1
|
O'Gorman B, Cooke SF, Blair CM. Precision peptide disruptors: The next generation of targeted therapeutics in oncology. Cell Signal 2025; 132:111783. [PMID: 40180165 DOI: 10.1016/j.cellsig.2025.111783] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Therapeutically targeting the pathologically remodelled protein-protein interaction network in cancer with peptide disruptors increasingly represents a clinically attractive approach to treating recalcitrant cancers. In this review, we map the pre-clinical and clinical-stage peptide disruptor landscape within an oncology-specific context and discuss key clinical examples that are making significant impact to patients; demonstrating a key role for peptide disruptors in precision medicine as a next-generation targeted therapeutic.
Collapse
Affiliation(s)
- Bria O'Gorman
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Sean F Cooke
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Connor M Blair
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
2
|
Gare CL, White AM, Malins LR. From lead to market: chemical approaches to transform peptides into therapeutics. Trends Biochem Sci 2025; 50:467-480. [PMID: 40011178 DOI: 10.1016/j.tibs.2025.01.009] [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: 11/11/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/28/2025]
Abstract
Peptides are a powerful drug modality with potential to access difficult targets. This recognition underlies their growth in the global pharmaceutical market, with peptides representing ~8% of drugs approved by the FDA over the past decade. Currently, the peptide therapeutic landscape is evolving, with high-throughput display technologies driving the identification of peptide leads with enhanced diversity. Yet, chemical modifications remain essential for improving the 'drug-like' properties of peptides and ultimately translating leads to market. In this review, we explore two recent therapeutic candidates (semaglutide, a peptide hormone analogue, and MK-0616, an mRNA display-derived candidate) as case studies that highlight general approaches to improving pharmacokinetics (PK) and potency. We also emphasize the critical link between advances in medicinal chemistry and the optimisation of highly efficacious peptide therapeutics.
Collapse
Affiliation(s)
- Caitlin L Gare
- Research School of Chemistry, Australian National University, Canberra 2601, Australian Capital Territory, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra 2601, Australian Capital Territory, Australia
| | - Andrew M White
- Research School of Chemistry, Australian National University, Canberra 2601, Australian Capital Territory, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra 2601, Australian Capital Territory, Australia
| | - Lara R Malins
- Research School of Chemistry, Australian National University, Canberra 2601, Australian Capital Territory, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra 2601, Australian Capital Territory, Australia.
| |
Collapse
|
3
|
Zuo Z, Wang Y, Fang Y, Zhao M, Wang Z, Yang Z, Jia B, Sun Y. A novel regulator of NLRP3 inflammasome: Peptides. Peptides 2025; 187:171381. [PMID: 40064242 DOI: 10.1016/j.peptides.2025.171381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
The NLRP3 inflammasome plays a crucial role as a critical regulator of the immune response and has been implicated in the pathogenesis of numerous diseases. Peptides, known for their remarkable potency, selectivity, and low toxicity, have been extensively employed in disease treatment. Recent research has unveiled the potential of peptides in modulating the activity of the NLRP3 inflammasome. This review begins by examining the structure of the NLRP3 inflammasome, encompassing NLRP3, ASC, and Caspase-1, along with the three activation pathways: canonical, non-canonical, and alternative. Subsequently, we provide a comprehensive summary of peptide modulators targeting the NLRP3 inflammasome and elucidate their underlying mechanisms. The efficacy of these modulators has been validated through in vitro and in vivo experiments on NLRP3 inflammasome regulation. Furthermore, we conduct sequence alignment of the identified peptides and investigate their binding sites on the NLRP3 protein. This work is a foundational exploration for advancing peptides as potential therapeutic agents for NLRP3-related diseases.
Collapse
Affiliation(s)
- Zhuo Zuo
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yaxing Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yanwei Fang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Mengya Zhao
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhe Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhouqi Yang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Bin Jia
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yulong Sun
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
4
|
Sasaki Y, Kagohashi K, Kawahara S, Kitai Y, Muromoto R, Oritani K, Kashiwakura JI, Matsuda T. STAP-1-derived peptide suppresses TCR-mediated T cell activation and ameliorates immune diseases by inhibiting STAP-1-LCK binding. Immunohorizons 2025; 9:vlaf015. [PMID: 40288812 PMCID: PMC12034384 DOI: 10.1093/immhor/vlaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Abstract
Signal-transducing adaptor protein-1 (STAP-1) is an adaptor protein specifically expressed in immune cells, such as T cells. We previously demonstrated that STAP-1 positively upregulates T cell receptor (TCR)-mediated T cell activation by interacting with LCK and phospholipase C-γ1 and affecting autoimmune demyelination and airway inflammation. In this study, we aimed to generate a new STAP-1-derived peptide, iSP1, to inhibit the STAP-1-LCK interaction. We also analyzed its function in vitro and in vivo. iSP1 successfully interfered with STAP-1-LCK binding and suppressed TCR-mediated signal transduction, interleukin-2 production, and human and murine T cell proliferation. Additionally, iSP1 prevented the progression of experimental autoimmune encephalomyelitis by inhibiting Th1 and Th17 cell infiltration. Our findings suggest iSP1 as a new therapeutic immunomodulatory agent for T cell-mediated autoimmune diseases.
Collapse
MESH Headings
- Animals
- Humans
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Mice
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Signal Transduction/drug effects
- Mice, Inbred C57BL
- Peptides/pharmacology
- Protein Binding/drug effects
- Cell Proliferation/drug effects
- Th17 Cells/immunology
- Interleukin-2/metabolism
- T-Lymphocytes/immunology
- Th1 Cells/immunology
- Female
Collapse
Affiliation(s)
- Yuto Sasaki
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kota Kagohashi
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shoya Kawahara
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuichi Kitai
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ryuta Muromoto
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare, Narita, Chiba, Japan
| | - Jun-Ichi Kashiwakura
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Life Science, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Hokkaido, Japan
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
5
|
Petrov D, Plais L, Schira K, Cai J, Keller M, Lessing A, Bassi G, Cazzamalli S, Neri D, Gloger A, Scheuermann J. Flexibility-tuning of dual-display DNA-encoded chemical libraries facilitates cyclic peptide ligand discovery. Nat Commun 2025; 16:3273. [PMID: 40188178 PMCID: PMC11972359 DOI: 10.1038/s41467-025-58507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
Cyclic peptides constitute an important drug modality since they offer significant advantages over small molecules and macromolecules. However, access to diverse chemical sets of cyclic peptides is difficult on a large library scale. DNA-encoded Chemical Libraries (DELs) provide a suitable tool to obtain large chemical diversity, but cyclic DELs made by standard DEL implementation cannot efficiently explore their conformational diversity. On the other hand, dual-display Encoded Self-Assembling Chemical (ESAC) Libraries can be used for modulating macrocycle flexibility since the two displayed peptides can be connected in an incremental fashion. In this work, we construct a 56 million dual-display ESAC library using a two-step cyclization strategy. We show that varying the level of conformational restraint is essential for the discovery of specific ligands for the three protein targets thrombin, human alkaline phosphatase and streptavidin.
Collapse
Affiliation(s)
- Dimitar Petrov
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Louise Plais
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Kristina Schira
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Junyu Cai
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Michelle Keller
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Alice Lessing
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Gabriele Bassi
- Philochem AG, Libernstrasse 3, 8112, Otelfingen, Switzerland
| | | | - Dario Neri
- Philochem AG, Libernstrasse 3, 8112, Otelfingen, Switzerland
| | - Andreas Gloger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland
| | - Jörg Scheuermann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093, Zurich, Switzerland.
| |
Collapse
|
6
|
Limbach MN, Lindberg ET, Shrestha C, Lou J, Steren CA, Best MD, Do TD. Biased Equilibrium Drives Cyclosporine Membrane Permeability: The Goldilocks Energy Barriers. J Med Chem 2025; 68:6588-6600. [PMID: 40077929 DOI: 10.1021/acs.jmedchem.4c03178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Conformational flexibility allows macrocyclic peptides like cyclosporine A (CycA) to cross membranes, yet drug design leveraging this property has largely failed. A key challenge is linking specific conformers to function, as different conformers govern permeability versus target binding. We reveal a mechanism that enhances CycA and alisporivir (ALI) permeability: trans-to-cis isomerization at MeVal11-MeBmt1 creates conformers that remain "soluble" in both membrane-like and aqueous environments. A biased equilibrium favors this conformer in protic environments, while a lipophilic conformer with cis MeLeu9-MeLeu10 dominates in aprotic conditions. This mechanism explains why CycH, Valspodar (VALSPO), and O-acetyl CycA (OAc-CycA) fail to cross membranes─they adopt similar states but lack this biased equilibrium. Our findings provide a new strategy for designing membrane-permeable N-methylated macrocycles and underscore the role of high-energy conformers as transition states between membrane permeability and target engagement─offering critical insights for drug development.
Collapse
Affiliation(s)
- Miranda N Limbach
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Edward T Lindberg
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Cynthiya Shrestha
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Carlos A Steren
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Thanh D Do
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| |
Collapse
|
7
|
Nomura K, Hochido H, Morita Y, Hashimoto S, Kawada H. Preparation of sterically hindered peptides using trifluoroacetyl protection: incorporation of N-alkyl-α,α-dialkyl amino acids into N-alkyl amino acids. Chem Commun (Camb) 2025; 61:4856-4859. [PMID: 40042883 DOI: 10.1039/d4cc05780e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
We have established a synthetic method for sterically hindered sequences, specifically the motif of N-alkyl-α,α-dialkyl amino acids to N-alkyl amino acids. The method consists of two steps, exploiting the unique properties of the trifluoroacetyl group for N-protection: enhancing the electrophilicity of Tfa-protected N-H amino acids, and increasing the acidity of the N-H moiety to enable site-selective N-alkylation.
Collapse
Affiliation(s)
- Kenichi Nomura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Hidekazu Hochido
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Yuya Morita
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Satoshi Hashimoto
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Hatsuo Kawada
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| |
Collapse
|
8
|
Zuo Q, Song X, Yan J, Bao G, Li Y, Shen J, He Z, Hu K, Sun W, Wang R. Triazination/IEDDA Cascade Modular Strategy Installing Pyridines/Pyrimidines onto Tyrosine Enables Peptide Screening and Optimization. J Am Chem Soc 2025; 147:9576-9589. [PMID: 39885681 DOI: 10.1021/jacs.4c17615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Modular chemical postmodification of peptides is a promising strategy that supports the optimization and innovation of hit peptide therapeutics by enabling rapid derivatization. However, current methods are primarily limited to traditional bio-orthogonal strategies and chemical ligation techniques, which require the preintroduction of non-natural amino acids and impose fixed methods that limit peptide diversity. Here, we developed the Tyrosine-1,2,3-Triazine Ligation (YTL) strategy, which constructs novel linkages (pyridine and pyrimidine) through a "one-pot, two-step" process combining SNAr and IEDDA reactions, promoting modular post modification of Tyr-containing peptides. After optimizing the YTL strategy and establishing standard procedures, we successfully applied it to the solid-phase postmodification of various biorelated peptides, such as the synthesis of dual-mode imaging probes and long-acting GLP-1 analogs. As a proof of concept, a library of 384 amphipathic peptides was constructed using YTL based on 96-well microfiltration plates. Modular modifications were then performed on the screened template tripeptide RYR, leading to the generation of 20 derivatives. The antibacterial activity of these derivatives was systematically characterized, identifying Z8 as a potential antibacterial candidate.
