1
|
Pandey D, Chauhan SC, Kashyap VK, Roy KK. Structural insights into small-molecule KRAS inhibitors for targeting KRAS mutant cancers. Eur J Med Chem 2024; 277:116771. [PMID: 39167893 DOI: 10.1016/j.ejmech.2024.116771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
The Kirsten rat sarcoma viral (KRAS) oncogene is the most frequently mutated isoform of RAS, associated with 85 % of RAS-driven cancers. KRAS functions as a signaling hub, participating in various cellular signaling pathways and regulating a wide range of important activities, including cell proliferation, differentiation, growth, metabolism, and migration. Despite being the most frequently altered oncogenic protein in solid tumors, over the past four decades, KRAS has historically been considered "undruggable" owing to a lack of pharmacologically targetable pockets within the mutant isoforms. However, improvements in drug design and development have culminated in the development of selective inhibitors for KRAS mutants. Recent developments have led to the successful targeting of the KRASG12C mutant through covalent inhibitors that exploit the unique cysteine residue introduced by the mutation at 12th position. These inhibitors bind covalently to C12, locking KRAS in its inactive GDP-bound state and preventing downstream signaling. Some of these inhibitors have shown encouraging results in KRASG12C mutant cancer patients but suffer from drug resistance, toxicity, and low therapeutic efficacy. Recently, there have been great advancements in the discovery of drugs that directly target the switch I (S-I), switch-II (S-II) and S-I/II interface sites of KRAS mutant proteins. These include KRASG12C inhibitors like AMG510 (Sotorasib) and MRTX849 (Adagrasib), which have got FDA approval for non-small cell lung cancer harboring the KRASG12C mutation. There is no approved drug for cancers harboring other KRAS mutations, although efforts have expanded to target other KRAS mutations and the Switch I/II interface, aiming to disrupt KRAS-driven oncogenic signaling. Structure-activity relationship (SAR) studies have been instrumental in optimizing the binding affinity, selectivity, and pharmacokinetic properties of these inhibitors, leading to the development of promising therapeutic agents like Sotorasib and Adagrasib. This review provides an overview of the KRAS pathway, KRAS binding sites, strategies for direct and indirect inhibition using small molecules, and SAR based on the co-crystal structures of inhibitors with KRAS mutants which is expected to offer new hope for patients with KRAS-driven cancers through the development of new KRAS-targeted drugs.
Collapse
Affiliation(s)
- Divya Pandey
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India
| | - Subhash C Chauhan
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| | - Vivek K Kashyap
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| | - Kuldeep K Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India.
| |
Collapse
|
2
|
Ghufran M, Rehman AU, Ayaz M, Ul-Haq Z, Uddin R, Azam SS, Wadood A. New lead compounds identification against KRas mediated cancers through pharmacophore-based virtual screening and in vitro assays. J Biomol Struct Dyn 2023; 41:8053-8067. [PMID: 36184737 DOI: 10.1080/07391102.2022.2128878] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2022] [Indexed: 10/07/2022]
Abstract
Cancer remains the leading cause of mortality and morbidity in the world, with 19.3 million new diagnoses and 10.1 million deaths in 2020. Cancer is caused due to mutations in proto-oncogenes and tumor-suppressor genes. Genetic analyses found that Ras (Rat sarcoma) is one of the most deregulated oncogenes in human cancers. The Ras oncogene family members including NRas (Neuroblastoma ras viral oncogene homolog), HRas (Harvey rat sarcoma) and KRas are involved in different types of human cancers. The mutant KRas is considered as the most frequent oncogene implicated in the development of lung, pancreatic and colon cancers. However, there is no efficient clinical drug even though it has been identified as an oncogene for 30 years. Therefore there is an emerging need to develop potent, new anticancer drugs. In this study, computer-aided drug designing approaches as well as experimental methods were employed to find new and potential anti-cancer drugs. The pharmacophore model was developed from an already known FDA approved anti-cancer drug Bortezomib using the software MOE. The validated pharmacophore model was then used to screen the in-house and commercially available databases. The pharmacophore-based virtual screening resulted in 26 and 86 hits from in-house and commercial databases respectively. Finally, 6/13 (in-house database) and 24/64 hits (commercial databases) were selected with different scaffolds having good interactions with the significant active residues of KRasG12D protein that were predicted as potent lead compounds. Finally, the results of pharmacophore-based virtual screening were further validated by molecular dynamics simulation analysis. The 6 hits of the in-house database were further evaluated experimentally. The experimental results showed that these compounds have good anti-cancer activity which validate the protocol of our in silico studies. KRasG12D protein is a very important anti-cancer target and potent inhibitors for this target are still not available, so small lead compound inhibitors were identified to inhibit the activity of this protein by blocking the GTP-binding pocket.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mehreen Ghufran
