151
|
Jana T, Ghosh A, Das Mandal S, Banerjee R, Saha S. PPIMpred: a web server for high-throughput screening of small molecules targeting protein-protein interaction. ROYAL SOCIETY OPEN SCIENCE 2017; 4:160501. [PMID: 28484602 PMCID: PMC5414239 DOI: 10.1098/rsos.160501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 03/20/2017] [Indexed: 05/31/2023]
Abstract
PPIMpred is a web server that allows high-throughput screening of small molecules for targeting specific protein-protein interactions, namely Mdm2/P53, Bcl2/Bak and c-Myc/Max. Three different kernels of support vector machine (SVM), namely, linear, polynomial and radial basis function (RBF), and two other machine learning techniques including Naive Bayes and Random Forest were used to train the models. A fivefold cross-validation technique was used to measure the performance of these classifiers. The RBF kernel of SVM outperformed and/or was comparable with all other methods with accuracy values of 83%, 79% and 90% for Mdm2/P53, Bcl2/Bak and c-Myc/Max, respectively. About 80% of the predicted SVM scores of training/testing datasets from Mdm2/P53 and Bcl2/Bak have significant IC50 values and docking scores. The proposed models achieved an accuracy of 66-90% with blind sets. The three mentioned (Mdm2/P53, Bcl2/Bak and c-Myc/Max) proposed models were screened in a large dataset of 265 242 small chemicals from National Cancer Institute open database. To further realize the robustness of this approach, hits with high and random SVM scores were used for molecular docking in AutoDock Vina wherein the molecules with high and random predicted SVM scores yielded moderately significant docking scores (p-values < 0.1). In addition to the above-mentioned classification scheme, this web server also allows users to get the structural and chemical similarities with known chemical modulators or drug-like molecules based on Tanimoto coefficient similarity search algorithm. PPIMpred is freely available at http://bicresources.jcbose.ac.in/ssaha4/PPIMpred/.
Collapse
Affiliation(s)
- Tanmoy Jana
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| | - Abhirupa Ghosh
- Department of Bioinformatics, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| | - Sukhen Das Mandal
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| | - Raja Banerjee
- Department of Bioinformatics, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, India
| | - Sudipto Saha
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| |
Collapse
|
152
|
Nguyen D, Liao W, Zeng SX, Lu H. Reviving the guardian of the genome: Small molecule activators of p53. Pharmacol Ther 2017; 178:92-108. [PMID: 28351719 DOI: 10.1016/j.pharmthera.2017.03.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
Abstract
The tumor suppressor p53 is one of the most important proteins for protection of genomic stability and cancer prevention. Cancers often inactivate it by either mutating its gene or disabling its function. Thus, activating p53 becomes an attractive approach for the development of molecule-based anti-cancer therapy. The past decade and half have witnessed tremendous progress in this area. This essay offers readers with a grand review on this progress with updated information about small molecule activators of p53 either still at bench work or in clinical trials.
Collapse
Affiliation(s)
- Daniel Nguyen
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States
| | - Wenjuan Liao
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States
| | - Shelya X Zeng
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States
| | - Hua Lu
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, LA 70012, United States.
| |
Collapse
|
153
|
Garcia BL, Skaff DA, Chatterjee A, Hanning A, Walker JK, Wyckoff GJ, Geisbrecht BV. Identification of C3b-Binding Small-Molecule Complement Inhibitors Using Cheminformatics. THE JOURNAL OF IMMUNOLOGY 2017; 198:3705-3718. [PMID: 28298523 DOI: 10.4049/jimmunol.1601932] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/21/2017] [Indexed: 01/08/2023]
Abstract
The complement system is an elegantly regulated biochemical cascade formed by the collective molecular recognition properties and proteolytic activities of more than two dozen membrane-bound or serum proteins. Complement plays diverse roles in human physiology, such as acting as a sentry against invading microorganisms, priming of the adaptive immune response, and removal of immune complexes. However, dysregulation of complement can serve as a trigger for a wide range of human diseases, which include autoimmune, inflammatory, and degenerative conditions. Despite several potential advantages of modulating complement with small-molecule inhibitors, small-molecule drugs are highly underrepresented in the current complement-directed therapeutics pipeline. In this study, we have employed a cheminformatics drug discovery approach based on the extensive structural and functional knowledge available for the central proteolytic fragment of the cascade, C3b. Using parallel in silico screening methodologies, we identified 45 small molecules that putatively bind C3b near ligand-guided functional hot spots. Surface plasmon resonance experiments resulted in the validation of seven dose-dependent C3b-binding compounds. Competition-based biochemical assays demonstrated the ability of several C3b-binding compounds to interfere with binding of the original C3b ligand that guided their discovery. In vitro assays of complement function identified a single complement inhibitory compound, termed cmp-5, and mechanistic studies of the cmp-5 inhibitory mode revealed it acts at the level of C5 activation. This study has led to the identification of a promising new class of C3b-binding small-molecule complement inhibitors and, to our knowledge, provides the first demonstration of cheminformatics-based, complement-directed drug discovery.
Collapse
Affiliation(s)
- Brandon L Garcia
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506
| | - D Andrew Skaff
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110
| | - Arindam Chatterjee
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104; and
| | | | - John K Walker
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104; and
| | - Gerald J Wyckoff
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110
| | - Brian V Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506;
| |
Collapse
|
154
|
Hanold LE, Fulton MD, Kennedy EJ. Targeting kinase signaling pathways with constrained peptide scaffolds. Pharmacol Ther 2017; 173:159-170. [PMID: 28185915 DOI: 10.1016/j.pharmthera.2017.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Kinases are amongst the largest families in the human proteome and serve as critical mediators of a myriad of cell signaling pathways. Since altered kinase activity is implicated in a variety of pathological diseases, kinases have become a prominent class of proteins for targeted inhibition. Although numerous small molecule and antibody-based inhibitors have already received clinical approval, several challenges may still exist with these strategies including resistance, target selection, inhibitor potency and in vivo activity profiles. Constrained peptide inhibitors have emerged as an alternative strategy for kinase inhibition. Distinct from small molecule inhibitors, peptides can provide a large binding surface area that allows them to bind shallow protein surfaces rather than defined pockets within the target protein structure. By including chemical constraints within the peptide sequence, additional benefits can be bestowed onto the peptide scaffold such as improved target affinity and target selectivity, cell permeability and proteolytic resistance. In this review, we highlight examples of diverse chemistries that are being employed to constrain kinase-targeting peptide scaffolds and highlight their application to modulate kinase signaling as well as their potential clinical implications.