Collapse
Affiliation(s)
- Quan Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Xinyi Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Jie Yan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Zeyuan He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| |
Collapse
|
9
|
Dubey GK, Chen PHC, Iskandar SE, Garikiparthy N, Zokian HJ, Lee HJJ, Weinglass A, Saldanha SA, Hallenbeck KK. Avoiding Misreading during Genetic Reprogramming in mRNA Display. Chembiochem 2025:e2500083. [PMID: 40055142 DOI: 10.1002/cbic.202500083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/07/2025] [Indexed: 04/09/2025]
Abstract
mRNA display is a powerful and increasingly accessible peptide discovery technology. It takes advantage of a reconstituted in vitro transcription and translation system to generate highly diverse affinity screening libraries. However, this process relies on the faithful translation of genetically encoded peptides, a conversion that is imperfect. Errors in translational decoding of mRNA can occur, decoupling the produced library from its genetic code. Because mRNA display affinity selections are analyzed with sequencing of the encoding DNA, rather than direct detection of the peptides, misreading silently reduces library diversity and complicates analysis. Herein, the presence of significant translational misreading during the production of mRNA display libraries is confirmed, best practices for genetic reprogramming are developed, and those rules are deployed to minimize the disconnect between genotype and phenotype in peptide affinity selections.
Collapse
Affiliation(s)
- Gopal K Dubey
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Peng-Hsun Chase Chen
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Sabrina E Iskandar
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Naga Garikiparthy
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Hratch J Zokian
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Hu-Jung Julie Lee
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Adam Weinglass
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - S Adrian Saldanha
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Kenneth K Hallenbeck
- Screening and Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, 07065, USA
| |
Collapse
|
10
|
Guo C, Ling N, Tian H, Wang Z, Gao M, Chen Y, Ji C. Comprehensive review of extraction, purification, structural characteristics, pharmacological activities, structure-activity relationship and application of seabuckthorn protein and peptides. Int J Biol Macromol 2025; 294:139447. [PMID: 39756720 DOI: 10.1016/j.ijbiomac.2024.139447] [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: 11/26/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Seabuckthorn (Hippophae rhamnoides) is an excellent plant that has the concomitant function of both medicine and foodstuff with high nutritional and health-promoting properties. As a pivotal bioactive component mainly existing in the seeds and leaves, seabuckthorn protein and its derived peptides have aroused wide attention owing to their multifaceted pharmacological activities, including anti-hypertensive, hypoglycemic, anti-obesity, anti-freeze, immunomodulatory, anti-inflammatory, sobriety, anti-oxidant and anti-neurodegenerative functions. Despite these promising attributes, the application of seabuckthorn peptides as functional food and medicines are impeded due to lack of a comprehensive understanding of pharmacological activities and intricate structure-activity relationship. Therefore, this review systematically summarizes the latest advancements in the extraction, purification, structural characteristics, pharmacological activities, digestion, absorption and transport, and application of seabuckthorn protein or peptides. Noteworthily, the structure-activity relationship is specifically delved into the hypoglycemic, anti-hypertensive, anti-obesity, anti-neurodegenerative and anti-oxidant peptides. Moreover, the shortcomings of current research and promising prospects are also highlighted. This comprehensive overview will provide a framework for future exploration and application of seabuckthorn protein or peptides in the realms of food and pharmaceuticals, offering a promising horizon for health benefits.
Collapse
Affiliation(s)
- Chunqiu Guo
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Na Ling
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| | - Haiyan Tian
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Zihao Wang
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Mingze Gao
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Yin Chen
- School of Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China
| | - Chenfeng Ji
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| |
Collapse
|
11
|
Dykstra R, Sindhikara D. Scoring Conformational Metastability of Macrocyclic Peptides with Binding Pose Metadynamics. J Chem Inf Model 2025; 65:1585-1604. [PMID: 39880805 DOI: 10.1021/acs.jcim.4c01408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Potency optimization of macrocyclic peptides can include both modifying intermolecular interactions and modifying the conformational stability of the bioactive conformation. However, the number of possible modifications is vast. To identify modifications that enhance the stability of the binding conformations in a cost-effective manner, there is a need for a high-throughput in-silico method that scores the conformational stability of these modified molecules. For the common case where a binding conformation of a similar compound is known, the relative stability of this conformation for a series of compounds can theoretically be estimated by modeling the metastability of the bound state via conformational sampling techniques. Herein, we survey several sampling methods and report solution-state binding pose metadynamics as the most efficient of such sampling methods. In this manuscript, we compare both estimations of metastability from shorter solution-state sampling methods to both experimental affinities and more rigorous sampling methods to properly isolate the conformational effect on potency. In our benchmark calculations on macrocyclic peptide data sets where chemical modifications can be expected to influence the stability of the binding pose, our solution-state binding pose metadynamics workflow, which estimates conformational metastability of the bioactive state, agrees with more rigorous REST2 simulations while using significantly less computational resources. Further, for both the cases where REST2 simulations converge, as well as some others, the binding pose metadynamics metastability estimations correlated well with experimentally measured potencies, suggesting binding pose metadynamics may be an efficient method for quickly estimating the effect of binding pose metastability on potency.
Collapse
Affiliation(s)
- Ryan Dykstra
- Department of Modeling and Informatics, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Dan Sindhikara
- Department of Modeling and Informatics, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
12
|
D'Alessio-Sands L, Gaynier J, Michel-Milian V, Agbowuro AA, Brackett CM. Current Strategies and Future Dimensions in the Development of KRAS Inhibitors for Targeted Anticancer Therapy. Drug Dev Res 2025; 86:e70042. [PMID: 39799558 DOI: 10.1002/ddr.70042] [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: 09/10/2024] [Revised: 11/05/2024] [Accepted: 12/15/2024] [Indexed: 01/15/2025]
Abstract
KRAS is a proto-oncogene that is found to be mutated in 15% of all metastatic cancers with high prevalence in pancreatic, lung, and colorectal cancers. Additionally, patients harboring KRAS mutations respond poorly to standard cancer therapy. As a result, KRAS is seen as an attractive target for targeted anticancer therapy. Over the last decade, this protein has evolved from being termed "undruggable" to producing two clinically approved drugs along with several more in clinical development, and many under preclinical investigations. This review details the development of various KRAS-targeted molecules with emphasis on the different drug design strategies employed by examining the following areas: (1) Direct inhibition of KRAS mutants using small molecule binders, (2) Inhibiting the activated state of KRAS mutants using a binary complex of small molecule binders and cyclophilin A, and (3) Targeted degradation of KRAS mutants using the PROTAC approach. We assess the pharmacological attributes and possible clinical benefits of the different molecules and look to the next frontiers in the application of KRAS inhibitors as anticancer agents.
Collapse
Affiliation(s)
| | - Joshua Gaynier
- South University School of Pharmacy, Savannah, Giorgia, USA
| | | | | | | |
Collapse
|
13
|
Kekec A, Tran LML, Plummer CW, Kalyani D. Late-stage installation and functionalization of alkyl pyridiniums: a general HTE amenable strategy to access diverse aryl alanine containing macrocyclic peptides. Chem Sci 2025; 16:2287-2294. [PMID: 39776654 PMCID: PMC11701726 DOI: 10.1039/d4sc06837h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
This manuscript describes a strategy to readily access diverse aryl and homoaryl alanine-containing pharmaceutically relevant macrocyclic peptides. A two-step sequence involving the late-stage installation of the pyridinium functionality on macrocyclic peptides followed by reductive couplings was implemented. These transformations are amenable to microscale high-throughput experimentation (HTE) and enable rapid access to aryl alanine-containing macrocyclic peptides that would otherwise be inaccessible via solid-phase peptide synthesis using commercially available amino acids. Numerous aryl and heteroaryl derivatives can be effectively used in these reactions. In addition, a systematic investigation was undertaken using an "informer" set of macrocyclic peptides which revealed the compatibility of the late-stage diversification with peptides containing diverse side chain functionalities.
Collapse
Affiliation(s)
- Ahmet Kekec
- Discovery Chemistry, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | | | | | | |
Collapse
|
14
|
Perdriau C, Luton A, Zimmeter K, Neuville M, Saragaglia C, Peluso-Iltis C, Osz J, Kauffmann B, Collie GW, Rochel N, Guichard G, Pasco M. Guanidinium-Stapled Helical Peptides for Targeting Protein-Protein Interactions. Angew Chem Int Ed Engl 2025; 64:e202416348. [PMID: 39714600 DOI: 10.1002/anie.202416348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
Peptide stapling has emerged as a versatile approach in drug discovery to reinforce secondary structure elements especially α-helices and improve properties of linear bioactive peptides. Inspired by the prevalence of arginine in protein-protein and protein-DNA interfaces, we investigated guanidinium-stapling as a means to constrain helical peptides. Guanidinium stapling was readily achieved on solid support, utilizing two orthogonally protected lysine or unatural α-amino acid residues with an amino function. This method allows for easy modulation of the nature and size of the staple as well as helix propensity. Evaluating a set of guanidinium-stapled peptides for their interaction with different protein targets identified several binders with increased target affinity. X-ray structure determination of four complexes revealed that all stapled peptides adopt a helical conformation upon protein binding. Notably, the disubstituted guanidinium generally exhibits a distinct cis/trans conformation and, in one instance, retains a conserved hydrogen bond with the protein surface. By identifying, for the first time, the guanidinium moiety as an effective helical peptide stapling group, this research significantly expands the repertoire of α-helix stapling techniques for the creation of useful protein mimics.
Collapse
Affiliation(s)
- Camille Perdriau
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | - Anaïs Luton
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | - Katharina Zimmeter
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | - Maxime Neuville
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
- IMMUPHARMA BIOTECH SAS, 15 rue de Bruxelles, 75009, Paris, France
| | - Claire Saragaglia
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | - Carole Peluso-Iltis
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, U1258/CNRS, UMR 7104/Univ. Strasbourg, 67404, Illkirch, France
| | - Judit Osz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, U1258/CNRS, UMR 7104/Univ. Strasbourg, 67404, Illkirch, France
| | - Brice Kauffmann
- Univ. Bordeaux, CNRS, INSERM, IECB, UAR3033, US001, F-33600, Pessac, France
| | - Gavin W Collie
- Discovery Sciences, R&D, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Natacha Rochel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM, U1258/CNRS, UMR 7104/Univ. Strasbourg, 67404, Illkirch, France
| | - Gilles Guichard
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| | - Morgane Pasco
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR5248, IECB, 2 rue Robert Escarpit, F-33600, Pessac, France
| |
Collapse
|
15
|
Ye Q, Shvartsbart A, Li Z, Gan P, Policarpo RL, Qi C, Roach JJ, Zhu W, McCammant MS, Hu B, Li G, Yin H, Carlsen P, Hoang G, Zhao L, Susick R, Zhang F, Lai CT, Allali Hassani A, Epling LB, Gallion A, Kurzeja-Lipinski K, Gallagher K, Roman V, Farren MR, Kong W, Deller MC, Zhang G, Covington M, Diamond S, Kim S, Yao W, Sokolsky A, Wang X. Discovery of INCB159020, an Orally Bioavailable KRAS G12D Inhibitor. J Med Chem 2025; 68:1924-1939. [PMID: 39772605 DOI: 10.1021/acs.jmedchem.4c02662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The inhibition of mutant KRAS proteins has emerged as a promising approach for treating KRAS-driven cancers, as evidenced by the clinical success of KRAS G12C inhibitors. KRAS G12D, the most common mutant, promises significant expansion of the addressable patient population; however, the reduced nucleophilicity of aspartate compared to cysteine poses significant challenges in balancing sufficient potency with ADME properties to support oral exposure. Herein, we describe the discovery of KRAS G12D inhibitor 23 (INCB159020), which achieves oral exposure in nonhuman primate (NHP). Starting from a weakly potent hit, structure-based drug design was utilized to drive significant potency. Focus on molecular rigidity and balanced polarity then allowed for successful optimization of properties required for oral exposure.