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
- Department of Pathology, Medical Teaching Institution Bacha Khan Medical College (BKMC) Mardan, Mardan, Pakistan
| | - Ashfaq Ur Rehman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, University of Karachi, Karachi, Pakistan
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, University of Karachi, Karachi, Pakistan
| | - Syed Sikander Azam
- Department of Bioinformatics, Quaid-e-Azam University, Islamabad, Pakistan
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| |
Collapse
|
3
|
Yang A, Li M, Fang M. The Research Progress of Direct KRAS G12C Mutation Inhibitors. Pathol Oncol Res 2021; 27:631095. [PMID: 34257597 PMCID: PMC8262225 DOI: 10.3389/pore.2021.631095] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/01/2021] [Indexed: 11/26/2022]
Abstract
Abstract: KRAS mutations have long been considered undruggable. However, a series of direct KRAS mutation inhibitors have been developed since the switch II pocket was discovered recently. This review will summarize progress in the development of direct KRAS G12C mutation inhibitors, current relevant drugs under study and challenges that need to be considered in future research.
Collapse
Affiliation(s)
- Ai Yang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingzhi Fang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Asl ER, Amini M, Najafi S, Mansoori B, Mokhtarzadeh A, Mohammadi A, Lotfinejad P, Bagheri M, Shirjang S, Lotfi Z, Rasmi Y, Baradaran B. Interplay between MAPK/ERK signaling pathway and MicroRNAs: A crucial mechanism regulating cancer cell metabolism and tumor progression. Life Sci 2021; 278:119499. [PMID: 33865878 DOI: 10.1016/j.lfs.2021.119499] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/29/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Mitogen-activated protein kinase (MAPK) signal transduction, as a highly conserved signaling pathway, is reported to be involved in various biological events, including metabolic reprogramming, cell proliferation, survival, and differentiation. Mutations in key molecules involved in MAPK/ERK signaling and dysregulation of this pathway are very common events in various human malignancies, which make the MAPK signaling a crucial signaling pathway participating in the regulation of glucose uptake by malignant cells and tumorigenesis. MicroRNAs (miRNAs), as small non-coding RNAs, are critical regulators of gene expression that play key roles in cancer initiation and progression. On the other hand, these small RNAs mutually regulate the MAPK signaling which is often overexpressed in the case of cancer progression; suggesting that crosstalk between miRNAs and this signaling pathway plays a pivotal role in the development of human cancers. Some miRNAs such as miR-20b, miR-34c-3p, miR-152, miR-181a, and miR-302b through inhibiting MAPK signaling, and miR-193a-3p, miR-330-3p, and miR-592 by activating this signaling pathway, play imperative roles in tumorigenesis. Therefore, in this review, we aimed to focus on the interplay between miRNAs and MAPK signaling in the various steps of tumorigenesis, including metabolic regulation, cell proliferation, apoptosis, metastasis, angiogenesis, and drug resistance.
Collapse
Affiliation(s)
- Elmira Roshani Asl
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Parisa Lotfinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences
| | - Mehdi Bagheri
- Department of Biology, Khorasan Razavi Science and Research Branch, Islamic Azad University, Neyshabur, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ziba Lotfi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.; Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran..
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran..
| |
Collapse
|
5
|
Gray JL, von Delft F, Brennan PE. Targeting the Small GTPase Superfamily through Their Regulatory Proteins. Angew Chem Int Ed Engl 2020; 59:6342-6366. [PMID: 30869179 PMCID: PMC7204875 DOI: 10.1002/anie.201900585] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/11/2019] [Indexed: 12/11/2022]
Abstract
The Ras superfamily of small GTPases are guanine-nucleotide-dependent switches essential for numerous cellular processes. Mutations or dysregulation of these proteins are associated with many diseases, but unsuccessful attempts to target the small GTPases directly have resulted in them being classed as "undruggable". The GTP-dependent signaling of these proteins is controlled by their regulators; guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in the Rho and Rab subfamilies, guanine nucleotide dissociation inhibitors (GDIs). This review covers the recent small molecule and biologics strategies to target the small GTPases through their regulators. It seeks to critically re-evaluate recent chemical biology practice, such as the presence of PAINs motifs and the cell-based readout using compounds that are weakly potent or of unknown specificity. It highlights the vast scope of potential approaches for targeting the small GTPases in the future through their regulatory proteins.
