist and structural biologist and a leading researcher in the fields of ubiquitin ligase biology and DNA repair.

Angela N. Koehler is an American biochemist who is the Karl Van Tassel (1925) Career Development Professor of Chemical Biology at the Broad Institute. Her research considers the development of chemical tools to understand transcriptional regulation, and the design of next-generation pharmaceuticals.

James Allen Wells is a Professor of Pharmaceutical Chemistry and Cellular & Molecular Pharmacology at the University of California, San Francisco (UCSF) and a member of the National Academy of Sciences. He received his B.A. degrees in biochemistry and psychology from University of California, Berkeley in 1973 and a PhD in biochemistry from Washington State University with Ralph Yount, PhD in 1979. He completed his postdoctoral studies at Stanford University School of Medicine with George Stark in 1982. He is a pioneer in protein engineering, phage display, fragment-based lead discovery, cellular apoptosis, and the cell surface proteome.

Trimethoprim-Halotag (TMP-HTag) is a small molecule chemical linker developed for the rapid and reversible control of protein localization in living cells (Ballister). TMP is an dihydrofolate reductase (DHFR) inhibitor chosen for its specificity in binding to the bacterial form of DHFR. The other half of the linker is a Halotag, a self labelling bacterial globular protein ligand that can bind covalently and irreversibly to the chloroalkane group of a Haloenzyme. Positioned between the TMP group and HaloTag is a flexible linker that can be modified to optimize protein linking efficiency. The modular structure of TMP-HaloTag makes it an ideal heterobifunctional tool for use in chemically induced dimerization (CID). Additionally, TMP- HTag can be modified to include photo-cleavable groups that allow for the control of CID using light.

Chimeric small molecule therapeutics are a class of drugs designed with multiple active domains to operate outside of the typical protein inhibition model. While most small molecule drugs inhibit target proteins by binding their active site, chimerics form protein-protein ternary structures to induce degradation or, less frequently, other protein modifications.