Collapse
Affiliation(s)
- Qinda Ye
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Artem Shvartsbart
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Zhenwu Li
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Pei Gan
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Rocco L Policarpo
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Chao Qi
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Jeremy J Roach
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Wenyu Zhu
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Matthew S McCammant
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Bin Hu
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Gencheng Li
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Haolin Yin
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Peter Carlsen
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Gia Hoang
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Le Zhao
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Robert Susick
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Fenglei Zhang
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Cheng-Tsung Lai
- Department of Computational Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Abdellah Allali Hassani
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Leslie B Epling
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Alexandra Gallion
- Department of Preclinical Pharmacology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Kerri Kurzeja-Lipinski
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Karen Gallagher
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Valerie Roman
- Department of Preclinical Pharmacology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Matthew R Farren
- Department of Preclinical Pharmacology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Weixi Kong
- Department of DMB, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Marc C Deller
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Guofeng Zhang
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Maryanne Covington
- Department of Applied Technology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Sharon Diamond
- Department of DMB, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Sunkyu Kim
- Department of Preclinical Pharmacology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Wenqing Yao
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
- Department of Preclinical Pharmacology, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803, United States
| | - Alexander Sokolsky
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| | - Xiaozhao Wang
- Department of Discovery Chemistry, Incyte Research Institute, Incyte Corporation, Wilmington, Delaware 19803 United States
| |
Collapse
|
16
|
Jing X, Mackay JP, Passioura T. Macrocyclic peptides as a new class of targeted protein degraders. RSC Chem Biol 2025:d4cb00199k. [PMID: 39822773 PMCID: PMC11733494 DOI: 10.1039/d4cb00199k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025] Open
Abstract
Targeted protein degraders, in the form of proteolysis targeting chimaeras (PROTACs) and molecular glues, leverage the ubiquitin-proteasome system to catalytically degrade specific target proteins of interest. Because such molecules can be extremely potent, they have attracted considerable attention as a therapeutic modality in recent years. However, while targeted degraders have great potential, they are likely to face many of the same challenges as more traditional small molecules when it comes to their development as therapeutics. In particular, existing targeted degrader design is largely only applicable to the same set of protein targets as traditional small molecules (i.e., ∼15% of the human proteome). Here, we consider the potential of macrocyclic peptides to overcome this limitation. Such molecules possess several features that make them well-suited for the role, including the ability to induce the formation of ternary protein complexes that can involve relatively flat surfaces and their structural commonality with E3 ligase-recruiting peptide degrons. For these reasons, macrocyclic peptides provide the opportunity both to broaden the number of targets accessible to degrader activity and to broaden the number of E3 ligases that can be harnessed to mediate that activity.
Collapse
Affiliation(s)
- Xuefei Jing
- School of Life and Environmental Sciences, The University of Sydney Sydney NSW 2006 Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney Sydney NSW 2006 Australia
| | - Toby Passioura
- School of Chemistry, The University of Sydney Sydney NSW 2006 Australia
- Insamo South Pty Ltd Chippendale NSW 2008 Australia
| |
Collapse
|
17
|
Wéber E, Ábrányi-Balogh P, Nagymihály B, Menyhárd DK, Péczka N, Gadanecz M, Schlosser G, Orgován Z, Bogár F, Bajusz D, Kecskeméti G, Szabó Z, Bartus É, Tököli A, Tóth GK, Szalai TV, Takács T, de Araujo E, Buday L, Perczel A, Martinek TA, Keserű GM. Target-Templated Construction of Functional Proteomimetics Using Photo-Foldamer Libraries. Angew Chem Int Ed Engl 2025; 64:e202410435. [PMID: 39329252 DOI: 10.1002/anie.202410435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 09/28/2024]
Abstract
Current methods for proteomimetic engineering rely on structure-based design. Here we describe a design strategy that allows the construction of proteomimetics against challenging targets without a priori characterization of the target surface. Our approach employs (i) a 100-membered photoreactive foldamer library, the members of which act as local surface mimetics, and (ii) the subsequent affinity maturation of the primary hits using systems chemistry. Two surface-oriented proteinogenic side chains drove the interactions between the short helical foldamer fragments and the proteins. Diazirine-based photo-crosslinking was applied to sensitively detect and localize binding even to shallow and dynamic patches on representatively difficult targets. Photo-foldamers identified functionally relevant protein interfaces, allosteric and previously unexplored targetable regions on the surface of STAT3 and an oncogenic K-Ras variant. Target-templated dynamic linking of foldamer hits resulted in two orders of magnitude affinity improvement in a single step. The dimeric K-Ras ligand mimicked protein-like catalytic functions. The photo-foldamer approach thus enables the highly efficient mapping of protein-protein interaction sites and provides a viable starting point for proteomimetic ligand development without a priori structural hypotheses.
Collapse
Affiliation(s)
- Edit Wéber
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Bence Nagymihály
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Dóra K Menyhárd
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Nikolett Péczka
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Márton Gadanecz
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, Eötvös Loránd University, Egyetem tér 1-3, H-1053, Budapest, Hungary
| | - Zoltán Orgován
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Gábor Kecskeméti
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Zoltán Szabó
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Éva Bartus
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Attila Tököli
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Gábor K Tóth
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Tibor V Szalai
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Szt. Gellért tér 4, H-1111, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Tamás Takács
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Egyetem tér 1-3, H-1053, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Elvin de Araujo
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Ontario, L5 L 1 C6, Mississauga, Canada
| | - László Buday
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - András Perczel
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Tamás A Martinek
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budafoki út 8, H-1111, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| |
Collapse
|
18
|
Mathiesen IR, Calder EDD, Kunzelmann S, Walport LJ. Discovering covalent cyclic peptide inhibitors of peptidyl arginine deiminase 4 (PADI4) using mRNA-display with a genetically encoded electrophilic warhead. Commun Chem 2024; 7:304. [PMID: 39702664 DOI: 10.1038/s42004-024-01388-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
Covalent drugs can achieve high potency with long dosing intervals. However, concerns remain about side-effects associated with off-target reactivity. Combining macrocyclic peptides with covalent warheads provides a solution to minimise off-target reactivity: the peptide enables highly specific target binding, positioning a weakly reactive warhead proximal to a suitable residue in the target. Here we demonstrate the direct discovery of covalent cyclic peptides using encoded libraries containing a weakly electrophilic cysteine-reactive fluoroamidine warhead. We combine direct incorporation of the warhead into peptide libraries using the flexible in vitro translation system with a peptide selection approach that identifies only covalent target binders. Using this approach, we identify potent and selective covalent inhibitors of the peptidyl arginine deiminase, PADI4 or PAD4, that react exclusively at the active site cysteine. We envisage this approach will enable covalent peptide inhibitor discovery for a range of related enzymes and expansion to alternative warheads in the future.
Collapse
Affiliation(s)
- Isabel R Mathiesen
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Ewen D D Calder
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Simone Kunzelmann
- Structural Biology Scientific Technology Platform, The Francis Crick Institute, London, UK
| | - Louise J Walport
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK.
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK.
| |
Collapse
|
19
|
Fechtmeyer PH, Yeh JTH. Temporal and Spatial Characterization of CUL3 KLHL20-driven Targeted Degradation of BET family, BRD Proteins by the Macrocycle-based Degrader BTR2004. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.07.627262. [PMID: 39677683 PMCID: PMC11643031 DOI: 10.1101/2024.12.07.627262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Targeted protein degradation (TPD) is a promising new therapeutic modality that leverages the endogenous cellular protein degradation machinery of the ubiquitin-proteasome system (UPS) to degrade selected proteins. Recently, we developed a synthetic macrocycle ligand to recruit CUL3KLHL20 E3 ligase for TPD. Using this KLHL20 ligand, we constructed the PROTAC BTR2004, which demonstrated potent degradation of BET family proteins BRD 2, 3, and 4. As the TPD field expands, it is important to understand the cellular and biochemical properties of all utilized E3 ligases. Herein we report the temporal and spatial processes of BTR2004-facilitated BET family protein degradation by KLHL20: The target protein degradation kinetics, BTR2004 intracellular activity half-life, and the onset of BTR2004 cell permeabilization. Employing proximity ligation and confocal microscopy techniques, we also illustrate the subcellular location of the ternary complex assembly upon BTR2004 treatment. These characterizations provide further insight into the processes that govern TPD and features that could be incorporated when designing future PROTAC molecules.
Collapse
|
20
|
Lamb HO, Benfield AH, Henriques ST. Peptides as innovative strategies to combat drug resistance in cancer therapy. Drug Discov Today 2024; 29:104206. [PMID: 39395530 DOI: 10.1016/j.drudis.2024.104206] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
Drug resistance is the leading cause of treatment failure in patients with cancer. Thus, innovative therapeutic strategies are required to overcome this critical challenge and improve patient outcomes. In this review, we examine the potential of peptide-based therapies to combat drug resistance in cancer. We highlight the unique strategies and mechanisms that can be explored by using peptides, including their ability to selectively target tumours, facilitate drug delivery into cancer cells, and inhibit key intracellular proteins that drive cancer progression and resistance. Peptides offer a promising approach to overcoming both intrinsic and adaptative cancer resistance against chemotherapy, targeted therapies, and biologics.
Collapse
Affiliation(s)
- Henry O Lamb
- School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Aurélie H Benfield
- School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Sónia Troeira Henriques
- School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Brisbane, QLD 4102, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
21
|
Tan Y, Yang J, Wang M, Peng Q, Li Y, Fu L, Zhang M, Wu J, Yang G, Hipolito CJ, Zhang Y, Qi J, Shi Y, Yin Y. De Novo Discovery of a Noncovalent Cell-Penetrating Bicyclic Peptide Inhibitor Targeting SARS-CoV-2 Main Protease. J Med Chem 2024; 67:20258-20274. [PMID: 39552553 DOI: 10.1021/acs.jmedchem.4c01639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Macrocyclic peptides have garnered significant attention as promising drug candidates. However, they typically face challenges in achieving and enhancing cell permeability for access to intracellular targets. In this study, we focused on the de novo screening of macrocyclic peptide inhibitors against the main protease (Mpro) of SARS-CoV-2 and identified novel noncovalently bound macrocyclic peptides that effectively inhibit proteolytic activity. High-resolution crystal structures further revealed molecular interactions between the macrocyclic peptides and Mpro. Subsequently, a specific macrocyclic peptide lacking cell permeability was further optimized and transformed into a low-toxicity, metabolically stable bicyclic peptide with a cell penetration capacity and therapeutic potential against SARS-CoV-2. The bicyclic peptide was achieved using a novel strategy that involved introducing both a bicyclic structure and a bridging perfluorobiphenyl group. Our study not only provides a lead peptide inhibitor for COVID-19 but also offers valuable insights into achieving cell penetration for macrocyclic peptides through strategic modifications.