Collapse
Affiliation(s)
- Janine L. Gray
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOld Road CampusOxfordOX3 7FZUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotOX11 0QXUK
| | - Frank von Delft
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotOX11 0QXUK
- Department of BiochemistryUniversity of JohannesburgAuckland Park2006South Africa
| | - Paul E. Brennan
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOld Road CampusOxfordOX3 7FZUK
- Alzheimer's Research (UK) Oxford Drug Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| |
Collapse
|
6
|
Liu Z, Wang M, Wang H, Fang L, Gou S. Targeting RAS-RAF pathway significantly improves antitumor activity of Rigosertib-derived platinum(IV) complexes and overcomes cisplatin resistance. Eur J Med Chem 2020; 194:112269. [PMID: 32248002 DOI: 10.1016/j.ejmech.2020.112269] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/22/2020] [Accepted: 03/22/2020] [Indexed: 02/07/2023]
Abstract
RAS-RAF pathway presents a valuable target for the cancer treatment due to its important roles in the regulation of tumor proliferation, apoptosis and the obtained resistance. To explore such target a RAS/CRAF interference agent, was therefore conjugated with Pt(IV) prodrugs via ester bond, resulting in total eleven multifunctional Pt(IV) complexes. The complexes could target genomic DNA and disrupt the signaling transduction from RAS protein to CRAF so that block the mitogen-activated protein kinase (MAPK) signaling pathway. Experiments in vitro indicated that all of the Pt(IV) complexes showed potent anti-tumor activity with IC50 values ranged from 8 nM to 22.55 μM, which were significantly improved as compared with cisplatin (CDDP) whose IC50 values ranged from 5.45 μM to 9.05 μM. Among them, 26 exerted the best anti-tumor activity in vitro, which not only exhibited excellent cytotoxicity against normal tumor cells, but also against CDDP-resistance cell lines (e.g. A549/CDDP and SKOV-3/CDDP). Importantly, 26 only showed little effect on normal cell lines such as HUEVC and LO2. Besides, the following biological mechanisms studies demonstrated that 26 could efficiently enter. A549 cells, significantly arrest cell cycle at G2/M phase, disrupt the signaling pathway and trigger endogenous caspase apoptosis pathway. Furthermore, results of a xenograft subcutaneous model of A549 tumor showed that 26 could effectively decrease tumor growth rates without causing loss of bodyweight.
Collapse
Affiliation(s)
- Zhikun Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Meng Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China
| | - Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
7
|
Gray JL, Delft F, Brennan PE. Targeting der kleinen GTPasen über ihre regulatorischen Proteine. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201900585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Janine L. Gray
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Target Discovery InstituteNuffield Department of MedicineUniversity of Oxford Old Road Campus Oxford OX3 7FZ Großbritannien
- Diamond Light Source Harwell Science and Innovation Campus Didcot OX11 0QX Großbritannien
| | - Frank Delft
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Diamond Light Source Harwell Science and Innovation Campus Didcot OX11 0QX Großbritannien
- Department of BiochemistryUniversity of Johannesburg Auckland Park 2006 Südafrika
| | - Paul E. Brennan
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Target Discovery InstituteNuffield Department of MedicineUniversity of Oxford Old Road Campus Oxford OX3 7FZ Großbritannien
- Alzheimer's Research (UK) Oxford Drug Discovery InstituteNuffield Department of MedicineUniversity of Oxford Oxford OX3 7FZ Großbritannien
| |
Collapse
|
8
|
Zhang Z, Shokat KM. Bifunctional Small-Molecule Ligands of K-Ras Induce Its Association with Immunophilin Proteins. Angew Chem Int Ed Engl 2019; 58:16314-16319. [PMID: 31557383 PMCID: PMC6980260 DOI: 10.1002/anie.201910124] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 12/20/2022]
Abstract
Here we report the design, synthesis, and characterization of bifunctional chemical ligands that induce the association of Ras with ubiquitously expressed immunophilin proteins such as FKBP12 and cyclophilin A. We show this approach is applicable to two distinct Ras ligand scaffolds, and that both the identity of the immunophilin ligand and the linker chemistry affect compound efficacy in biochemical and cellular contexts. These ligands bind to Ras in an immunophilin-dependent fashion and mediate the formation of tripartite complexes of Ras, immunophilin, and the ligand. The recruitment of cyclophilin A to GTP-bound Ras blocks its interaction with B-Raf in biochemical assays. Our study demonstrates the feasibility of ligand-induced association of Ras with intracellular proteins and suggests it as a promising therapeutic strategy for Ras-driven cancers.