References

  1. "Novartis and Berkeley researchers team up to tackle the industry's toughest drug targets". Chemical & Engineering News. Retrieved 2020-08-27.
  2. "Daniel Nomura, PhD". 26 January 2021.
  3. "Droia Ventures – Investing in Impact – Life Science". Droia Ventures – Investing in Impact. Retrieved 2022-11-21.
  4. Nomura, Daniel K.; Long, Jonathan Z.; Niessen, Sherry; Hoover, Heather S.; Ng, Shu-Wing; Cravatt, Benjamin F. (2010-01-08). "Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis". Cell. 140 (1): 49–61. doi:10.1016/j.cell.2009.11.027. ISSN   1097-4172. PMC   2885975 . PMID   20079333.
  5. Nomura, Daniel K.; Morrison, Bradley E.; Blankman, Jacqueline L.; Long, Jonathan Z.; Kinsey, Steven G.; Marcondes, Maria Cecilia G.; Ward, Anna M.; Hahn, Yun Kyung; Lichtman, Aron H.; Conti, Bruno; Cravatt, Benjamin F. (2011-11-11). "Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation". Science. 334 (6057): 809–813. Bibcode:2011Sci...334..809N. doi:10.1126/science.1209200. ISSN   1095-9203. PMC   3249428 . PMID   22021672.
  6. Jarvis, Lisa M. "Targeted protein degraders are redefining how small molecules look and act". Chemical & Engineering News. Retrieved 2020-08-27.
  7. Chung, Clive Yik-Sham; Shin, Hijai R.; Berdan, Charles A.; Ford, Breanna; Ward, Carl C.; Olzmann, James A.; Zoncu, Roberto; Nomura, Daniel K. (August 2019). "Covalent targeting of the vacuolar H+-ATPase activates autophagy via mTORC1 inhibition". Nature Chemical Biology. 15 (8): 776–785. doi:10.1038/s41589-019-0308-4. ISSN   1552-4469. PMC   6641988 . PMID   31285595.
  8. Isobe, Yosuke; Okumura, Mikiko; McGregor, Lynn M.; Brittain, Scott M.; Jones, Michael D.; Liang, Xiaoyou; White, Ross; Forrester, William; McKenna, Jeffrey M.; Tallarico, John A.; Schirle, Markus (2020-06-22). "Manumycin polyketides act as molecular glues between UBR7 and P53". Nature Chemical Biology. 16 (11): 1189–1198. doi:10.1038/s41589-020-0557-2. ISSN   1552-4469. PMC   7572527 . PMID   32572277. S2CID   219976949.
  9. Boike, Lydia; Cioffi, Alexander G.; Majewski, Felix C.; Co, Jennifer; Henning, Nathaniel J.; Jones, Michael D.; Liu, Gang; McKenna, Jeffrey M.; Tallarico, John A.; Schirle, Markus; Nomura, Daniel K. (2021-01-21). "Discovery of a Functional Covalent Ligand Targeting an Intrinsically Disordered Cysteine within MYC". Cell Chemical Biology. 28 (1): 4–13.e17. doi: 10.1016/j.chembiol.2020.09.001 . ISSN   2451-9456. PMC   7854864 . PMID   32966806. S2CID   221888024.
  10. Spradlin, Jessica N.; Hu, Xirui; Ward, Carl C.; Brittain, Scott M.; Jones, Michael D.; Ou, Lisha; To, Milton; Proudfoot, Andrew; Ornelas, Elizabeth; Woldegiorgis, Mikias; Olzmann, James A. (July 2019). "Harnessing the anti-cancer natural product nimbolide for targeted protein degradation". Nature Chemical Biology. 15 (7): 747–755. doi:10.1038/s41589-019-0304-8. ISSN   1552-4469. PMC   6592714 . PMID   31209351.
  11. Ward, Carl C.; Kleinman, Jordan I.; Brittain, Scott M.; Lee, Patrick S.; Chung, Clive Yik Sham; Kim, Kenneth; Petri, Yana; Thomas, Jason R.; Tallarico, John A.; McKenna, Jeffrey M.; Schirle, Markus; Nomura, Daniel K. (2019-11-15). "Covalent Ligand Screening Uncovers a RNF4 E3 Ligase Recruiter for Targeted Protein Degradation Applications". ACS Chemical Biology. 14 (11): 2430–2440. doi:10.1021/acschembio.8b01083. ISSN   1554-8929. PMC   7422721 . PMID   31059647.
  12. Henning, Nathaniel J.; Manford, Andrew G.; Spradlin, Jessica N.; Brittain, Scott M.; Zhang, Erika; McKenna, Jeffrey M.; Tallarico, John A.; Schirle, Markus; Rape, Michael; Nomura, Daniel K. (2022-01-19). "Discovery of a Covalent FEM1B Recruiter for Targeted Protein Degradation Applications". Journal of the American Chemical Society. 144 (2): 701–708. doi:10.1021/jacs.1c03980. ISSN   0002-7863. PMC   8928484 . PMID   34994556.
  13. Henning, Nathaniel J.; Boike, Lydia; Spradlin, Jessica N.; Ward, Carl C.; Liu, Gang; Zhang, Erika; Belcher, Bridget P.; Brittain, Scott M.; Hesse, Matthew J.; Dovala, Dustin; McGregor, Lynn M.; Valdez Misiolek, Rachel; Plasschaert, Lindsey W.; Rowlands, David J.; Wang, Feng (April 2022). "Deubiquitinase-targeting chimeras for targeted protein stabilization". Nature Chemical Biology. 18 (4): 412–421. doi: 10.1038/s41589-022-00971-2 . ISSN   1552-4469. PMC   10125259 . PMID   35210618. S2CID   233745282.
  14. Toriki, Ethan S.; Papatzimas, James W.; Nishikawa, Kaila; Dovala, Dustin; McGregor, Lynn M.; Hesse, Matthew J.; McKenna, Jeffrey M.; Tallarico, John A.; Schirle, Markus; Nomura, Daniel K. (2022-11-04). "Rational Chemical Design of Molecular Glue Degraders": 2022.11.04.512693. doi:10.1101/2022.11.04.512693. S2CID   253371370.{{cite journal}}: Cite journal requires |journal= (help)
  15. "Novartis, UC Berkeley join forces for 'undruggable' targets". Fierce Biotech. 28 September 2017. Retrieved 2020-08-27.
  16. Staff, Hyunkyu Michael Lee | (2017-10-02). "UC Berkeley researchers partners with Novartis to develop new cures". The Daily Californian. Retrieved 2020-08-28.
  17. "C&EN's 2019 10 Start-Ups to Watch". Chemical & Engineering News. Retrieved 2020-08-27.
  18. "Vicinitas Therapeutics Launches With $65 Million in Series A Financing to Advance Precision Medicines to Stabilize Key Proteins". Bloomberg.com. 2022-07-28. Retrieved 2022-11-21.
  19. "Daniel Nomura, PhD". 26 January 2021.
  20. "Daniel Nomura receives ASPIRE Award from the Mark Foundation | College of Chemistry".
  21. "NCI Outstanding Investigator Award Recipients - NCI". www.cancer.gov. 2022-11-02. Retrieved 2022-12-04.