Collapse
Affiliation(s)
- Yahong Tan
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jinyue Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Min Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qi Peng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongqi Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lifeng Fu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mengmeng Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jiang Wu
- AI and Life Sciences Institute (Hong Kong) Limited, 6/F., Building 17W, No. 17 Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, Shatin 999077, Hong Kong
| | - Guanya Yang
- AI and Life Sciences Institute (Hong Kong) Limited, 6/F., Building 17W, No. 17 Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, Shatin 999077, Hong Kong
| | - Christopher John Hipolito
- Screening & Compound Profiling, Quantitative Biosciences, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Youming Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Shi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Life Science Academy, Beijing 102209, China
| | - Yizhen Yin
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao 266237, China
- Shandong Research Institute of Industrial Technology, Jinan 250101, China
| |
Collapse
|
22
|
Swenson CS, Mandava G, Thomas DM, Moellering RE. Tackling Undruggable Targets with Designer Peptidomimetics and Synthetic Biologics. Chem Rev 2024; 124:13020-13093. [PMID: 39540650 PMCID: PMC12036645 DOI: 10.1021/acs.chemrev.4c00423] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The development of potent, specific, and pharmacologically viable chemical probes and therapeutics is a central focus of chemical biology and therapeutic development. However, a significant portion of predicted disease-causal proteins have proven resistant to targeting by traditional small molecule and biologic modalities. Many of these so-called "undruggable" targets feature extended, dynamic protein-protein and protein-nucleic acid interfaces that are central to their roles in normal and diseased signaling pathways. Here, we discuss the development of synthetically stabilized peptide and protein mimetics as an ever-expanding and powerful region of chemical space to tackle undruggable targets. These molecules aim to combine the synthetic tunability and pharmacologic properties typically associated with small molecules with the binding footprints, affinities and specificities of biologics. In this review, we discuss the historical and emerging platforms and approaches to design, screen, select and optimize synthetic "designer" peptidomimetics and synthetic biologics. We examine the inspiration and design of different classes of designer peptidomimetics: (i) macrocyclic peptides, (ii) side chain stabilized peptides, (iii) non-natural peptidomimetics, and (iv) synthetic proteomimetics, and notable examples of their application to challenging biomolecules. Finally, we summarize key learnings and remaining challenges for these molecules to become useful chemical probes and therapeutics for historically undruggable targets.
Collapse
Affiliation(s)
- Colin S Swenson
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Gunasheil Mandava
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Deborah M Thomas
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Raymond E Moellering
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
23
|
Yeste-Vázquez A, Paulussen FM, Wendt M, Klintrot R, Schulte C, Wallraven K, van Gijzel L, Simeonov B, van der Gaag M, Gerber A, Maric HM, Hennig S, Grossmann TN. Structure-Based Design of Bicyclic Helical Peptides That Target the Oncogene β-Catenin. Angew Chem Int Ed Engl 2024; 63:e202411749. [PMID: 39167026 DOI: 10.1002/anie.202411749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
The inhibition of intracellular protein-protein interactions is challenging, in particular, when involved interfaces lack pronounced cavities. The transcriptional co-activator protein and oncogene β-catenin is a prime example of such a challenging target. Despite extensive targeting efforts, available high-affinity binders comprise only large molecular weight inhibitors. This hampers the further development of therapeutically useful compounds. Herein, we report the design of a considerably smaller peptidomimetic scaffold derived from the α-helical β-catenin-binding motif of Axin. Sequence maturation and bicyclization provided a stitched peptide with an unprecedented crosslink architecture. The binding mode and site were confirmed by a crystal structure. Further derivatization yielded a β-catenin inhibitor with single-digit micromolar activity in a cell-based assay. This study sheds light on how to design helix mimetics with reduced molecular weight thereby improving their biological activity.
Collapse
Affiliation(s)
- Alejandro Yeste-Vázquez
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Felix M Paulussen
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Mathias Wendt
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Rasmus Klintrot
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Clemens Schulte
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of, Wuerzburg, Germany
| | - Kerstin Wallraven
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lieke van Gijzel
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Boris Simeonov
- Department of Neurosurgery, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maurice van der Gaag
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Neurosurgery, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alan Gerber
- Department of Neurosurgery, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hans M Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of, Wuerzburg, Germany
| | - Sven Hennig
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| | - Tom N Grossmann
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Koch G, Engstrom A, Taechalertpaisarn J, Faris J, Ono S, Naylor MR, Lokey RS. Chromatographic Determination of Permeability-Relevant Lipophilicity Facilitates Rapid Analysis of Macrocyclic Peptide Scaffolds. J Med Chem 2024; 67:19612-19622. [PMID: 39453819 PMCID: PMC11571107 DOI: 10.1021/acs.jmedchem.4c01956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/27/2024]
Abstract
Hydrocarbon-determined shake-flask measurements have demonstrated great utility for optimizing lipophilicity during early drug discovery. Alternatively, chromatographic methods confer reduced experimental error and improved handling of complex mixtures. In this study, we developed a chromatographic approach for estimating hydrocarbon-water shake-flask partition coefficients for a variety of macrocyclic peptides and other bRo5 molecules including PROTACs. The model accurately predicts experimental shake-flask measurements with high reproducibility across a wide range of lipophilicities. The chromatographic retention times revealed subtle conformational effects and correlated with the ability to sequester hydrogen bond donors in low dielectric media. Estimations of shake-flask lipophilicity from our model also accurately predicted trends in MDCK passive cell permeability for a variety of thioether-cyclized decapeptides. This method provides a convenient, high-throughput approach for measuring lipophilic permeability efficiency and predicting passive cell permeability in bRo5 compounds that is suitable for multiplexing pure compounds or investigating the properties of complex library mixtures.
Collapse
Affiliation(s)
- Grant Koch
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156 High St., Santa Cruz, California 95064, United States
| | - Alexander Engstrom
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156 High St., Santa Cruz, California 95064, United States
| | - Jaru Taechalertpaisarn
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156 High St., Santa Cruz, California 95064, United States
| | - Justin Faris
- Department
of Discovery Chemistry, Revolution Medicines,
Inc., Redwood City, California 94063, United States
| | - Satoshi Ono
- Innovative
Research Division, Mitsubishi Tanabe Pharma
Corporation, Kanagawa 227-0033, Japan
| | - Matthew R. Naylor
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156 High St., Santa Cruz, California 95064, United States
| | - R. Scott Lokey
- Department
of Chemistry and Biochemistry, University
of California, Santa Cruz, 1156 High St., Santa Cruz, California 95064, United States
| |
Collapse
|
25
|
Colas K, Bindl D, Suga H. Selection of Nucleotide-Encoded Mass Libraries of Macrocyclic Peptides for Inaccessible Drug Targets. Chem Rev 2024; 124:12213-12241. [PMID: 39451037 PMCID: PMC11565579 DOI: 10.1021/acs.chemrev.4c00422] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024]
Abstract
Technological advances and breakthrough developments in the pharmaceutical field are knocking at the door of the "undruggable" fortress with increasing insistence. Notably, the 21st century has seen the emergence of macrocyclic compounds, among which cyclic peptides are of particular interest. This new class of potential drug candidates occupies the vast chemical space between classic small-molecule drugs and larger protein-based therapeutics, such as antibodies. As research advances toward clinical targets that have long been considered inaccessible, macrocyclic peptides are well-suited to tackle these challenges in a post-rule of 5 pharmaceutical landscape. Facilitating their discovery is an arsenal of high-throughput screening methods that exploit massive randomized libraries of genetically encoded compounds. These techniques benefit from the incorporation of non-natural moieties, such as non- proteinogenic amino acids or stabilizing hydrocarbon staples. Exploiting these features for the strategic architectural design of macrocyclic peptides has the potential to tackle challenging targets such as protein-protein interactions, which have long resisted research efforts. This Review summarizes the basic principles and recent developments of the main high-throughput techniques for the discovery of macrocyclic peptides and focuses on their specific deployment for targeting undruggable space. A particular focus is placed on the development of new design guidelines and principles for the cyclization and structural stabilization of cyclic peptides and the resulting success stories achieved against well-known inaccessible drug targets.
Collapse
Affiliation(s)
- Kilian Colas
- University of Tokyo, Department of Chemistry, Graduate School of Science 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Daniel Bindl
- University of Tokyo, Department of Chemistry, Graduate School of Science 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- University of Tokyo, Department of Chemistry, Graduate School of Science 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
26
|
Chène P. Direct Inhibition of the YAP : TEAD Interaction: An Unprecedented Drug Discovery Challenge. ChemMedChem 2024; 19:e202400361. [PMID: 38863297 DOI: 10.1002/cmdc.202400361] [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: 05/10/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/13/2024]
Abstract
The Hippo pathway, which is key in organ morphogenesis, is frequently deregulated in cancer. The TEAD (TEA domain family member) transcription factors are the most distal elements of this pathway, and their activity is regulated by proteins such as YAP (Yes-associated protein). The identification of inhibitors of the YAP : TEAD interaction is one approach to develop novel anticancer drugs: the first clinical candidate (IAG933) preventing the association between these two proteins by direct competition has just been reported. The discovery of this molecule was particularly challenging because the interface between these two proteins is large (~3500 Å2 buried in complex formation) and made up of distinct contact areas. The most critical of these involves an omega-loop (Ω-loop), a secondary structure element rarely found in protein-protein interactions. This review summarizes how the knowledge gained from structure-function studies of the interaction between the Ω-loop of YAP and TEAD was used to devise the strategy to identify potent low-molecular weight compounds that show a pronounced anti-tumor effect.
Collapse
Affiliation(s)
- Patrick Chène
- Disease Area Oncology, Biomedical Research, CH-4056, Basel, Switzerland
- Novartis, WSJ 386 4.13.06, CH-4002, Basel, Switzerland
| |
Collapse
|
27
|
Hurd CA, Bush JT, Powell AJ, Walport LJ. mRNA Display in Cell Lysates Enables Identification of Cyclic Peptides Targeting the BRD3 Extraterminal Domain. Angew Chem Int Ed Engl 2024; 63:e202406414. [PMID: 38899853 DOI: 10.1002/anie.202406414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/21/2024]
Abstract
mRNA display is a powerful technology to screen libraries of >1012 cyclic peptides against a protein target, enabling the rapid discovery of high affinity ligands. These cyclic peptides are particularly well suited to challenging protein targets that have been difficult to drug with small molecules. However, target choice can still be limited as screens are typically performed against purified proteins which often demands the use of isolated domains and precludes the use of aggregation-prone targets. Herein, we report a method to perform mRNA display selections in mammalian cell lysates without the need for prior target purification, vastly expanding the potential target scope of mRNA display. We have applied the methodology to identify low to sub-nanomolar peptide binders for two targets: a NanoLuc subunit (LgBiT) and full-length bromodomain-containing protein 3 (BRD3). Our cyclic peptides for BRD3 were found to bind to the extraterminal (ET) domain of BRD3 and the closely related BRD proteins, BRD2 and BRD4. While many chemical probes exist for the bromodomains of BRD proteins, the ET domain is relatively underexplored, making these peptides valuable additions to the BRD toolbox.