Collapse
Affiliation(s)
- Ziyang Zhang
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA, 94143, USA
| | - Kevan M Shokat
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA, 94143, USA
| |
Collapse
|
9
|
Zhang Z, Shokat KM. Bifunctional Small‐Molecule Ligands of K‐Ras Induce Its Association with Immunophilin Proteins. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201910124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ziyang Zhang
- Cellular and Molecular PharmacologyHoward Hughes Medical InstituteUniversity of California, San Francisco 600 16th Street San Francisco CA 94143 USA
| | - Kevan M. Shokat
- Cellular and Molecular PharmacologyHoward Hughes Medical InstituteUniversity of California, San Francisco 600 16th Street San Francisco CA 94143 USA
| |
Collapse
|
10
|
Tsubamoto M, Le TK, Li M, Watanabe T, Matsumi C, Parvatkar P, Fujii H, Kato N, Sun J, Ohkanda J. A Guanidyl-Based Bivalent Peptidomimetic Inhibits K-Ras Prenylation and Association with c-Raf. Chemistry 2019; 25:13531-13536. [PMID: 31393030 DOI: 10.1002/chem.201903129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/07/2019] [Indexed: 01/06/2023]
Abstract
Unusual lipid modification of K-Ras makes Ras-directed cancer therapy a challenging task. Aiming to disrupt electrostatic-driven protein-protein interactions (PPIs) of K-Ras with FTase and GGTase I, a series of bivalent dual inhibitors that recognize the active pocket and the common acidic surface of FTase and GGTase I were designed. The structure-activity-relationship study resulted in 8 b, in which a biphenyl-based peptidomimetic FTI-277 was attached to a guanidyl-containing gallate moiety through an alkyl linker. Cell-based evaluation demonstrated that 8 b exhibited substantial inhibition of K-Ras processing without apparent interference with Rap-1A processing. Fluorescent imaging showed that 8 b disrupts localization of K-Ras to the plasma membrane and impairs interaction with c-Raf, whereas only FTI-277 was found to be inactive. These results suggest that targeting the PPI interface of K-Ras may provide an alternative method of inhibiting K-Ras.
Collapse
Affiliation(s)
- Mai Tsubamoto
- The Institute of Scientific Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Toan Khanh Le
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan
| | - Minghua Li
- Department of Pharmaceutical Science, University of South Florida, Tampa, Florida, 33612, USA
| | - Taku Watanabe
- Ina Laboratory, Medical & Biological Laboratories, CO., Ltd., Ina, Nagano, 396-0002, Japan
| | - Chiemi Matsumi
- Ina Laboratory, Medical & Biological Laboratories, CO., Ltd., Ina, Nagano, 396-0002, Japan
| | - Prakash Parvatkar
- Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Hiroshi Fujii
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan
| | - Nobuo Kato
- The Institute of Scientific Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Jiazhi Sun
- Department of Pharmaceutical Science, University of South Florida, Tampa, Florida, 33612, USA
| | - Junko Ohkanda
- Academic Assembly, Institute of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan
| |
Collapse
|
11
|
Tolpygin AO, Cherkasov AV, Fukin GK, Kovylina TA, Lyssenko KA, Trifonov AA. Calcium Amido Complexes Coordinated by Tridentate Amidinate Ligands: Synthesis, Structures and Catalytic Activity in Olefin Hydrophosphination and Polymerization of Cyclic Esters. Eur J Inorg Chem 2019. [DOI: 10.1002/ejic.201900610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Aleksei O. Tolpygin
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences Tropinina str. 49 603950 Nizhny Novgorod, GSP‐445 Russia
| | - Anton V. Cherkasov
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences Tropinina str. 49 603950 Nizhny Novgorod, GSP‐445 Russia
| | - Georgy K. Fukin
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences Tropinina str. 49 603950 Nizhny Novgorod, GSP‐445 Russia
| | - Tatyana A. Kovylina
