13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Small molecule PROTACs: an emerging technology for targeted therapy in drug discovery

      review-article
      , , ,
      RSC Advances
      The Royal Society of Chemistry

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Curing malignant carcinomas is a grand ambition in the development of human health. Over the past decades, targeted therapies have become one of the most successful ways of achieving this. Of these approaches, small molecule inhibitors and monoclonal antibodies are two major methods, however several barriers to their development and clinical use still exist. The use of proteolysis-targeting chimeras (PROTACs) is a new technology through utilizing a intracellular ubiquitin-proteasome system to induce targeted protein degradation, is receiving much attention in the field of targeted therapies. Hetero-bifunctional PROTACs have the potential to eliminate the “undruggable” proteome that comprises about 85% of human proteins, which indicates their great prospects in therapeutic fields. However, there are some hurdles preventing current PROTACs moving from bench to clinic, such as delivery and bioavailability. This review provides an overview of the development of PROTAC technology and will briefly summarize the future possible directions of this approach.

          Abstract

          An overview of the latest developments in PROTAC technology and the possible directions of this approach is presented.

          Related collections

          Most cited references67

          • Record: found
          • Abstract: found
          • Article: not found

          Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation.

          The intracellular levels of many proteins are regulated by ubiquitin-dependent proteolysis. One of the best-characterized enzymes that catalyzes the attachment of ubiquitin to proteins is a ubiquitin ligase complex, Skp1-Cullin-F box complex containing Hrt1 (SCF). We sought to artificially target a protein to the SCF complex for ubiquitination and degradation. To this end, we tested methionine aminopeptidase-2 (MetAP-2), which covalently binds the angiogenesis inhibitor ovalicin. A chimeric compound, protein-targeting chimeric molecule 1 (Protac-1), was synthesized to recruit MetAP-2 to SCF. One domain of Protac-1 contains the I kappa B alpha phosphopeptide that is recognized by the F-box protein beta-TRCP, whereas the other domain is composed of ovalicin. We show that MetAP-2 can be tethered to SCF(beta-TRCP), ubiquitinated, and degraded in a Protac-1-dependent manner. In the future, this approach may be useful for conditional inactivation of proteins, and for targeting disease-causing proteins for destruction.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Induced protein degradation: an emerging drug discovery paradigm

            Small-molecule drug discovery has traditionally focused on occupancy of a binding site that directly affects protein function. This article discusses emerging technologies, such as proteolysis-targeting chimaeras (PROTACs), that exploit cellular quality control machinery to selectively degrade target proteins, which could have advantages over traditional approaches, including the potential to target proteins that are not currently therapeutically tractable.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Catalytic in vivo protein knockdown by small-molecule PROTACs.

              The current predominant therapeutic paradigm is based on maximizing drug-receptor occupancy to achieve clinical benefit. This strategy, however, generally requires excessive drug concentrations to ensure sufficient occupancy, often leading to adverse side effects. Here, we describe major improvements to the proteolysis targeting chimeras (PROTACs) method, a chemical knockdown strategy in which a heterobifunctional molecule recruits a specific protein target to an E3 ubiquitin ligase, resulting in the target's ubiquitination and degradation. These compounds behave catalytically in their ability to induce the ubiquitination of super-stoichiometric quantities of proteins, providing efficacy that is not limited by equilibrium occupancy. We present two PROTACs that are capable of specifically reducing protein levels by >90% at nanomolar concentrations. In addition, mouse studies indicate that they provide broad tissue distribution and knockdown of the targeted protein in tumor xenografts. Together, these data demonstrate a protein knockdown system combining many of the favorable properties of small-molecule agents with the potent protein knockdown of RNAi and CRISPR.
                Bookmark

                Author and article information

                Journal
                RSC Adv
                RSC Adv
                RA
                RSCACL
                RSC Advances
                The Royal Society of Chemistry
                2046-2069
                30 May 2019
                29 May 2019
                30 May 2019
                : 9
                : 30
                : 16967-16976
                Affiliations
                [a] Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University Shanghai 200241 China yhchen@ 123456bio.ecnu.edu.cn
                [b] School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China qhzhao@ 123456chem.ecnu.edu.cn
                Author information
                https://orcid.org/0000-0003-4031-447X
                https://orcid.org/0000-0003-1733-7980
                Article
                c9ra03423d
                10.1039/c9ra03423d
                9064693
                35519875
                85d4ec60-0e71-423f-b8b6-76b1721a148e
                This journal is © The Royal Society of Chemistry
                History
                : 7 May 2019
                : 14 May 2019
                Page count
                Pages: 10
                Funding
                Funded by: National Natural Science Foundation of China, doi 10.13039/501100001809;
                Award ID: 81673304
                Funded by: Shanghai Municipal Education Commission, doi 10.13039/501100003395;
                Award ID: 2017-01-07-00-05-E00011
                Funded by: National Basic Research Program of China (973 Program), doi 10.13039/501100012166;
                Award ID: 2018YFA0507001
                Categories
                Chemistry
                Custom metadata
                Paginated Article

                Comments

                Comment on this article