Collapse
Affiliation(s)
- Catherine A Hurd
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, W12 0BZ
- Crick-GSK Biomedical LinkLabs, The Francis Crick Institute, London, NW1 1AT
| | - Jacob T Bush
- Crick-GSK Biomedical LinkLabs, GSK, Gunnels Wood Road, Stevenage, SG1 2NY
| | - Andrew J Powell
- Crick-GSK Biomedical LinkLabs, GSK, Gunnels Wood Road, Stevenage, SG1 2NY
| | - Louise J Walport
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, W12 0BZ
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, NW1 1AT
| |
Collapse
|
28
|
Darling WTP, Wieske LHE, Cook DT, Aliev AE, Caron L, Humphrys EJ, Figueiredo AM, Hansen DF, Erdélyi M, Tabor AB. The Influence of Disulfide, Thioacetal and Lanthionine-Bridges on the Conformation of a Macrocyclic Peptide. Chemistry 2024; 30:e202401654. [PMID: 38953277 DOI: 10.1002/chem.202401654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Cyclisation of peptides by forming thioether (lanthionine), disulfide (cystine) or methylene thioacetal bridges between side chains is established as an important tool to stabilise a given structure, enhance metabolic stability and optimise both potency and selectivity. However, a systematic comparative study of the effects of differing bridging modalities on peptide conformation has not previously been carried out. In this paper, we have used the NMR deconvolution algorithm, NAMFIS, to determine the conformational ensembles, in aqueous solution, of three cyclic analogues of angiotensin(1-7), incorporating either disulfide, or non-reducible thioether or methylene thioacetal bridges. We demonstrate that the major solution conformations are conserved between the different bridged peptides, but the distribution of conformations differs appreciably. This suggests that subtle differences in ring size and bridging structure can be exploited to fine-tune the conformational properties of cyclic peptides, which may modulate their bioactivities.
Collapse
Affiliation(s)
- William T P Darling
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Lianne H E Wieske
- Department of Chemistry-BMC, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Declan T Cook
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Abil E Aliev
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Laurent Caron
- Biosynth Laboratories Ltd (formerly Cambridge Research Biochemicals Ltd), 17-18 Belasis Court, Belasis Hall Technology Park, Billingham, TS23 4AZ, UK
| | - Emily J Humphrys
- Biosynth Laboratories Ltd (formerly Cambridge Research Biochemicals Ltd), 17-18 Belasis Court, Belasis Hall Technology Park, Billingham, TS23 4AZ, UK
| | - Angelo Miguel Figueiredo
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, UCL Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - D Flemming Hansen
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, UCL Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Máté Erdélyi
- Department of Chemistry-BMC, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Alethea B Tabor
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| |
Collapse
|
29
|
Wang F, Ma J, Yang L, Hu P, Tang S, Wang J, Li Z. Discovery of novel CXCR4 inhibitors for the treatment of inflammation by virtual screening and biological evaluation. Eur J Med Chem 2024; 275:116605. [PMID: 38885550 DOI: 10.1016/j.ejmech.2024.116605] [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: 05/14/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
C-X-C chemokine receptor type 4 (CXCR4) exerts considerable influence on the pathogenesis of inflammatory disorders and offers a potent avenue for drug intervention. This research utilizes a hybrid virtual screening methodology constructed using computer-aided drug design to discover novel CXCR4 inhibitors for the treatment of inflammation. First, a compound library was screened by Lipinski's five rules and adsorption, distribution, metabolism, excretion and toxicity properties. Second, the HypoGen algorithm was used in constructing a 3D-QSAR pharmacophore model and verify it layer by layer, and the obtained optimal pharmacophore 1 (Hypo 1) was used as a 3D query for compound screening. Then, hit compounds were obtained through molecular docking (Libdock and CDOCKER). The toxicity of the compounds to MDA-MB-231 cells was evaluated in vitro, and their binding affinity to the target was evaluated according to how they compete with 12G5 antibody for CXCR4 on the surfaces of the MDA-MB-231 cells. Compound Hit14 showed the strongest binding affinity among the hit compounds and inhibited cell migration and invasion in Matrigel invasion and wound healing assay at a concentration of 100 nM, demonstrating a better effect than AMD3100. Western Blot experiments further showed that Hit14 blocked the CXCR4/CXCL12-mediated phosphorylation of Akt. Meanwhile, cellular thermal displacement assay analysis showed that CXCR4 protein bound to Hit14 had high thermal stability. Finally, through in vivo experiments, we found that Hit14 inhibited mouse ear inflammation and reduced ear swelling and damage. Therefore, Hit14 is a promising drug for the further development of CXCR4 inhibitors for inflammation treatment.
Collapse
Affiliation(s)
- Fang Wang
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jie Ma
- The Central Hospital of Wuhan, Tongji Medical College of HUST, Wuhan, China
| | - Lili Yang
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ping Hu
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Siming Tang
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jing Wang
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Zeng Li
- The Key Laboratory for Joint Construction of Synthetic Bioprotein of Anhui Province, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.
| |
Collapse
|
30
|
Otani Y, Ichinose A, Wang X, Huang Z, Kasahara A, Ishii M, Watanabe E, Kanamitsu K, Tai K, Kusuhara H, Ueda T, Takeuchi K, Ohwada T. An N-ortho-nitrobenzylated benzanilide amino acid enables control of the conformation and membrane permeability of cyclic peptides. Chem Commun (Camb) 2024; 60:9242-9245. [PMID: 39115107 DOI: 10.1039/d4cc02738h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
We designed and synthesized an N-ortho-nitrobenzylated benzanilide-based amino acid having a cis-amide structure that facilitates cyclization of peptides containing it. Photo-induced removal of the nitrobenzyl group from this residue in the resulting cyclized peptides dramatically alters their conformation and passive membrane permeability via complete cis-amide to trans-amide conversion.
Collapse
Affiliation(s)
- Yuko Otani
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Asami Ichinose
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Xihong Wang
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Zhihan Huang
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Akitomo Kasahara
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Mayumi Ishii
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Eri Watanabe
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Kayoko Kanamitsu
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Kempei Tai
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Hiroyuki Kusuhara
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Takumi Ueda
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6, Yamadaoka, Suita-Shi, Osaka, Japan
| | - Koh Takeuchi
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Tomohiko Ohwada
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
31
|
Li M, Li J, Lu X, Schroder R, Chandramohan A, Wuelfing WP, Templeton AC, Xu W, Gindy M, Kesisoglou F, Ling J, Sawyer T, Verma CS, Partridge AW, Su Y. Molecular Mechanism of P53 Peptide Permeation through Lipid Membranes from Solid-State NMR Spectroscopy and Molecular Dynamics Simulations. J Am Chem Soc 2024; 146:23075-23091. [PMID: 39110018 DOI: 10.1021/jacs.4c04230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Macrocyclic peptides show promise in targeting high-value therapeutically relevant binding sites due to their high affinity and specificity. However, their clinical application is often hindered by low membrane permeability, which limits their effectiveness against intracellular targets. Previous studies focused on peptide conformations in various solvents, leaving a gap in understanding their interactions with and translocation through lipid bilayers. Addressing this, our study explores the membrane interactions of stapled peptides, a subclass of macrocyclic peptides, using solid-state nuclear magnetic resonance (ssNMR) spectroscopy and molecular dynamics (MD) simulations. We conducted ssNMR measurements on ATSP-7041M, a prototypical stapled peptide, to understand its interaction with lipid membranes, leading to an MD-informed model for peptide membrane permeation. Our findings reveal that ATSP-7041M adopts a stable α-helical structure upon membrane binding, facilitated by a cation-π interaction between its phenylalanine side chain and the lipid headgroup. This interaction makes the membrane-bound state energetically favorable, facilitating membrane affinity and insertion. The bound peptide displayed asymmetric insertion depths, with the C-terminus penetrating deeper (approximately 9 Å) than the N-terminus (approximately 4.3 Å) relative to the lipid headgroups. Contrary to expectations, peptide dynamics was not hindered by membrane binding and exhibited rapid motions similar to cell-penetrating peptides. These dynamic interactions and peptide-lipid affinity appear to be crucial for membrane permeation. MD simulations indicated a thermodynamically stable transmembrane conformation of ATSP-7041M, reducing the energy barrier for translocation. Our study offers an in silico view of ATSP-7041M's translocation from the extracellular to the intracellular region, highlighting the significance of peptide-lipid interactions and dynamics in enabling peptide transit through membranes.
Collapse
Affiliation(s)
- Mingyue Li
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jianguo Li
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
- Singapore Eye Research Institute, 20 College Road Discovery Tower, Singapore 169856, Singapore
| | - Xingyu Lu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
- Instrumentation and Service Center for Molecular Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Ryan Schroder
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - W Peter Wuelfing
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Allen C Templeton
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Wei Xu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marian Gindy
- Small Molecule Science and Technology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Filippos Kesisoglou
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jing Ling
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Tomi Sawyer
- Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Chandra S Verma
- Bioinformatics Institute at A*STAR (Agency for Science, Technology and Research), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
- School of Biological Sciences, Nanyang Technological University, 50 Nanyang Drive, Singapore 637551, Singapore
| | | | - Yongchao Su
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
32
|
Thorpe MP, Smith AN, Blackwell DJ, Hopkins CR, Knollmann BC, Akers WS, Johnston JN. The backbone constitution drives passive permeability independent of side chains in depsipeptide and peptide macrocycles inspired by ent-verticilide. Chem Sci 2024; 15:d4sc02758b. [PMID: 39211739 PMCID: PMC11348715 DOI: 10.1039/d4sc02758b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
The number of peptide-like scaffolds found in late-stage drug development is increasing, but a critical unanswered question in the field is whether substituents (side chains) or the backbone drive passive permeability. The backbone is scrutinized in this study. Five series of macrocyclic peptidic compounds were prepared, and their passive permeability was determined (PAMPA, Caco-2), to delineate structure-permeability relationships. Each series was based on the cell-permeable antiarrhythmic compound ent-verticilide, a cyclic oligomeric depsipeptide (COD) containing repeating ester/N-Me amide didepsipeptide monomers. One key finding is that native lipophilic ester functionality can impart a favorable level of permeability, but ester content alone is not the final determinant - the analog with highest P app was discovered by a single ester-to-N-H amide replacement. Furthermore, the relative composition of esters and N-Me amides in a series had more nuanced permeability behavior. Overall, a systematic approach to structure-permeability correlations suggests that a combinatorial-like investigation of functionality in peptidic or peptide-like compounds could better identify leads with optimal passive permeability, perhaps prior to modification of side chains.