Collapse
Affiliation(s)
- Laura E Hanold
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, United States
| | - Melody D Fulton
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, United States
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, United States.
| |
Collapse
|
155
|
Comparative Study of Elastic Network Model and Protein Contact Network for Protein Complexes: The Hemoglobin Case. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2483264. [PMID: 28243596 PMCID: PMC5294226 DOI: 10.1155/2017/2483264] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/17/2016] [Accepted: 12/20/2016] [Indexed: 01/12/2023]
Abstract
The overall topology and interfacial interactions play key roles in understanding structural and functional principles of protein complexes. Elastic Network Model (ENM) and Protein Contact Network (PCN) are two widely used methods for high throughput investigation of structures and interactions within protein complexes. In this work, the comparative analysis of ENM and PCN relative to hemoglobin (Hb) was taken as case study. We examine four types of structural and dynamical paradigms, namely, conformational change between different states of Hbs, modular analysis, allosteric mechanisms studies, and interface characterization of an Hb. The comparative study shows that ENM has an advantage in studying dynamical properties and protein-protein interfaces, while PCN is better for describing protein structures quantitatively both from local and from global levels. We suggest that the integration of ENM and PCN would give a potential but powerful tool in structural systems biology.
Collapse
|
156
|
Choi S, Choi KY. Screening-based approaches to identify small molecules that inhibit protein–protein interactions. Expert Opin Drug Discov 2017; 12:293-303. [DOI: 10.1080/17460441.2017.1280456] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sehee Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- CK Biotechnology Inc., 416 Advanced Science and Technology Center, 50 Yonsei-ro, Seoul, Korea
| |
Collapse
|
157
|
Zhou Y, Zhang N, Chen W, Zhao L, Zhong R. Underlying mechanisms of cyclic peptide inhibitors interrupting the interaction of CK2α/CK2β: comparative molecular dynamics simulation studies. Phys Chem Chem Phys 2017; 18:9202-10. [PMID: 26974875 DOI: 10.1039/c5cp06276d] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Protein-protein interactions (PPIs) are fundamental to all biological processes. Recently, the CK2β-derived cyclic peptide Pc has been demonstrated to efficiently antagonize the CK2α/CK2β interaction and strongly affect the phosphorylation of CK2β-dependent CK2 substrate specificity. The binding affinity of Pc to CK2α is destroyed to different extents by two single-point mutations of Tyr188 to Ala (Y188A) and Phe190 to Ala (F190A), which exert negative effects on the inhibitory activity (IC50) of Pc against the CK2α/CK2β interaction from 3.0 μM to 54.0 μM and ≫100 μM, respectively. However, the structural influences of Y188A and F190A mutations on the CK2α-Pc complex remain unclear. In this study, comparative molecular dynamics (MD) simulations, principal component analysis (PCA), domain cross-correlation map (DCCM) analysis and energy calculations were performed on wild type (WT), Y188A mutant, and F190A mutant systems. The results revealed that ordered communications between hydrophobic and polar interactions were essential for CK2α-Pc binding in the WT system. In addition to the loss of the hydrogen bond between Gln36 of CK2α and Gly189 of Pc in the two mutants, the improper recognition mechanisms occurred through different pathways. These pathways included the weakened hydrophobic interactions in the Y188A mutant as well as decreased polar and hydrophobic interactions in the F190A mutant. The energy analysis results qualitatively elucidated the instability of the two mutants and energetic contributions of the key residues. This study not only revealed the structural mechanisms for the decreased binding affinity of Y188A and F190A mutant CK2α-Pc complexes, but also provided valuable clues for the rational design of CK2α/CK2β subunit interaction inhibitors with high affinity and specificity.
Collapse
Affiliation(s)
- Yue Zhou
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Na Zhang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Wenjuan Chen
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Lijiao Zhao
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Rugang Zhong
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
158
|
Wiedmann MM, Tan YS, Wu Y, Aibara S, Xu W, Sore HF, Verma CS, Itzhaki L, Stewart M, Brenton JD, Spring DR. Development of Cell-Permeable, Non-Helical Constrained Peptides to Target a Key Protein-Protein Interaction in Ovarian Cancer. Angew Chem Int Ed Engl 2017; 56:524-529. [PMID: 27918136 PMCID: PMC5291322 DOI: 10.1002/anie.201609427] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/18/2016] [Indexed: 02/04/2023]
Abstract
There is a lack of current treatment options for ovarian clear cell carcinoma (CCC) and the cancer is often resistant to platinum-based chemotherapy. Hence there is an urgent need for novel therapeutics. The transcription factor hepatocyte nuclear factor 1β (HNF1β) is ubiquitously overexpressed in CCC and is seen as an attractive therapeutic target. This was validated through shRNA-mediated knockdown of the target protein, HNF1β, in five high- and low-HNF1β-expressing CCC lines. To inhibit the protein function, cell-permeable, non-helical constrained proteomimetics to target the HNF1β-importin α protein-protein interaction were designed, guided by X-ray crystallographic data and molecular dynamics simulations. In this way, we developed the first reported series of constrained peptide nuclear import inhibitors. Importantly, this general approach may be extended to other transcription factors.