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences Tropinina str. 49 603950 Nizhny Novgorod, GSP‐445 Russia
| | - Konstantin A. Lyssenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences Vavilova str. 28 119991 Moscow, GSP‐1 Russia
| | - Alexander A. Trifonov
- G. A. Razuvaev Institute of Organometallic Chemistry of Russian Academy of Sciences Tropinina str. 49 603950 Nizhny Novgorod, GSP‐445 Russia
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences Vavilova str. 28 119991 Moscow, GSP‐1 Russia
| |
Collapse
|
12
|
Affiliation(s)
- Jie Wang
- Shanghai Key Laboratory of New Drug Design, School of PharmacyEast China University of Science and Technology Shanghai 200237 China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of PharmacyEast China University of Science and Technology Shanghai 200237 China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of PharmacyEast China University of Science and Technology Shanghai 200237 China
| |
Collapse
|
13
|
Ghosh AK, Samanta I, Mondal A, Liu WR. Covalent Inhibition in Drug Discovery. ChemMedChem 2019; 14:889-906. [PMID: 30816012 DOI: 10.1002/cmdc.201900107] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Indexed: 12/11/2022]
Abstract
Although covalent inhibitors have been used as therapeutics for more than a century, there has been general resistance in the pharmaceutical industry against their further development due to safety concerns. This inclination has recently been reverted after the development of a wide variety of covalent inhibitors to address human health conditions along with the US Food and Drug Administration (FDA) approval of several covalent therapeutics for use in humans. Along with this exciting resurrection of an old drug discovery concept, this review surveys enzymes that can be targeted by covalent inhibitors for the treatment of human diseases. We focus on protein kinases, RAS proteins, and a few other enzymes that have been studied extensively as targets for covalent inhibition, with the aim to address challenges in designing effective covalent drugs and to provide suggestions in the area that have yet to be explored.
Collapse
Affiliation(s)
- Avick Kumar Ghosh
- Department of Chemistry, Texas A&M University, Corner of Ross and Spence Streets, College Station, TX, 77843, USA
| | - Indranil Samanta
- Department of Chemistry, Texas A&M University, Corner of Ross and Spence Streets, College Station, TX, 77843, USA
| | - Anushree Mondal
- Department of Chemistry, Texas A&M University, Corner of Ross and Spence Streets, College Station, TX, 77843, USA
| | - Wenshe Ray Liu
- Department of Chemistry, Texas A&M University, Corner of Ross and Spence Streets, College Station, TX, 77843, USA
| |
Collapse
|
14
|
Murarka S, Martín-Gago P, Schultz-Fademrecht C, Al Saabi A, Baumann M, Fansa EK, Ismail S, Nussbaumer P, Wittinghofer A, Waldmann H. Development of Pyridazinone Chemotypes Targeting the PDEδ Prenyl Binding Site. Chemistry 2017; 23:6083-6093. [PMID: 27809361 DOI: 10.1002/chem.201603222] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Indexed: 12/18/2022]
Abstract
The K-Ras GTPase is a major target in anticancer drug discovery. However, direct interference with signaling by K-Ras has not led to clinically useful drugs yet. Correct localization and signaling by farnesylated K-Ras is regulated by the prenyl binding protein PDEδ. Interfering with binding of PDEδ to K-Ras by means of small molecules provides a novel opportunity to suppress oncogenic signaling. Here we describe the identification and structure-guided development of novel K-Ras-PDEδ inhibitor chemotypes based on pyrrolopyridazinones and pyrazolopyridazinones that bind to the farnesyl binding pocket of PDEδ with low nanomolar affinity. We delineate the structure-property relationship and in vivo pharmacokinetic (PK) and toxicokinetic (Tox) studies for pyrazolopyridazinone-based K-Ras-PDEδ inhibitors. These findings may inspire novel drug discovery efforts aimed at the development of drugs targeting oncogenic Ras.