Collapse
Affiliation(s)
- Madelaine P Thorpe
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University Nashville TN 37235-1822 USA
| | - Abigail N Smith
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University Nashville TN 37235-1822 USA
| | - Daniel J Blackwell
- Department of Medicine, Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center Medical Research Bldg IV, Room 1265, 2215B Garland Ave Nashville TN 37232-0575 USA
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68198 USA
| | - Bjorn C Knollmann
- Department of Medicine, Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center Medical Research Bldg IV, Room 1265, 2215B Garland Ave Nashville TN 37232-0575 USA
| | - Wendell S Akers
- Pharmaceutical Sciences Research Center, College of Pharmacy, Lipscomb University Nashville TN 37204 USA
| | - Jeffrey N Johnston
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University Nashville TN 37235-1822 USA
| |
Collapse
|
33
|
Wu J, Wang Y, Cai W, Chen D, Peng X, Dong H, Li J, Liu H, Shi S, Tang S, Li Z, Sui H, Wang Y, Wu C, Zhang Y, Fu X, Yin Y. Ribosomal translation of fluorinated non-canonical amino acids for de novo biologically active fluorinated macrocyclic peptides. Chem Sci 2024:d4sc04061a. [PMID: 39129776 PMCID: PMC11310889 DOI: 10.1039/d4sc04061a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Fluorination has emerged as a promising strategy in medicinal chemistry to improve the pharmacological profiles of drug candidates. Similarly, incorporating fluorinated non-canonical amino acids into macrocyclic peptides expands chemical diversity and enhances their pharmacological properties, from improved metabolic stability to enhanced cell permeability and target interactions. However, only a limited number of fluorinated non-canonical amino acids, which are canonical amino acid analogs, have been incorporated into macrocyclic peptides by ribosomes for de novo construction and target-based screening of fluorinated macrocyclic peptides. In this study, we report the ribosomal translation of a series of distinct fluorinated non-canonical amino acids, including mono-to tri-fluorinated variants, as well as fluorinated l-amino acids, d-amino acids, β-amino acids, etc. This enabled the de novo discovery of fluorinated macrocyclic peptides with high affinity for EphA2, and particularly the identification of those exhibiting broad-spectrum activity against Gram-negative bacteria by targeting the BAM complex. This study not only expands the scope of ribosomally translatable fluorinated amino acids but also underscores the versatility of fluorinated macrocyclic peptides as potent therapeutic agents.
Collapse
Affiliation(s)
- Junjie Wu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University Qingdao 266237 China
| | - Yuchan Wang
- College of Life Sciences, Fujian Normal University Fuzhou 350117 China
| | - Wenfeng Cai
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University Qingdao 266237 China
| | - Danyan Chen
- College of Life Sciences, Fujian Normal University Fuzhou 350117 China
| | - Xiangda Peng
- Shanghai Zelixir Biotech Company Ltd Shanghai 200030 China
| | - Huilei Dong
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Jinjing Li
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Hongtan Liu
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Shuting Shi
- College of Life Sciences, Fujian Normal University Fuzhou 350117 China
| | - Sen Tang
- College of Life Sciences, Fujian Normal University Fuzhou 350117 China
| | - Zhifeng Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University Qingdao 266237 China
| | - Haiyan Sui
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University Qingdao 266237 China
| | - Yan Wang
- College of Life Sciences, Fujian Normal University Fuzhou 350117 China
| | - Chuanliu Wu
- College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Youming Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University Qingdao 266237 China
| | - Xinmiao Fu
- College of Life Sciences, Fujian Normal University Fuzhou 350117 China
| | - Yizhen Yin
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University Qingdao 266237 China
- Shandong Research Institute of Industrial Technology Jinan 250101 China
| |
Collapse
|
34
|
Kage M, Hayashi R, Matsuo A, Tamiya M, Kuramoto S, Ohara K, Irie M, Chiyoda A, Takano K, Ito T, Kotake T, Takeyama R, Ishikawa S, Nomura K, Furuichi N, Morita Y, Hashimoto S, Kawada H, Nishimura Y, Nii K, Sase H, Ohta A, Kojima T, Iikura H, Tanada M, Shiraishi T. Structure-activity relationships of middle-size cyclic peptides, KRAS inhibitors derived from an mRNA display. Bioorg Med Chem 2024; 110:117830. [PMID: 38981216 DOI: 10.1016/j.bmc.2024.117830] [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: 05/27/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cyclic peptides are attracting attention as therapeutic agents due to their potential for oral absorption and easy access to tough intracellular targets. LUNA18, a clinical KRAS inhibitor, was transformed-without scaffold hopping-from the initial hit by using an mRNA display library that met our criteria for drug-likeness. In drug discovery using mRNA display libraries, hit compounds always possess a site linked to an mRNA tag. Here, we describe our examination of the Structure-Activity Relationship (SAR) using X-ray structures for chemical optimization near the site linked to the mRNA tag, equivalent to the C-terminus. Structural modifications near the C-terminus demonstrated a relatively wide range of tolerance for side chains. Furthermore, we show that a single atom modification is enough to change the pharmacokinetic (PK) profile. Since there are four positions where side chain modification is permissible in terms of activity, it is possible to flexibly adjust the pharmacokinetic profile by structurally optimizing the side chain. The side chain transformation findings demonstrated here may be generally applicable to hits obtained from mRNA display libraries.
Collapse
Affiliation(s)
- Mirai Kage
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Ryuji Hayashi
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Atsushi Matsuo
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Minoru Tamiya
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Shino Kuramoto
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Kazuhiro Ohara
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Machiko Irie
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Aya Chiyoda
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Koji Takano
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Toshiya Ito
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Tomoya Kotake
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Ryuuichi Takeyama
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Shiho Ishikawa
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Kenichi Nomura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Noriyuki Furuichi
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Yuya Morita
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Satoshi Hashimoto
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Hatsuo Kawada
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Yoshikazu Nishimura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Keiji Nii
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Hitoshi Sase
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Atsushi Ohta
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Tetsuo Kojima
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Hitoshi Iikura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Mikimasa Tanada
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Takuya Shiraishi
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| |
Collapse
|
35
|
Li Y, Yang L, Li X, Zhang X. Inhibition of GTPase KRAS G12D: a review of patent literature. Expert Opin Ther Pat 2024; 34:701-721. [PMID: 38884569 DOI: 10.1080/13543776.2024.2369630] [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: 04/03/2024] [Accepted: 06/14/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION KRAS is a critical oncogenic protein intricately involved in tumor progression, and the difficulty in targeting KRAS has led it to be classified as an 'undruggable target.' Among the various KRAS mutations, KRASG12D is highly prevalent and represents a promising therapeutic target, yet there are currently no approved inhibitors for it. AREA COVERED This review summarizes numerous patents and literature featuring inhibitors or degraders of KRASG12D through searching relevant information in PubMed, SciFinder and Web of Science databases from 2021 to February 2024, providing an overview of the research progress on inhibiting KRASG12D in terms of design strategies, chemical structures, biological activities, and clinical advancements. EXPERT OPINION Since the approval of AMG510 (Sotorasib), there has been an increasing focus on the inhibition of KRASG12D, leading to numerous reports of related inhibitors and degraders. Among them, MRTX1133, as the first KRASG12D inhibitor to enter clinical trials, has demonstrated excellent tumor suppression in various KRASG12D-bearing human tumor xenograft models. It is important to note, however, that understanding the mechanisms of acquired resistance caused by KRAS inhibition and developing additional combination therapies is crucial. Moreover, seeking covalent inhibition of KRASG12D also holds significant potential.
Collapse
Affiliation(s)
- Yuhang Li
- Department of Chemistry, China Pharmaceutical University, Nanjing, China
| | - Le Yang
- Department of Chemistry, China Pharmaceutical University, Nanjing, China
| | - Xiaoran Li
- Department of Chemistry, China Pharmaceutical University, Nanjing, China
- AceMapAI Joint Lab, China Pharmaceutical University, Nanjing, China
| | - Xiaojin Zhang
- Department of Chemistry, China Pharmaceutical University, Nanjing, China
- AceMapAI Joint Lab, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
36
|
Dai B, Chen JN, Zeng Q, Geng H, Wu YD. Accurate Structure Prediction for Cyclic Peptides Containing Proline Residues with High-Temperature Molecular Dynamics. J Phys Chem B 2024; 128:7322-7331. [PMID: 39028892 DOI: 10.1021/acs.jpcb.4c02004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Cyclic peptides (CPs) are emerging as promising drug candidates. Numerous natural CPs and their analogs are effective therapeutics against various diseases. Notably, many of them contain peptidyl cis-prolyl bonds. Due to the high rotational barrier of peptide bonds, conventional molecular dynamics simulations struggle to effectively sample the cis/trans-isomerization of peptide bonds. Previous studies have highlighted the high accuracy of the residue-specific force field (RSFF) and the high sampling efficiency of high-temperature molecular dynamics (high-T MD). Herein, we propose a protocol that combines high-T MD with RSFF2C and a recently developed reweighting method based on probability densities for accurate structure prediction of proline-containing CPs. Our method successfully predicted 19 out of 23 CPs with the backbone rmsd < 1.0 Å compared to X-ray structures. Furthermore, we performed high-T MD and density reweighting on the sunflower trypsin inhibitor (SFTI-1)/trypsin complex to demonstrate its applicability in studying CP-complexes containing cis-prolines. Our results show that the conformation of SFTI-1 in aqueous solution is consistent with its bound conformation, potentially facilitating its binding.
Collapse
Affiliation(s)
- Botao Dai
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Jia-Nan Chen
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Qing Zeng
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Hao Geng
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yun-Dong Wu
- Lab of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518132, China
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
37
|
You S, McIntyre G, Passioura T. The coming of age of cyclic peptide drugs: an update on discovery technologies. Expert Opin Drug Discov 2024; 19:961-973. [PMID: 38872502 DOI: 10.1080/17460441.2024.2367024] [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: 04/25/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
INTRODUCTION Cyclic peptides are an established class of pharmaceuticals, with the ability to bind to a broader range of protein targets than traditional small molecules while also being capable of oral availability and cell penetration. Historically, cyclic peptide drugs have been discovered almost exclusively through natural product mining approaches; however, the last two decades have seen the development of display screening approaches capable of rapidly identifying de novo (i.e. not natural product derived) cyclic peptide ligands to targets of interest. AREAS COVERED In this review, the authors describe the current clinical landscape for cyclic peptide pharmaceuticals. This article focuses on the discovery approaches that have led to the development of different classes of molecules and how the development of newer technologies, particularly phage and mRNA display, has broadened the clinical applicability of such molecules. EXPERT OPINION The field of de novo cyclic peptide drug discovery is reaching maturity, with the first drugs identified through display screening approaches reaching the market in recent years. Many more are in clinical trials; however, significant technical challenges remain. Technological improvements will be required over the coming years to facilitate the identification of membrane permeable cyclic peptides capable of oral availability and targeting intracellular proteins.
Collapse
Affiliation(s)
- Sophia You
- Insamo South, Chippendale, NSW, Australia
| | | | - Toby Passioura
- School of Chemistry, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
38
|
Yeffet D, Columbus I, Parvari G, Eichen Y, Saphier S, Ghindes-Azaria L, Redy-Keisar O, Amir D, Drug E, Gershonov E, Binyamin I, Cohen Y, Karton-Lifshin N, Zafrani Y. Addressing the Opioids Lipophilicity Challenge via a Straightforward and Simultaneous 1H NMR-Based log P/ D Determination, Both Separately and in Mixtures. J Med Chem 2024; 67:12399-12409. [PMID: 39013123 DOI: 10.1021/acs.jmedchem.4c01153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
A systematic study of trends in the lipophilicity of prominent representatives of the opioid family, including natural, semisynthetic, synthetic, and endogenous neuropeptide opioids, is described. This was enabled by a straightforward 1H NMR-based logP/D determination method developed for compounds holding at least one aromatic hydrogen atom. Moreover, the new method enables a direct simultaneous logD determination of opioid mixtures, overcoming the high sensitivity of this family to the measurement conditions, which is critical when a determination of the exact ΔlogD values of matched pairs is required. Interpretation of the experimental ΔlogD7.4 values of selected matched pairs, focusing inter alia on the 3-OMe and 14-OMe motifs in morphinan opioids, is suggested with the aid of DFT calculations and may be useful for the discovery of new opioid therapeutics.