Collapse
Affiliation(s)
- Mareike M. Wiedmann
- Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
- Cancer Research UK Cambridge InstituteUniversity of CambridgeLi Ka Shing Centre, Robinson WayCambridgeCB2 0REUK
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and ResearchA*STAR30 Biopolis Street, #07-01 MatrixSingapore138671Singapore
| | - Yuteng Wu
- Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Shintaro Aibara
- SciLifeLabTomtebodavägen 23A171 65 SolnaStockholmSweden
- MRC Laboratory of Molecular BiologyFrancis Crick Avenue, Cambridge Biomedical CampusCambridgeCB2 0QHUK
| | - Wenshu Xu
- Department of PharmacologyTennis Court RoadCambridgeCB2 1PDUK
| | - Hannah F. Sore
- Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Chandra S. Verma
- Bioinformatics Institute, Agency for Science, Technology and ResearchA*STAR30 Biopolis Street, #07-01 MatrixSingapore138671Singapore
- School of Biological SciencesNanyang Technological University60 Nanyang DriveSingapore637551Singapore
- Department of Biological SciencesNational University of Singapore14 Science Drive 4Singapore117543Singapore
| | - Laura Itzhaki
- Department of PharmacologyTennis Court RoadCambridgeCB2 1PDUK
| | - Murray Stewart
- MRC Laboratory of Molecular BiologyFrancis Crick Avenue, Cambridge Biomedical CampusCambridgeCB2 0QHUK
| | - James D. Brenton
- Cancer Research UK Cambridge InstituteUniversity of CambridgeLi Ka Shing Centre, Robinson WayCambridgeCB2 0REUK
| | - David R. Spring
- Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| |
Collapse
|
159
|
Abstract
Physiological characteristics of diseases bring about both challenges and opportunities for targeted drug delivery. Various drug delivery platforms have been devised ranging from macro- to micro- and further into the nanoscopic scale in the past decades. Recently, the favorable physicochemical properties of nanomaterials, including long circulation, robust tissue and cell penetration attract broad interest, leading to extensive studies for therapeutic benefits. Accumulated knowledge about the physiological barriers that affect the in vivo fate of nanomedicine has led to more rational guidelines for tailoring the nanocarriers, such as size, shape, charge, and surface ligands. Meanwhile, progresses in material chemistry and molecular pharmaceutics generate a panel of physiological stimuli-responsive modules that are equipped into the formulations to prepare “smart” drug delivery systems. The capability of harnessing physiological traits of diseased tissues to control the accumulation of or drug release from nanomedicine has further improved the controlled drug release profiles with a precise manner. Successful clinical translation of a few nano-formulations has excited the collaborative efforts from the research community, pharmaceutical industry, and the public towards a promising future of smart drug delivery.
Collapse
Affiliation(s)
- Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Wenyan Ji
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Grace Wright
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
160
|
Denizli M, Aslan B, Mangala LS, Jiang D, Rodriguez-Aguayo C, Lopez-Berestein G, Sood AK. Chitosan Nanoparticles for miRNA Delivery. Methods Mol Biol 2017; 1632:219-230. [PMID: 28730442 PMCID: PMC7423176 DOI: 10.1007/978-1-4939-7138-1_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
RNA interference techniques represent a promising strategy for therapeutic applications. In addition to small interfering RNA-based approaches, which have been widely studied and translated into clinical investigations, microRNA-based approaches are attractive owing to their "one hit, multiple targets" concept. To overcome challenges with in vivo delivery of microRNAs related to stability, cellular uptake, and specific delivery, our group has developed and characterized chitosan nanoparticles for nucleotide delivery. This platform allows for robust target modulation and antitumor activity following intravenous administration.
Collapse
Affiliation(s)
- Merve Denizli
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingegowda S Mangala
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Bldg., CPB6.3275, 1515 Holcombe Blvd., Unit 1362, Houston, TX, 77030, USA
| | - Dahai Jiang
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Bldg., CPB6.3275, 1515 Holcombe Blvd., Unit 1362, Houston, TX, 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Bldg., CPB6.3275, 1515 Holcombe Blvd., Unit 1362, Houston, TX, 77030, USA.
- Program in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
161
|
Ohkanda J. A Strategy of Designing Mid-sized Synthetic Molecules that Recognize Interfaces of Protein-protein Interactions. J SYN ORG CHEM JPN 2017. [DOI: 10.5059/yukigoseikyokaishi.75.735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Junko Ohkanda
- Academic Assembly, Institute of Agriculture, Shinshu University
| |
Collapse
|
162
|
Zhang B, Zhang J, Peng S, Liu R, Li X, Hou Y, Han X, Fang J. Thioredoxin reductase inhibitors: a patent review. Expert Opin Ther Pat 2016; 27:547-556. [DOI: 10.1080/13543776.2017.1272576] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Shoujiao Peng
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Ruijuan Liu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Yanan Hou
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xiao Han
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| |
Collapse
|
163
|
Wiedmann MM, Tan YS, Wu Y, Aibara S, Xu W, Sore HF, Verma CS, Itzhaki L, Stewart M, Brenton JD, Spring DR. Development of Cell-Permeable, Non-Helical Constrained Peptides to Target a Key Protein-Protein Interaction in Ovarian Cancer. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201609427] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Mareike M. Wiedmann
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
- Cancer Research UK Cambridge Institute; University of Cambridge; Li Ka Shing Centre, Robinson Way Cambridge CB2 0RE UK
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research; A*STAR; 30 Biopolis Street, #07-01 Matrix Singapore 138671 Singapore
| | - Yuteng Wu
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Shintaro Aibara
- SciLifeLab; Tomtebodavägen 23A 171 65 Solna Stockholm Sweden
- MRC Laboratory of Molecular Biology; Francis Crick Avenue, Cambridge Biomedical Campus Cambridge CB2 0QH UK
| | - Wenshu Xu
- Department of Pharmacology; Tennis Court Road Cambridge CB2 1PD UK
| | - Hannah F. Sore
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Chandra S. Verma
- Bioinformatics Institute, Agency for Science, Technology and Research; A*STAR; 30 Biopolis Street, #07-01 Matrix Singapore 138671 Singapore
- School of Biological Sciences; Nanyang Technological University; 60 Nanyang Drive Singapore 637551 Singapore
- Department of Biological Sciences; National University of Singapore; 14 Science Drive 4 Singapore 117543 Singapore
| | - Laura Itzhaki
- Department of Pharmacology; Tennis Court Road Cambridge CB2 1PD UK
| | - Murray Stewart
- MRC Laboratory of Molecular Biology; Francis Crick Avenue, Cambridge Biomedical Campus Cambridge CB2 0QH UK
| | - James D. Brenton
- Cancer Research UK Cambridge Institute; University of Cambridge; Li Ka Shing Centre, Robinson Way Cambridge CB2 0RE UK
| | - David R. Spring
- Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| |
Collapse
|
164
|
Abstract
Peptide-based drug discovery has experienced a remarkable resurgence within the past decade due to the emerging class of inhibitors known as stapled peptides. Stapled peptides are therapeutic protein mimetics that have been locked within a specific conformational structure by hydrocarbon stapling. These peptides are highly important in selectively impairing disease-relevant protein–protein interactions and exhibit significant pharmacokinetic advantages over other forms of therapeutics in terms of affinity, specificity, size, synthetic accessibility and resistance to proteolytic degradation. A series of stapled peptides are currently in development, and the potential successes of these peptides, either as single-agent treatments or as combinational treatments with other therapeutic modalities, could potentially change the landscape of protein therapeutic development. Here, we provide examples of successful discovery efforts to illustrate the research strategies of stapled peptides in drug design and development.