Collapse
Affiliation(s)
- Sandip Murarka
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Pablo Martín-Gago
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | | | - Alaa Al Saabi
- Lead Discovery Center GmbH, 44227, Dortmund, Germany
| | | | - Eyad K Fansa
- Max-Planck-Institut für Molekulare Physiologie, Structural Biology Group, 44227, Dortmund, Germany
| | - Shehab Ismail
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | | | - Alfred Wittinghofer
- Max-Planck-Institut für Molekulare Physiologie, Structural Biology Group, 44227, Dortmund, Germany
| | - Herbert Waldmann
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie, Chemische Biologie, Otto-Hahn-Straße 6, 44221, Dortmund, Germany
| |
Collapse
|
15
|
Martín-Gago P, Fansa EK, Klein CH, Murarka S, Janning P, Schürmann M, Metz M, Ismail S, Schultz-Fademrecht C, Baumann M, Bastiaens PIH, Wittinghofer A, Waldmann H. A PDE6δ-KRas Inhibitor Chemotype with up to Seven H-Bonds and Picomolar Affinity that Prevents Efficient Inhibitor Release by Arl2. Angew Chem Int Ed Engl 2017; 56:2423-2428. [PMID: 28106325 DOI: 10.1002/anie.201610957] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Indexed: 12/11/2022]
Abstract
Small-molecule inhibition of the interaction between the KRas oncoprotein and the chaperone PDE6δ impairs KRas spatial organization and signaling in cells. However, despite potent binding in vitro (KD <10 nm), interference with Ras signaling and growth inhibition require 5-20 μm compound concentrations. We demonstrate that these findings can be explained by fast release of high-affinity inhibitors from PDE6δ by the release factor Arl2. This limitation is overcome by novel highly selective inhibitors that bind to PDE6δ with up to 7 hydrogen bonds, resulting in picomolar affinity. Their release by Arl2 is greatly decreased, and representative compounds selectively inhibit growth of KRas mutated and -dependent cells with the highest activity recorded yet. Our findings indicate that very potent inhibitors of the KRas-PDE6δ interaction may impair the growth of tumors driven by oncogenic KRas.
Collapse
Affiliation(s)
- Pablo Martín-Gago
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Eyad K Fansa
- Structural Biology Group, Max Planck Institute for Molecular Physiology, 44227, Dortmund, Germany
| | - Christian H Klein
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Sandip Murarka
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Petra Janning
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Marc Schürmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Malte Metz
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Shehab Ismail
- Structural Biology Group, Max Planck Institute for Molecular Physiology, 44227, Dortmund, Germany
| | | | | | - Philippe I H Bastiaens
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
- TU Dortmund, Faculty of Chemistry and Chemical Biology, 44227, Dortmund, Germany
| | - Alfred Wittinghofer
- Structural Biology Group, Max Planck Institute for Molecular Physiology, 44227, Dortmund, Germany
| | - Herbert Waldmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
- TU Dortmund, Faculty of Chemistry and Chemical Biology, 44227, Dortmund, Germany
| |
Collapse
|
16
|
Martín-Gago P, Fansa EK, Klein CH, Murarka S, Janning P, Schürmann M, Metz M, Ismail S, Schultz-Fademrecht C, Baumann M, Bastiaens PIH, Wittinghofer A, Waldmann H. A PDE6δ-KRas Inhibitor Chemotype with up to Seven H-Bonds and Picomolar Affinity that Prevents Efficient Inhibitor Release by Arl2. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201610957] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Pablo Martín-Gago
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
| | - Eyad K. Fansa
- Structural Biology Group; Max Planck Institute for Molecular Physiology; 44227 Dortmund Germany
| | - Christian H. Klein
- Department of Systemic Cell Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
| | - Sandip Murarka
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
| | - Petra Janning
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
| | - Marc Schürmann
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
| | - Malte Metz
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
| | - Shehab Ismail
- Structural Biology Group; Max Planck Institute for Molecular Physiology; 44227 Dortmund Germany
| | | | | | - Philippe I. H. Bastiaens
- Department of Systemic Cell Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
- TU Dortmund; Faculty of Chemistry and Chemical Biology; 44227 Dortmund Germany
| | - Alfred Wittinghofer
- Structural Biology Group; Max Planck Institute for Molecular Physiology; 44227 Dortmund Germany
| | - Herbert Waldmann
- Department of Chemical Biology; Max Planck Institute of Molecular Physiology; 44227 Dortmund Germany
- TU Dortmund; Faculty of Chemistry and Chemical Biology; 44227 Dortmund Germany
| |
Collapse
|
17
|
Marín-Ramos NI, Piñar C, Vázquez-Villa H, Martín-Fontecha M, González Á, Canales Á, Algar S, Mayo PP, Jiménez-Barbero J, Gajate C, Mollinedo F, Pardo L, Ortega-Gutiérrez S, Viso A, López-Rodríguez ML. Development of a Nucleotide Exchange Inhibitor That Impairs Ras Oncogenic Signaling. Chemistry 2016; 23:1676-1685. [PMID: 27885731 DOI: 10.1002/chem.201604905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Indexed: 12/11/2022]
Abstract