Collapse
Affiliation(s)
- Dina Yeffet
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Ishay Columbus
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Galit Parvari
- Schulich Faculty of Chemistry Technion, Israel Institute of Technology, Technion City, Haifa 3200008, Israel
| | - Yoav Eichen
- Schulich Faculty of Chemistry Technion, Israel Institute of Technology, Technion City, Haifa 3200008, Israel
| | - Sigal Saphier
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Lee Ghindes-Azaria
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Orit Redy-Keisar
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Dafna Amir
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Eyal Drug
- Department of Analytical Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Eytan Gershonov
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Iris Binyamin
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Yoram Cohen
- School of Chemistry, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Naama Karton-Lifshin
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | - Yossi Zafrani
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| |
Collapse
|
39
|
Lamartina CW, Chartier CA, Hirano JM, Shah NH, Rovis T. Crafting Unnatural Peptide Macrocycles via Rh(III)-Catalyzed Carboamidation. J Am Chem Soc 2024. [PMID: 39024122 DOI: 10.1021/jacs.4c05248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Contemporary developments in the field of peptide macrocyclization methodology are imperative for enabling the advance of drug design in medicinal chemistry. This report discloses a Rh(III)-catalyzed macrocyclization via carboamidation, reacting acryloyl-peptide-dioxazolone precursors and arylboronic acids to form complex cyclic peptides with concomitant incorporation of noncanonical α-amino acids. The diverse and modular technology allows for expedient access to a wide variety of cyclic peptides from 4 to 15 amino acids in size and features simultaneous formation of unnatural phenylalanine and tyrosine derivatives with up to >20:1 diastereoselectivity. The reaction showcases an expansive substrate scope with 45 examples and is compatible with the majority of standard protected amino acids used in Fmoc-solid phase peptide synthesis. The methodology is applied to the synthesis of multiple peptidomimetic macrocyclic analogs, including derivatives of cyclosomatostatin and gramicidin S.
Collapse
Affiliation(s)
| | - Cassandra A Chartier
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Jillian M Hirano
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Neel H Shah
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Tomislav Rovis
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| |
Collapse
|
40
|
Krishna Sudhakar H, Yau JTK, Alcock LJ, Lau YH. Accessing diverse bicyclic peptide conformations using 1,2,3-TBMB as a linker. Org Biomol Chem 2024. [PMID: 39007293 DOI: 10.1039/d4ob00901k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Bicyclic peptides are a powerful modality for engaging challenging drug targets such as protein-protein interactions. Here, we use 1,2,3-tris(bromomethyl)benzene (1,2,3-TBMB) to access bicyclic peptides with diverse conformations that differ from conventional bicyclisation products formed with 1,3,5-TBMB. Bicyclisation at cysteine residues under aqueous buffer conditions proceeds efficiently, with broad substrate scope, compatibility with high-throughput screening, and clean conversion (>90%) for 96 of the 115 peptides tested. We envisage that the 1,2,3-TBMB linker will be applicable to a variety of peptide screening techniques in drug discovery.
Collapse
Affiliation(s)
| | - Jackie Tsz Ki Yau
- School of Chemistry, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Lisa J Alcock
- School of Chemistry, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Yu Heng Lau
- School of Chemistry, The University of Sydney, Camperdown, NSW 2006, Australia.
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Camperdown, NSW 2006, Australia.
| |
Collapse
|
41
|
Ma X, Sloman DL, Duggal R, Anderson KD, Ballard JE, Bharathan I, Brynczka C, Gathiaka S, Henderson TJ, Lyons TW, Miller R, Munsell EV, Orth P, Otte RD, Palani A, Rankic DA, Robinson MR, Sather AC, Solban N, Song XS, Wen X, Xu Z, Yang Y, Yang R, Day PJ, Stoeck A, Bennett DJ, Han Y. Discovery of MK-1084: An Orally Bioavailable and Low-Dose KRAS G12C Inhibitor. J Med Chem 2024; 67:11024-11052. [PMID: 38924388 DOI: 10.1021/acs.jmedchem.4c00572] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Oncogenic mutations in the RAS gene account for 30% of all human tumors; more than 60% of which present as KRAS mutations at the hotspot codon 12. After decades of intense pursuit, a covalent inhibition strategy has enabled selective targeting of this previously "undruggable" target. Herein, we disclose our journey toward the discovery of MK-1084, an orally bioavailable and low-dose KRASG12C covalent inhibitor currently in phase I clinical trials (NCT05067283). We leveraged structure-based drug design to identify a macrocyclic core structure, and hypothesis-driven optimization of biopharmaceutical properties to further improve metabolic stability and tolerability.
Collapse
Affiliation(s)
- Xiaoshen Ma
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - David L Sloman
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Ruchia Duggal
- Department of Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Kenneth D Anderson
- Department of Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jeanine E Ballard
- Department of Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Indu Bharathan
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Christopher Brynczka
- Department of Nonclinical Drug Safety, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Symon Gathiaka
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Timothy J Henderson
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Thomas W Lyons
- Department of Process Research and Development, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Richard Miller
- Department of Discovery Quantitative Biosciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Erik V Munsell
- Department of Discovery Pharmaceutical Sciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Peter Orth
- Department of Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, New Jersey 07065, United States
| | - Ryan D Otte
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Anandan Palani
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Danica A Rankic
- Department of Process Research and Development, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Michelle R Robinson
- Department of Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Aaron C Sather
- Department of Process Research and Development, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Nicolas Solban
- Department of Discovery Quantitative Biosciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Xuelei Sherry Song
- Department of Discovery Quantitative Biosciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Xin Wen
- Department of Process Research and Development, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Zangwei Xu
- Department of Discovery Quantitative Biosciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Yi Yang
- Department of Discovery Quantitative Biosciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Ruojing Yang
- Department of Discovery Quantitative Biosciences, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Phil J Day
- Department of Structural Biology, Astex Pharmaceuticals, 436 Cambridge Science Park, Cambridge CB4 0QA, U.K
| | - Alexander Stoeck
- Department of Discovery Biology, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - David Jonathan Bennett
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| | - Yongxin Han
- Department of Discovery Chemistry, Merck & Co., Inc., 33 Ave. Louis Pasteur, Boston, Massachusetts 02215, United States
| |
Collapse
|
42
|
Yamada T, Mihara K, Ueda T, Yamauchi D, Shimizu M, Ando A, Mayumi K, Nakata Z, Mikamiyama H. Discovery and Hit to Lead Optimization of Macrocyclic Peptides as Novel Tropomyosin Receptor Kinase A Antagonists. J Med Chem 2024; 67:11197-11208. [PMID: 38950284 DOI: 10.1021/acs.jmedchem.4c00715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Tropomyosin receptor kinases (Trks) are receptor tyrosine kinases activated by neurotrophic factors, called neurotrophins. Among them, TrkA interacts with the nerve growth factor (NGF), which leads to pain induction. mRNA-display screening was carried out to discover a hit compound 2, which inhibits protein-protein interactions between TrkA and NGF. Subsequent structure optimization improving phosphorylation inhibitory activity and serum stability was pursued using a unique process that took advantage of the peptide being synthesized by translation from mRNA. This gave peptide 19, which showed an analgesic effect in a rat incisional pain model. The peptides described here can serve as a new class of analgesics, and the structure optimization methods reported provide a strategy for discovering new peptide drugs.
Collapse
Affiliation(s)
- Toru Yamada
- Biopharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Kousuke Mihara
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Taichi Ueda
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Daisuke Yamauchi
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Masaya Shimizu
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Azusa Ando
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Kei Mayumi
- Pharmaceutical Development Division, Yodoyabashi Office, Osaka , Osaka 541-0042, Japan
| | - Zenzaburo Nakata
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| | - Hidenori Mikamiyama
- Pharmaceutical Research Division, Shionogi Pharmaceutical Research Center, Toyonaka , Osaka 561-0825, Japan
| |
Collapse
|
43
|
Nielsen AL, Bognar Z, Mothukuri GK, Zarda A, Schüttel M, Merz ML, Ji X, Will EJ, Chinellato M, Bartling CRO, Strømgaard K, Cendron L, Angelini A, Heinis C. Large Libraries of Structurally Diverse Macrocycles Suitable for Membrane Permeation. Angew Chem Int Ed Engl 2024; 63:e202400350. [PMID: 38602024 DOI: 10.1002/anie.202400350] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 04/12/2024]
Abstract
Macrocycles offer an attractive format for drug development due to their good binding properties and potential to cross cell membranes. To efficiently identify macrocyclic ligands for new targets, methods for the synthesis and screening of large combinatorial libraries of small cyclic peptides were developed, many of them using thiol groups for efficient peptide macrocyclization. However, a weakness of these libraries is that invariant thiol-containing building blocks such as cysteine are used, resulting in a region that does not contribute to library diversity but increases molecule size. Herein, we synthesized a series of structurally diverse thiol-containing elements and used them for the combinatorial synthesis of a 2,688-member library of small, structurally diverse peptidic macrocycles with unprecedented skeletal complexity. We then used this library to discover potent thrombin and plasma kallikrein inhibitors, some also demonstrating favorable membrane permeability. X-ray structure analysis of macrocycle-target complexes showed that the size and shape of the newly developed thiol elements are key for binding. The strategy and library format presented in this work significantly enhance structural diversity by allowing combinatorial modifications to a previously invariant region of peptide macrocycles, which may be broadly applied in the development of membrane permeable therapeutics.
Collapse
Affiliation(s)
- Alexander L Nielsen
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Zsolt Bognar
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Ganesh K Mothukuri
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Anne Zarda
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Mischa Schüttel
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Manuel L Merz
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Xinjian Ji
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Edward J Will
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | | | - Christian R O Bartling
- Center for Biopharmaceuticals and Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Kristian Strømgaard
- Center for Biopharmaceuticals and Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Laura Cendron
- Department of Biology, University of Padova, 35131, Padova, Italy
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, Venezia Mestre, Venice, 30172, Italy
- European Centre for Living Technologies (ECLT), Ca' Bottacin, Dorsoduro 3911, Calle Crosera, Venice, 30124, Italy
| | - Christian Heinis
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| |
Collapse
|
44
|
Zuo Q, Li Y, Lai X, Bao G, Chen L, He Z, Song X, E R, Wang P, Shi Y, Luo H, Sun W, Wang R. Cysteine-Specific Multifaceted Bioconjugation of Peptides and Proteins Using 5-Substituted 1,2,3-Triazines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308491. [PMID: 38466927 DOI: 10.1002/advs.202308491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/08/2024] [Indexed: 03/13/2024]
Abstract
Peptide and protein postmodification have gained significant attention due to their extensive impact on biomolecule engineering and drug discovery, of which cysteine-specific modification strategies are prominent due to their inherent nucleophilicity and low abundance. Herein, the study introduces a novel approach utilizing multifunctional 5-substituted 1,2,3-triazine derivatives to achieve multifaceted bioconjugation targeting cysteine-containing peptides and proteins. On the one hand, this represents an inaugural instance of employing 1,2,3-triazine in biomolecular-specific modification within a physiological solution. On the other hand, as a powerful combination of precision modification and biorthogonality, this strategy allows for the one-pot dual-orthogonal functionalization of biomolecules utilizing the aldehyde group generated simultaneously. 1,2,3-Triazine derivatives with diverse functional groups allow conjugation to peptides or proteins, while bi-triazines enable peptide cyclization and dimerization. The examination of the stability of bi-triazines revealed their potential for reversible peptide modification. This work establishes a comprehensive platform for identifying cysteine-selective modifications, providing new avenues for peptide-based drug development, protein bioconjugation, and chemical biology research.