Collapse
|
165
|
Villoutreix B. Combining bioinformatics, chemoinformatics and experimental approaches to design chemical probes: Applications in the field of blood coagulation. ANNALES PHARMACEUTIQUES FRANÇAISES 2016; 74:253-66. [PMID: 27133312 DOI: 10.1016/j.pharma.2016.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/21/2016] [Accepted: 03/21/2016] [Indexed: 11/08/2022]
|
166
|
Guo Z, Li B, Cheng LT, Zhou S, McCammon JA, Che J. Identification of protein-ligand binding sites by the level-set variational implicit-solvent approach. J Chem Theory Comput 2016; 11:753-65. [PMID: 25941465 PMCID: PMC4410907 DOI: 10.1021/ct500867u] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Indexed: 12/25/2022]
Abstract
![]()
Protein–ligand
binding is a key biological process at the
molecular level. The identification and characterization of small-molecule
binding sites on therapeutically relevant proteins have tremendous
implications for target evaluation and rational drug design. In this
work, we used the recently developed level-set variational implicit-solvent
model (VISM) with the Coulomb field approximation (CFA) to locate
and characterize potential protein–small-molecule binding sites.
We applied our method to a data set of 515 protein–ligand complexes
and found that 96.9% of the cocrystallized ligands bind to the VISM-CFA-identified
pockets and that 71.8% of the identified pockets are occupied by cocrystallized
ligands. For 228 tight-binding protein–ligand complexes (i.e,
complexes with experimental pKd values
larger than 6), 99.1% of the cocrystallized ligands are in the VISM-CFA-identified
pockets. In addition, it was found that the ligand binding orientations
are consistent with the hydrophilic and hydrophobic descriptions provided
by VISM. Quantitative characterization of binding pockets with topological
and physicochemical parameters was used to assess the “ligandability”
of the pockets. The results illustrate the key interactions between
ligands and receptors and can be very informative for rational drug
design.
Collapse
Affiliation(s)
- Zuojun Guo
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | | | | | | | | | | |
Collapse
|
167
|
Modell AE, Blosser SL, Arora PS. Systematic Targeting of Protein-Protein Interactions. Trends Pharmacol Sci 2016; 37:702-713. [PMID: 27267699 DOI: 10.1016/j.tips.2016.05.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/14/2016] [Accepted: 05/16/2016] [Indexed: 12/22/2022]
Abstract
Over the past decade, protein-protein interactions (PPIs) have gone from being neglected as 'undruggable' to being considered attractive targets for the development of therapeutics. Recent advances in computational analysis, fragment-based screening, and molecular design have revealed promising strategies to address the basic molecular recognition challenge: how to target large protein surfaces with specificity. Several systematic and complementary workflows have been developed to yield successful inhibitors of PPIs. Here we review the major contemporary approaches utilized for the discovery of inhibitors and focus on a structure-based workflow, from the selection of a biological target to design.
Collapse
Affiliation(s)
- Ashley E Modell
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Sarah L Blosser
- Department of Chemistry, New York University, New York, NY 10003, USA
| | - Paramjit S Arora
- Department of Chemistry, New York University, New York, NY 10003, USA
| |
Collapse
|
168
|
Sarkar D, Patra P, Ghosh A, Saha S. Computational Framework for Prediction of Peptide Sequences That May Mediate Multiple Protein Interactions in Cancer-Associated Hub Proteins. PLoS One 2016; 11:e0155911. [PMID: 27218803 PMCID: PMC4878775 DOI: 10.1371/journal.pone.0155911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 05/08/2016] [Indexed: 01/26/2023] Open
Abstract
A considerable proportion of protein-protein interactions (PPIs) in the cell are estimated to be mediated by very short peptide segments that approximately conform to specific sequence patterns known as linear motifs (LMs), often present in the disordered regions in the eukaryotic proteins. These peptides have been found to interact with low affinity and are able bind to multiple interactors, thus playing an important role in the PPI networks involving date hubs. In this work, PPI data and de novo motif identification based method (MEME) were used to identify such peptides in three cancer-associated hub proteins—MYC, APC and MDM2. The peptides corresponding to the significant LMs identified for each hub protein were aligned, the overlapping regions across these peptides being termed as overlapping linear peptides (OLPs). These OLPs were thus predicted to be responsible for multiple PPIs of the corresponding hub proteins and a scoring system was developed to rank them. We predicted six OLPs in MYC and five OLPs in MDM2 that scored higher than OLP predictions from randomly generated protein sets. Two OLP sequences from the C-terminal of MYC were predicted to bind with FBXW7, component of an E3 ubiquitin-protein ligase complex involved in proteasomal degradation of MYC. Similarly, we identified peptides in the C-terminal of MDM2 interacting with FKBP3, which has a specific role in auto-ubiquitinylation of MDM2. The peptide sequences predicted in MYC and MDM2 look promising for designing orthosteric inhibitors against possible disease-associated PPIs. Since these OLPs can interact with other proteins as well, these inhibitors should be specific to the targeted interactor to prevent undesired side-effects. This computational framework has been designed to predict and rank the peptide regions that may mediate multiple PPIs and can be applied to other disease-associated date hub proteins for prediction of novel therapeutic targets of small molecule PPI modulators.
Collapse
Affiliation(s)
| | - Piya Patra
- Maulana Abdul Kalam Azad University of Technology, Kolkata, India
| | - Abhirupa Ghosh
- Maulana Abdul Kalam Azad University of Technology, Kolkata, India
| | - Sudipto Saha
- Bioinformatics Centre, Bose Institute, Kolkata, India
- * E-mail: ;
| |
Collapse
|
169
|
Roskoski R. Cyclin-dependent protein kinase inhibitors including palbociclib as anticancer drugs. Pharmacol Res 2016; 107:249-275. [DOI: 10.1016/j.phrs.2016.03.012] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 02/07/2023]
|
170
|
Beekman AM, Howell LA. Small-Molecule and Peptide Inhibitors of the Pro-Survival Protein Mcl-1. ChemMedChem 2016; 11:802-13. [PMID: 26696548 PMCID: PMC4991272 DOI: 10.1002/cmdc.201500497] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/02/2015] [Indexed: 01/11/2023]
Abstract
The ability of protein-protein interactions to regulate cellular processes in both beneficial and detrimental ways has made them obvious drug targets. The Bcl-2 family of proteins undergo a series of protein-protein interactions which regulate the intrinsic cell-death pathway. The pro-survival members of the Bcl-2 family, including Bcl-2, Bcl-xL , and Mcl-1, are commonly overexpressed in a number of human cancers. Effective modulators of members of the Bcl-2 family have been developed and are undergoing clinical trials, but the efficient modulation of Mcl-1 is still not represented in the clinic. In addition, Mcl-1 is a major cause of resistance to radio- and chemotherapies, including inhibitors that target other Bcl-2 family members. Subsequently, the inhibition of Mcl-1 has become of significant interest to the scientific community. This review covers the progress made to date in modulating the activity of Mcl-1, by both stapled peptides and small molecules. The development of peptides as drug candidates, and the advancement of experimental and computational techniques used to discover small molecules are also highlighted.