Despite more than three decades of intense effort, no anti-Ras therapies have reached clinical application. Contributing to this failure has been an underestimation of Ras complexity and a dearth of structural information. In this regard, recent studies have revealed the highly dynamic character of the Ras surface and the existence of transient pockets suitable for small-molecule binding, opening up new possibilities for the development of Ras modulators. Herein, a novel Ras inhibitor (compound 12) is described that selectively impairs mutated Ras activity in a reversible manner without significantly affecting wild-type Ras, reduces the Ras-guanosine triphosphate (GTP) levels, inhibits the activation of the mitogen-activated protein kinase (MAPK) pathway, and exhibits remarkable cytotoxic activity in Ras-driven cellular models. The use of molecular dynamics simulations and NMR spectroscopy experiments has enabled the molecular bases responsible for the interactions between compound 12 and Ras protein to be explored. The new Ras inhibitor binds partially to the GTP-binding region and extends into the adjacent hydrophobic pocket delimited by switch II. Hence, Ras inhibitor 12 could represent a new compound for the development of more efficacious drugs to target Ras-driven cancers; a currently unmet clinical need.
Collapse
Affiliation(s)
- Nagore I Marín-Ramos
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain.,CEI Campus Moncloa, UCM-UPM and CSIC, 28040, Madrid, Spain
| | - Carmen Piñar
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Henar Vázquez-Villa
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Mar Martín-Fontecha
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Ángel González
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Ángeles Canales
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Sergio Algar
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Paloma P Mayo
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Edif. 801A, 48160, Derio, Spain.,Ikerbasque, Basque Foundation for Science, 48103, Bilbao, Spain
| | - Consuelo Gajate
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Faustino Mollinedo
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Leonardo Pardo
- Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain
| | - Silvia Ortega-Gutiérrez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Alma Viso
- Instituto de Química Orgánica General, IQOG-CSIC, Juan de la Cierva, 3, 28006, Madrid, Spain
| | - María L López-Rodríguez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| |
Collapse
|
18
|
Mao Y, Yao H, Wang H, Cheng P, Long D. Microsecond Timescale Dynamics of GDP-Bound Ras Underlies the Formation of Novel Inhibitor-Binding Pockets. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201608653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yunyun Mao
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Haijie Yao
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Hui Wang
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Peng Cheng
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Dong Long
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
- Hefei National Laboratory for Physical Sciences at the Microscale; University of Science and Technology of China, Hefei; Anhui 230027 China
| |
Collapse
|
19
|
Mao Y, Yao H, Wang H, Cheng P, Long D. Microsecond Timescale Dynamics of GDP-Bound Ras Underlies the Formation of Novel Inhibitor-Binding Pockets. Angew Chem Int Ed Engl 2016; 55:15629-15632. [DOI: 10.1002/anie.201608653] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 10/07/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Yunyun Mao
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Haijie Yao
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Hui Wang
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Peng Cheng
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
| | - Dong Long
- School of Life Sciences; University of Science and Technology of China; 443 Huangshan Street, Hefei Anhui 230027 China
- Hefei National Laboratory for Physical Sciences at the Microscale; University of Science and Technology of China, Hefei; Anhui 230027 China
| |
Collapse
|
20
|
Dubiella C, Baur R, Cui H, Huber EM, Groll M. Selektive Hemmung des Immunproteasoms durch strukturbasierte Wahl eines nichtkatalytischen Cysteins als Angriffspunkt. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201506631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Christian Dubiella
- Center for Integrated Protein Science Munich (CIPSM), Fakultät für Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching (Deutschland)
| | - Regina Baur
- Center for Integrated Protein Science Munich (CIPSM), Fakultät für Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching (Deutschland)
| | - Haissi Cui
- Center for Integrated Protein Science Munich (CIPSM), Fakultät für Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching (Deutschland)
| | - Eva M. Huber
- Center for Integrated Protein Science Munich (CIPSM), Fakultät für Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching (Deutschland)
| | - Michael Groll
- Center for Integrated Protein Science Munich (CIPSM), Fakultät für Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching (Deutschland)
| |
Collapse
|
21
|
Dubiella C, Baur R, Cui H, Huber EM, Groll M. Selective Inhibition of the Immunoproteasome by Structure-Based Targeting of a Non-catalytic Cysteine. Angew Chem Int Ed Engl 2015; 54:15888-91. [PMID: 26563572 DOI: 10.1002/anie.201506631] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/15/2015] [Indexed: 11/09/2022]
Abstract
Clinically applied proteasome inhibitors induce cell death by concomitant blockage of constitutive and immunoproteasomes. In contrast, selective immunoproteasome inhibition is less cytotoxic and has the potential to modulate chronic inflammation and autoimmune diseases. In this study, we rationally designed decarboxylated peptides that covalently target a non-catalytic cysteine of the immunoproteasome subunit β5i with α-chloroacetamide-containing sidechains. The enhanced isoform specificity decreased cytotoxic effects and the compound suppressed the production of inflammatory cytokines. Structure-based optimization led to over 150-fold selectivity for subunit β5i over β5c. This new compound class provides a promising starting point for the development of selective immunoproteasome inhibitors as potential anti-inflammatory agents.