Collapse
Affiliation(s)
- Quan Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Xuanliang Lai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Lu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Zeyuan He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Xinyi Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Ruiyao E
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Pengxin Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Yuntao Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Huixin Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| |
Collapse
|
45
|
Sigal M, Matsumoto S, Beattie A, Katoh T, Suga H. Engineering tRNAs for the Ribosomal Translation of Non-proteinogenic Monomers. Chem Rev 2024; 124:6444-6500. [PMID: 38688034 PMCID: PMC11122139 DOI: 10.1021/acs.chemrev.3c00894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/21/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Ribosome-dependent protein biosynthesis is an essential cellular process mediated by transfer RNAs (tRNAs). Generally, ribosomally synthesized proteins are limited to the 22 proteinogenic amino acids (pAAs: 20 l-α-amino acids present in the standard genetic code, selenocysteine, and pyrrolysine). However, engineering tRNAs for the ribosomal incorporation of non-proteinogenic monomers (npMs) as building blocks has led to the creation of unique polypeptides with broad applications in cellular biology, material science, spectroscopy, and pharmaceuticals. Ribosomal polymerization of these engineered polypeptides presents a variety of challenges for biochemists, as translation efficiency and fidelity is often insufficient when employing npMs. In this Review, we will focus on the methodologies for engineering tRNAs to overcome these issues and explore recent advances both in vitro and in vivo. These efforts include increasing orthogonality, recruiting essential translation factors, and creation of expanded genetic codes. After our review on the biochemical optimizations of tRNAs, we provide examples of their use in genetic code manipulation, with a focus on the in vitro discovery of bioactive macrocyclic peptides containing npMs. Finally, an analysis of the current state of tRNA engineering is presented, along with existing challenges and future perspectives for the field.
Collapse
Affiliation(s)
- Maxwell Sigal
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satomi Matsumoto
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Adam Beattie
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayuki Katoh
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
46
|
Yin H, Tang Q, Xia H, Bi F. Targeting RAF dimers in RAS mutant tumors: From biology to clinic. Acta Pharm Sin B 2024; 14:1895-1923. [PMID: 38799634 PMCID: PMC11120325 DOI: 10.1016/j.apsb.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/02/2024] [Accepted: 02/20/2024] [Indexed: 05/29/2024] Open
Abstract
RAS mutations occur in approximately 30% of tumors worldwide and have a poor prognosis due to limited therapies. Covalent targeting of KRAS G12C has achieved significant success in recent years, but there is still a lack of efficient therapeutic approaches for tumors with non-G12C KRAS mutations. A highly promising approach is to target the MAPK pathway downstream of RAS, with a particular focus on RAF kinases. First-generation RAF inhibitors have been authorized to treat BRAF mutant tumors for over a decade. However, their use in RAS-mutated tumors is not recommended due to the paradoxical ERK activation mainly caused by RAF dimerization. To address the issue of RAF dimerization, type II RAF inhibitors have emerged as leading candidates. Recent clinical studies have shown the initial effectiveness of these agents against RAS mutant tumors. Promisingly, type II RAF inhibitors in combination with MEK or ERK inhibitors have demonstrated impressive efficacy in RAS mutant tumors. This review aims to clarify the importance of RAF dimerization in cellular signaling and resistance to treatment in tumors with RAS mutations, as well as recent progress in therapeutic approaches to address the problem of RAF dimerization in RAS mutant tumors.
Collapse
Affiliation(s)
- Huanhuan Yin
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiulin Tang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongwei Xia
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Bi
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Kirschner T, Müller MP, Rauh D. Targeting KRAS Diversity: Covalent Modulation of G12X and Beyond in Cancer Therapy. J Med Chem 2024; 67:6044-6051. [PMID: 38621359 DOI: 10.1021/acs.jmedchem.3c02403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The GTPase KRAS acts as a switch in cellular signaling, transitioning between inactive GDP-bound and active GTP-bound states. In about 20% of human cancers, oncogenic RAS mutations disrupt this balance, favoring the active form and promoting proliferative signaling, thus rendering KRAS an appealing target for precision medicine in oncology. In 2013, Shokat and co-workers achieved a groundbreaking feat by covalently targeting a previously undiscovered allosteric pocket (switch II pocket (SWIIP)) of KRASG12C. This breakthrough led to the development and approval of sotorasib (AMG510) and adagrasib (MRTX849), revolutionizing the treatment of KRASG12C-dependent lung cancer. Recent achievements in targeting various KRASG12X mutants, using SWIIP as a key binding pocket, are discussed. Insights from successful KRASG12C targeting informed the design of molecules addressing other mutations, often in a covalent manner. These findings offer promise for innovative approaches in addressing commonly occurring KRAS mutations such as G12D, G12V, G12A, G12S, and G12R in various cancers.
Collapse
Affiliation(s)
- Tonia Kirschner
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Matthias P Müller
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| | - Daniel Rauh
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund, Germany
| |
Collapse
|
48
|
Ball A, Mohammed S, Doigneaux C, Gardner RM, Easton JW, Turner S, Essex JW, Pairaudeau G, Tavassoli A. Identification and Development of Cyclic Peptide Inhibitors of Hypoxia Inducible Factors 1 and 2 That Disrupt Hypoxia-Response Signaling in Cancer Cells. J Am Chem Soc 2024; 146:8877-8886. [PMID: 38503564 PMCID: PMC10996005 DOI: 10.1021/jacs.3c10508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Hypoxia inducible factor (HIF) is a heterodimeric transcription factor composed of an oxygen-regulated α subunit and a constitutively expressed β subunit that serves as the master regulator of the cellular response to low oxygen concentrations. The HIF transcription factor senses and responds to hypoxia by significantly altering transcription and reprogramming cells to enable adaptation to a hypoxic microenvironment. Given the central role played by HIF in the survival and growth of tumors in hypoxia, inhibition of this transcription factor serves as a potential therapeutic approach for treating a variety of cancers. Here, we report the identification, optimization, and characterization of a series of cyclic peptides that disrupt the function of HIF-1 and HIF-2 transcription factors by inhibiting the interaction of both HIF-1α and HIF-2α with HIF-1β. These compounds are shown to bind to HIF-α and disrupt the protein-protein interaction between the α and β subunits of the transcription factor, resulting in disruption of hypoxia-response signaling by our lead molecule in several cancer cell lines.
Collapse
Affiliation(s)
- Andrew
T. Ball
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Soran Mohammed
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Cyrielle Doigneaux
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Reece M. Gardner
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - James W. Easton
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Steven Turner
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Jonathan W. Essex
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Garry Pairaudeau
- Discovery
Sciences IMED Biotech Unit, AstraZeneca, 310 Cambridge Science Park, Milton
Road, Cambridge CB4 0WG, U.K.
| | - Ali Tavassoli
- School
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| |
Collapse
|
49
|
Faris J, Adaligil E, Popovych N, Ono S, Takahashi M, Nguyen H, Plise E, Taechalertpaisarn J, Lee HW, Koehler MFT, Cunningham CN, Lokey RS. Membrane Permeability in a Large Macrocyclic Peptide Driven by a Saddle-Shaped Conformation. J Am Chem Soc 2024; 146:4582-4591. [PMID: 38330910 PMCID: PMC10885153 DOI: 10.1021/jacs.3c10949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
The effort to modulate challenging protein targets has stimulated interest in ligands that are larger and more complex than typical small-molecule drugs. While combinatorial techniques such as mRNA display routinely produce high-affinity macrocyclic peptides against classically undruggable targets, poor membrane permeability has limited their use toward primarily extracellular targets. Understanding the passive membrane permeability of macrocyclic peptides would, in principle, improve our ability to design libraries whose leads can be more readily optimized against intracellular targets. Here, we investigate the permeabilities of over 200 macrocyclic 10-mers using the thioether cyclization motif commonly found in mRNA display macrocycle libraries. We identified the optimal lipophilicity range for achieving permeability in thioether-cyclized 10-mer cyclic peptide-peptoid hybrid scaffolds and showed that permeability could be maintained upon extensive permutation in the backbone. In one case, changing a single amino acid from d-Pro to d-NMe-Ala, representing the loss of a single methylene group in the side chain, resulted in a highly permeable scaffold in which the low-dielectric conformation shifted from the canonical cross-beta geometry of the parent compounds into a novel saddle-shaped fold in which all four backbone NH groups were sequestered from the solvent. This work provides an example by which pre-existing physicochemical knowledge of a scaffold can benefit the design of macrocyclic peptide mRNA display libraries, pointing toward an approach for biasing libraries toward permeability by design. Moreover, the compounds described herein are a further demonstration that geometrically diverse, highly permeable scaffolds exist well beyond conventional drug-like chemical space.
Collapse
Affiliation(s)
- Justin
H. Faris
- Department
of Chemistry and Biochemistry, University
of California, Santa
Cruz, California 95064, United States
| | - Emel Adaligil
- Department
of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| | - Nataliya Popovych
- Department
of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Satoshi Ono
- Innovative
Research Division, Mitsubishi Tanabe Pharma
Corporation, Kanagawa 227-0033, Japan
| | - Mifune Takahashi
- Department
of Drug Metabolism and Pharmacokinetics, Genentech, South
San Francisco, California 94080, United States
| | - Huy Nguyen
- Department
of Analytical Research, Genentech, South San Francisco, California 94080, United States
| | - Emile Plise
- Department
of Drug Metabolism and Pharmacokinetics, Genentech, South
San Francisco, California 94080, United States
| | - Jaru Taechalertpaisarn
- Department
of Chemistry and Biochemistry, University
of California, Santa
Cruz, California 95064, United States
| | - Hsiau-Wei Lee
- Department
of Chemistry and Biochemistry, University
of California, Santa
Cruz, California 95064, United States
| | - Michael F. T. Koehler
- Department
of Medicinal Chemistry, Genentech, South San Francisco, California 94080, United States
| | - Christian N. Cunningham
- Department
of Peptide Therapeutics, Genentech, South San Francisco, California 94080, United States
| | - R. Scott Lokey
- Department
of Chemistry and Biochemistry, University
of California, Santa
Cruz, California 95064, United States
| |
Collapse
|
50
|
Pal S, 't Hart P. Imbuing peptide libraries with drug-likeness. Nat Chem Biol 2024; 20:140-141. [PMID: 38242957 DOI: 10.1038/s41589-023-01524-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Affiliation(s)
- Sunit Pal
- Max Planck Institute of Molecular Physiology, Chemical Genomics Centre, Dortmund, Germany
| | - Peter 't Hart
- Max Planck Institute of Molecular Physiology, Chemical Genomics Centre, Dortmund, Germany.
| |
Collapse
|