Collapse
Affiliation(s)
- Andrew M Beekman
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR4 7TJ, UK
| | - Lesley A Howell
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR4 7TJ, UK.
| |
Collapse
|
171
|
Bayó-Puxan N, Rodríguez-Mias R, Goldflam M, Kotev M, Ciudad S, Hipolito CJ, Varese M, Suga H, Campos-Olivas R, Barril X, Guallar V, Teixidó M, García J, Giralt E. Combined Use of Oligopeptides, Fragment Libraries, and Natural Compounds: A Comprehensive Approach To Sample the Druggability of Vascular Endothelial Growth Factor. ChemMedChem 2016; 11:928-39. [PMID: 26553526 PMCID: PMC5063151 DOI: 10.1002/cmdc.201500467] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Indexed: 12/28/2022]
Abstract
The modulation of protein-protein interactions (PPIs) is emerging as a highly promising tool to fight diseases. However, whereas an increasing number of compounds are able to disrupt peptide-mediated PPIs efficiently, the inhibition of domain-domain PPIs appears to be much more challenging. Herein, we report our results related to the interaction between vascular endothelial growth factor (VEGF) and its receptor (VEGFR). The VEGF-VEGFR interaction is a typical domain-domain PPI that is highly relevant for the treatment of cancer and some retinopathies. Our final goal was to identify ligands able to bind VEGF at the region used by the growth factor to interact with its receptor. We undertook an extensive study, combining a variety of experimental approaches, including NMR-spectroscopy-based screening of small organic fragments, peptide libraries, and medicinal plant extracts. The key feature of the successful ligands that emerged from this study was their capacity to expose hydrophobic functional groups able to interact with the hydrophobic hot spots at the interacting VEGF surface patch.
Collapse
Affiliation(s)
- Núria Bayó-Puxan
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Ricard Rodríguez-Mias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Michael Goldflam
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Martin Kotev
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Sonia Ciudad
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Christopher J Hipolito
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, 113-8654, Japan
| | - Monica Varese
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, 113-8654, Japan
| | | | - Xavier Barril
- Department of Physical Chemistry, University of Barcelona, Barcelona, 08028, Spain
- The Institute of Biomedicine of the University of Barcelona, Barcelona, 08007, Spain
- Catalan Institution for Research and Advanced Studies, Barcelona, 08010, Spain
| | - Víctor Guallar
- Catalan Institution for Research and Advanced Studies, Barcelona, 08010, Spain
- Department of Life Sciences, Barcelona Supercomputing Center, Barcelona, 08034, Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Jesús García
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Ernest Giralt
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain.
- Department of Organic Chemistry, University of Barcelona, Barcelona, 08028, Spain.
| |
Collapse
|
172
|
Hain AUP, Miller AS, Levitskaya J, Bosch J. Virtual Screening and Experimental Validation Identify Novel Inhibitors of the Plasmodium falciparum Atg8-Atg3 Protein-Protein Interaction. ChemMedChem 2016; 11:900-10. [PMID: 26748931 PMCID: PMC8614111 DOI: 10.1002/cmdc.201500515] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 12/07/2015] [Indexed: 01/16/2023]
Abstract
New therapies are needed against malaria, a parasitic infection caused by Plasmodium falciparum, as drug resistance emerges against the current treatment, artemisinin. We previously characterized the Atg8-Atg3 protein-protein interaction (PPI), which is essential for autophagy and parasite survival. Herein we illustrate the use of virtual library screening to selectively block the PPI in the parasite without inhibiting the homologous interaction in humans by targeting the A-loop of PfAtg8. This A-loop is important for Atg3 binding in Plasmodium, but is absent from the human Atg8 homologues. In this proof-of-concept study, we demonstrate a shift in lipidation state of PfAtg8 and inhibition of P. falciparum growth in both blood- and liver-stage cultures upon drug treatment. Our results illustrate how in silico screening and structure-aided drug design against a PPI can be used to identify new hits for drug development. Additionally, as we targeted a region of Atg8 that is conserved within apicomplexans, we predict that our small molecule will have cross-reactivity against other disease-causing apicomplexans, such as Toxoplasma, Cryptosporidium, Theileria, Neospora, Eimeria, and Babesia.
Collapse
Affiliation(s)
- Adelaide U P Hain
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health and, Johns Hopkins Malaria Research Institute, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Alexia S Miller
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Jelena Levitskaya
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health and, Johns Hopkins Malaria Research Institute, 615 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
173
|
Wu Y, Olsen LB, Lau YH, Jensen CH, Rossmann M, Baker YR, Sore HF, Collins S, Spring DR. Development of a Multifunctional Benzophenone Linker for Peptide Stapling and Photoaffinity Labelling. Chembiochem 2016; 17:689-92. [PMID: 26919579 PMCID: PMC4862033 DOI: 10.1002/cbic.201500648] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Indexed: 01/18/2023]
Abstract
Photoaffinity labelling is a useful method for studying how proteins interact with ligands and biomolecules, and can help identify and characterise new targets for the development of new therapeutics. We present the design and synthesis of a novel multifunctional benzophenone linker that serves as both a photo-crosslinking motif and a peptide stapling reagent. Using double-click stapling, we attached the benzophenone to the peptide via the staple linker, rather than by modifying the peptide sequence with a photo-crosslinking amino acid. When applied to a p53-derived peptide, the resulting photoreactive stapled peptide was able to preferentially crosslink with MDM2 in the presence of competing protein. This multifunctional linker also features an extra alkyne handle for downstream applications such as pull-down assays, and can be used to investigate the target selectivity of stapled peptides.