Collapse
Affiliation(s)
- Christian Dubiella
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching (Germany).
| | - Regina Baur
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching (Germany)
| | - Haissi Cui
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching (Germany)
| | - Eva M Huber
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching (Germany)
| | - Michael Groll
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748 Garching (Germany).
| |
Collapse
|
22
|
Cromm PM, Spiegel J, Grossmann TN, Waldmann H. Direkte Modulation von Aktivität und Funktion kleiner GTPasen. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201504357] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
23
|
Cromm PM, Spiegel J, Grossmann TN, Waldmann H. Direct Modulation of Small GTPase Activity and Function. Angew Chem Int Ed Engl 2015; 54:13516-37. [DOI: 10.1002/anie.201504357] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Indexed: 12/19/2022]
|
24
|
Upadhyaya P, Qian Z, Selner NG, Clippinger SR, Wu Z, Briesewitz R, Pei D. Inhibition of Ras signaling by blocking Ras-effector interactions with cyclic peptides. Angew Chem Int Ed Engl 2015; 54:7602-6. [PMID: 25950772 PMCID: PMC4591930 DOI: 10.1002/anie.201502763] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Indexed: 12/31/2022]
Abstract
Ras genes are frequently activated in human cancers, but the mutant Ras proteins remain largely "undruggable" through the conventional small-molecule approach owing to the absence of any obvious binding pockets on their surfaces. By screening a combinatorial peptide library, followed by structure-activity relationship (SAR) analysis, we discovered a family of cyclic peptides possessing both Ras-binding and cell-penetrating properties. These cell-permeable cyclic peptides inhibit Ras signaling by binding to Ras-GTP and blocking its interaction with downstream proteins and they induce apoptosis of cancer cells. Our results demonstrate the feasibility of developing cyclic peptides for the inhibition of intracellular protein-protein interactions and of direct Ras inhibitors as a novel class of anticancer agents.
Collapse
Affiliation(s)
- Punit Upadhyaya
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210 (USA)
| | - Ziqing Qian
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210 (USA)
| | - Nicholas G Selner
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210 (USA)
| | - Sarah R Clippinger
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210 (USA)
| | - Zhengrong Wu
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210 (USA)
| | - Roger Briesewitz
- Department of Pharmacology, The Ohio State University, 5065 Graves Hall, 333 West 10th Avenue, Columbus, OH 43210 (USA).
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12th Avenue, Columbus, OH 43210 (USA).
| |
Collapse
|
25
|
Upadhyaya P, Qian Z, Selner NG, Clippinger SR, Wu Z, Briesewitz R, Pei D. Inhibition of Ras Signaling by Blocking Ras-Effector Interactions with Cyclic Peptides. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201502763] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
26
|
Rudolph J, Stokoe D. Selektive Inhibierung von mutiertem Ras-Protein durch kovalent bindende Liganden. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201400233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
Rudolph J, Stokoe D. Selective Inhibition of Mutant Ras Protein through Covalent Binding. Angew Chem Int Ed Engl 2014; 53:3777-9. [DOI: 10.1002/anie.201400233] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Indexed: 01/25/2023]
|