Collapse
Affiliation(s)
- Yuteng Wu
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Lasse B Olsen
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Yu Heng Lau
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Claus Hatt Jensen
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Maxim Rossmann
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Ysobel R Baker
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Hannah F Sore
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Súil Collins
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - David R Spring
- University Chemical Laboratory, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
174
|
Smith EW, Nevins AM, Qiao Z, Liu Y, Getschman AE, Vankayala SL, Kemp MT, Peterson FC, Li R, Volkman BF, Chen Y. Structure-Based Identification of Novel Ligands Targeting Multiple Sites within a Chemokine-G-Protein-Coupled-Receptor Interface. J Med Chem 2016; 59:4342-51. [PMID: 27058821 DOI: 10.1021/acs.jmedchem.5b02042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CXCL12 is a human chemokine that recognizes the CXCR4 receptor and is involved in immune responses and metastatic cancer. Interactions between CXCL12 and CXCR4 are an important drug target but, like other elongated protein-protein interfaces, present challenges for small molecule ligand discovery due to the relatively shallow and featureless binding surfaces. Calculations using an NMR complex structure revealed a binding hot spot on CXCL12 that normally interacts with the I4/I6 residues from CXCR4. Virtual screening was performed against the NMR model, and subsequent testing has verified the specific binding of multiple docking hits to this site. Together with our previous results targeting two other binding pockets that recognize sulfotyrosine residues (sY12 and sY21) of CXCR4, including a new analog against the sY12 binding site reported herein, we demonstrate that protein-protein interfaces can often possess multiple sites for engineering specific small molecule ligands that provide lead compounds for subsequent optimization by fragment based approaches.
Collapse
Affiliation(s)
- Emmanuel W Smith
- Department of Molecular Medicine, University of South Florida , 12901 Bruce B. Downs Boulevard, Tampa, Florida 33612, United States
| | - Amanda M Nevins
- Department of Biochemistry, Medical College of Wisconsin , 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Zhen Qiao
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, and Cancer Genes and Molecular Regulation Program, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center , 986805 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yan Liu
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, and Cancer Genes and Molecular Regulation Program, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center , 986805 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin , 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Sai L Vankayala
- Department of Chemistry, University of South Florida , 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - M Trent Kemp
- Department of Chemistry, University of South Florida , 4202 East Fowler Avenue, Tampa, Florida 33620, United States
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin , 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Rongshi Li
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, and Cancer Genes and Molecular Regulation Program, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center , 986805 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin , 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Yu Chen
- Department of Molecular Medicine, University of South Florida , 12901 Bruce B. Downs Boulevard, Tampa, Florida 33612, United States
| |
Collapse
|
175
|
Aboalhaija NH, Zihlif MA, Taha MO. Discovery of new selective cytotoxic agents against Bcl-2 expressing cancer cells using ligand-based modeling. Chem Biol Interact 2016; 250:12-26. [PMID: 26954606 DOI: 10.1016/j.cbi.2016.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/28/2016] [Accepted: 03/02/2016] [Indexed: 12/11/2022]
|
176
|
Huang X, Dixit VM. Drugging the undruggables: exploring the ubiquitin system for drug development. Cell Res 2016; 26:484-98. [PMID: 27002218 PMCID: PMC4822129 DOI: 10.1038/cr.2016.31] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dynamic modulation of protein levels is tightly controlled in response to physiological cues. In mammalian cells, much of the protein degradation is carried out by the ubiquitin-proteasome system (UPS). Similar to kinases, components of the ubiquitin system are often dysregulated, leading to a variety of diseases, including cancer and neurodegeneration, making them attractive drug targets. However, so far there are only a handful of drugs targeting the ubiquitin system that have been approved by the FDA. Here, we review possible therapeutic intervention nodes in the ubiquitin system, analyze the challenges, and highlight the most promising strategies to target the UPS.
Collapse
Affiliation(s)
- Xiaodong Huang
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
177
|
Oo C, Kalbag SS. Leveraging the attributes of biologics and small molecules, and releasing the bottlenecks: a new wave of revolution in drug development. Expert Rev Clin Pharmacol 2016; 9:747-9. [DOI: 10.1586/17512433.2016.1160778] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Charles Oo
- SunLife Biopharma, Morris Plains, NJ, USA
| | | |
Collapse
|
178
|
Nim S, Jeon J, Corbi-Verge C, Seo MH, Ivarsson Y, Moffat J, Tarasova N, Kim PM. Pooled screening for antiproliferative inhibitors of protein-protein interactions. Nat Chem Biol 2016; 12:275-81. [PMID: 26900867 PMCID: PMC5756068 DOI: 10.1038/nchembio.2026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 01/04/2016] [Indexed: 11/30/2022]
Abstract
Protein-protein interactions (PPIs) are emerging as a promising new class of drug targets. Here, we present a novel high-throughput approach to screen inhibitors of PPIs in cells. We designed a library of 50,000 human peptide binding motifs and used a pooled lentiviral system to express them intracellularly and screen for their effects on cell proliferation. We thereby identified inhibitors that drastically reduced the viability of a pancreas cancer line (RWP1) while leaving a control line virtually unaffected. We identified their target interactions computationally, and validated a subset in experiments. We also discovered their potential mechanisms of action including apoptosis and cell cycle arrest. Finally, we confirmed that synthetic lipopeptide versions of our inhibitors have similarly specific and dosage dependent effects on cancer cell growth. Our screen reveals new drug targets and peptide drug leads and it provides a rich dataset covering phenotypes for inhibition of thousands of interactions.
Collapse
Affiliation(s)
- Satra Nim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Jouhyun Jeon
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Carles Corbi-Verge
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Moon-Hyeong Seo
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ylva Ivarsson
- Department of Chemistry, Biomedical Center (BMC), Uppsala University, Uppsala, Sweden
| | - Jason Moffat
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Nadya Tarasova
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Philip M Kim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
179
|
Borkin D, Pollock J, Kempinska K, Purohit T, Li X, Wen B, Zhao T, Miao H, Shukla S, He M, Sun D, Cierpicki T, Grembecka J. Property Focused Structure-Based Optimization of Small Molecule Inhibitors of the Protein-Protein Interaction between Menin and Mixed Lineage Leukemia (MLL). J Med Chem 2016; 59:892-913. [PMID: 26744767 PMCID: PMC5092235 DOI: 10.1021/acs.jmedchem.5b01305] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Development of potent small molecule inhibitors of protein-protein interactions with optimized druglike properties represents a challenging task in lead optimization process. Here, we report synthesis and structure-based optimization of new thienopyrimidine class of compounds, which block the protein-protein interaction between menin and MLL fusion proteins that plays an important role in acute leukemias with MLL translocations. We performed simultaneous optimization of both activity and druglike properties through systematic exploration of substituents introduced to the indole ring of lead compound 1 (MI-136) to identify compounds suitable for in vivo studies in mice. This work resulted in the identification of compound 27 (MI-538), which showed significantly increased activity, selectivity, polarity, and pharmacokinetic profile over 1 and demonstrated a pronounced effect in a mouse model of MLL leukemia. This study, which reports detailed structure-activity and structure-property relationships for the menin-MLL inhibitors, demonstrates challenges in optimizing inhibitors of protein-protein interactions for potential therapeutic applications.
Collapse
MESH Headings
- Animals
- Caco-2 Cells
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Histone-Lysine N-Methyltransferase/chemistry
- Histone-Lysine N-Methyltransferase/metabolism
- Humans
- Injections, Intraventricular
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Models, Molecular
- Molecular Structure
- Myeloid-Lymphoid Leukemia Protein/chemistry
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Protein Binding/drug effects
- Proto-Oncogene Proteins/chemistry
- Proto-Oncogene Proteins/metabolism
- Pyrimidines/administration & dosage
- Pyrimidines/chemistry
- Pyrimidines/pharmacology
- Small Molecule Libraries/administration & dosage
- Small Molecule Libraries/chemistry
- Small Molecule Libraries/pharmacology
- Structure-Activity Relationship
- Thiophenes/administration & dosage
- Thiophenes/chemistry
- Thiophenes/pharmacology
Collapse
Affiliation(s)
- Dmitry Borkin
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Jonathan Pollock
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Katarzyna Kempinska
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Trupta Purohit
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Xiaoqin Li
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ting Zhao
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hongzhi Miao
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Shirish Shukla
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Miao He
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
180
|
An Intriguing Correlation Based on the Superimposition of Residue Pairs with Inhibitors that Target Protein-Protein Interfaces. Sci Rep 2016; 6:18543. [PMID: 26730437 PMCID: PMC4698585 DOI: 10.1038/srep18543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/19/2015] [Indexed: 11/26/2022] Open
Abstract
Druggable sites on protein-protein interfaces are difficult to predict. To survey inhibitor-binding sites onto which residues are superimposed at protein-protein interfaces, we analyzed publicly available information for 39 inhibitors that target the protein-protein interfaces of 8 drug targets. By focusing on the differences between residues that were superimposed with inhibitors and non-superimposed residues, we observed clear differences in the distances and changes in the solvent-accessible surface areas (∆SASA). Based on the observation that two or more residues were superimposed onto inhibitors in 37 (95%) of 39 protein-inhibitor complexes, we focused on the two-residue relationships. Application of a cross-validation procedure confirmed a linear negative correlation between the absolute value of the dihedral angle and the sum of the ∆SASAs of the residues. Finally, we applied the regression equation of this correlation to four inhibitors that bind to new sites not bound by the 39 inhibitors as well as additional inhibitors of different targets. Our results shed light on the two-residue correlation between the absolute value of the dihedral angle and the sum of the ∆SASA, which may be a useful relationship for identifying the key two-residues as potential targets of protein-protein interfaces.
Collapse
|
181
|
K. Comfort K. The rise of nanotoxicology: A successful collaboration between engineering and biology. AIMS BIOENGINEERING 2016. [DOI: 10.3934/bioeng.2016.3.230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
182
|
Hu G, Xiao F, Li Y, Li Y, Vongsangnak W. Protein-Protein Interface and Disease: Perspective from Biomolecular Networks. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 160:57-74. [PMID: 27928579 DOI: 10.1007/10_2016_40] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protein-protein interactions are involved in many important biological processes and molecular mechanisms of disease association. Structural studies of interfacial residues in protein complexes provide information on protein-protein interactions. Characterizing protein-protein interfaces, including binding sites and allosteric changes, thus pose an imminent challenge. With special focus on protein complexes, approaches based on network theory are proposed to meet this challenge. In this review we pay attention to protein-protein interfaces from the perspective of biomolecular networks and their roles in disease. We first describe the different roles of protein complexes in disease through several structural aspects of interfaces. We then discuss some recent advances in predicting hot spots and communication pathway analysis in terms of amino acid networks. Finally, we highlight possible future aspects of this area with respect to both methodology development and applications for disease treatment.
Collapse
Affiliation(s)
- Guang Hu
- Center for Systems Biology, School of Electronic and Information Engineering, Soochow University, Suzhou, 215006, China.
| | - Fei Xiao
- School of Basic Medicine and Biological Sciences, Medical College of Soochow University, Suzhou, 215123, China
| | - Yuqian Li
- School of Electronic Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuan Li
- Center for Systems Biology, School of Electronic and Information Engineering, Soochow University, Suzhou, 215006, China
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand.
- Computational Biomodelling Laboratory for Agricultural Science and Technology (CBLAST), Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
183
|
Stucchi M, Grazioso G, Lammi C, Manara S, Zanoni C, Arnoldi A, Lesma G, Silvani A. Disrupting the PCSK9/LDLR protein–protein interaction by an imidazole-based minimalist peptidomimetic. Org Biomol Chem 2016; 14:9736-9740. [DOI: 10.1039/c6ob01642a] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report on a tetraimidazole-based β-strand minimalist peptidomimetic as a novel inhibitor of LDLR–PCSK9 protein–protein interaction, a promising target for hypercholesterolemia.
Collapse
Affiliation(s)
- Mattia Stucchi
- Dipartimento di Chimica
- Università degli Studi di Milano
- 20133 Milano
- Italy
- Dipartimento di Scienze della Vita
| | - Giovanni Grazioso
- Dipartimento di Scienze Farmaceutiche
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - Carmen Lammi
- Dipartimento di Scienze Farmaceutiche
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - Silvia Manara
- Dipartimento di Scienze Farmaceutiche
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - Chiara Zanoni
- Dipartimento di Scienze Farmaceutiche
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - Anna Arnoldi
- Dipartimento di Scienze Farmaceutiche
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - Giordano Lesma
- Dipartimento di Chimica
- Università degli Studi di Milano
- 20133 Milano
- Italy
| | - Alessandra Silvani
- Dipartimento di Chimica
- Università degli Studi di Milano
- 20133 Milano
- Italy
| |
Collapse
|
184
|
Callies O, Hernández Daranas A. Application of isothermal titration calorimetry as a tool to study natural product interactions. Nat Prod Rep 2016; 33:881-904. [DOI: 10.1039/c5np00094g] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The study of molecular interactions of natural products by isothermal titration calorimetry (ITC) is a potent tool to get new insights of the underpinning driving forces.
Collapse
Affiliation(s)
- O. Callies
- Institute of Bioorganic Chemistry “Antonio González”
- Center for Biomedical Research of the Canary Islands
- University of La Laguna
- 38206 La Laguna
- Spain
| | - A. Hernández Daranas
- Institute of Bioorganic Chemistry “Antonio González”
- Center for Biomedical Research of the Canary Islands
- University of La Laguna
- 38206 La Laguna
- Spain
| |
Collapse
|
185
|
Winter A, Sigurdardottir AG, DiCara D, Valenti G, Blundell TL, Gherardi E. Developing Antagonists for the Met-HGF/SF Protein–Protein Interaction Using a Fragment-Based Approach. Mol Cancer Ther 2015; 15:3-14. [DOI: 10.1158/1535-7163.mct-15-0446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/29/2015] [Indexed: 11/16/2022]
|
186
|
Yeh JE, Frank DA. STAT3-Interacting Proteins as Modulators of Transcription Factor Function: Implications to Targeted Cancer Therapy. ChemMedChem 2015; 11:795-801. [DOI: 10.1002/cmdc.201500482] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/01/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Jennifer E. Yeh
- Department of Medical Oncology; Dana-Farber Cancer Institute; 450 Brookline Avenue Boston MA 02215 USA
| | - David A. Frank
- Department of Medical Oncology; Dana-Farber Cancer Institute; 450 Brookline Avenue Boston MA 02215 USA
| |
Collapse
|
187
|
Parvatkar P, Kato N, Uesugi M, Sato SI, Ohkanda J. Intracellular Generation of a Diterpene-Peptide Conjugate that Inhibits 14-3-3-Mediated Interactions. J Am Chem Soc 2015; 137:15624-7. [PMID: 26632868 DOI: 10.1021/jacs.5b09817] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Synthetic agents that disrupt intracellular protein-protein interactions (PPIs) are highly desirable for elucidating signaling networks and developing new therapeutics. However, designing cell-penetrating large molecules equipped with the many functional groups necessary for binding to large interfaces remains challenging. Here, we describe a rational strategy for the intracellular oxime ligation-mediated generation of an amphipathic bivalent inhibitor composed of a peptide and diterpene natural product, fusicoccin, which binds 14-3-3 protein with submicromolar affinity. Our results demonstrate that co-treatment of cells with small module molecules, the aldehyde-containing fusicoccin 1 and the aminooxy-containing peptide 2, generates the corresponding conjugate 3 in cells, resulting in significant cytotoxicity. In contrast, chemically synthesized 3 is not cytotoxic, likely due to its inability to penetrate cells. Compound 3, but not 1 or 2, disrupts endogenous 14-3-3/cRaf interactions, suggesting that cell death is caused by inhibition of 14-3-3 activity. These results suggest that intracellular generation of large-sized molecules may serve as a new approach for modulating PPIs.
Collapse
Affiliation(s)
- Prakash Parvatkar
- Institute for Chemical Research and Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Gokasho, Uji, Kyoto 611-0011, Japan
| | - Nobuo Kato
- Institute of Scientific and Industrial Research, Osaka University , 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Motonari Uesugi
- Institute for Chemical Research and Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Gokasho, Uji, Kyoto 611-0011, Japan
| | - Shin-Ichi Sato
- Institute for Chemical Research and Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Gokasho, Uji, Kyoto 611-0011, Japan
| | - Junko Ohkanda
- Institute for Chemical Research and Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Gokasho, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
188
|
Nevola L, Giralt E. Modulating protein-protein interactions: the potential of peptides. Chem Commun (Camb) 2015; 51:3302-15. [PMID: 25578807 DOI: 10.1039/c4cc08565e] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Protein-protein interactions (PPIs) have emerged as important and challenging targets in chemical biology and medicinal chemistry. The main difficulty encountered in the discovery of small molecule modulators derives from the large contact surfaces involved in PPIs when compared with those that participate in protein-small molecule interactions. Because of their intrinsic features, peptides can explore larger surfaces and therefore represent a useful alternative to modulate PPIs. The use of peptides as therapeutics has been held back by their instability in vivo and poor cell internalization. However, more than 200 peptide drugs and homologous compounds (proteins or antibodies) containing peptide bonds are (or have been) on the market, and many alternatives are now available to tackle these limitations. This review will focus on the latest progress in the field, spanning from "lead" identification methods to binding evaluation techniques, through an update of the most successful examples described in the literature.
Collapse
Affiliation(s)
- Laura Nevola
- Institute for Research in Biomedicine (IRB Barcelona), C/Baldiri Reixac 10, 08028 Barcelona, Spain.
| | | |
Collapse
|
189
|
Kanthala S, Pallerla S, Jois S. Current and future targeted therapies for non-small-cell lung cancers with aberrant EGF receptors. Future Oncol 2015; 11:865-78. [PMID: 25757687 DOI: 10.2217/fon.14.312] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Expression of the EGF receptors (EGFRs) is abnormally high in many types of cancer, including 25% of lung cancers. Successful treatments target mutations in the EGFR tyrosine kinase domain with EGFR tyrosine kinase inhibitors (TKIs). However, almost all patients develop resistance to this treatment, and acquired resistance to first-generation TKI has prompted the clinical development of a second generation of EGFR TKI. Because of the development of resistance to treatment of TKIs, there is a need to collect genomic information about EGFR levels in non-small-cell lung cancer patients. Herein, we focus on current molecular targets that have therapies available as well as other targets for which therapies will be available in the near future.
Collapse
Affiliation(s)
- Shanthi Kanthala
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | | | | |
Collapse
|
190
|
Yu X, Guttenberger N, Fuchs E, Peters M, Weber H, Breinbauer R. Diversity-Oriented Synthesis of a Library of Star-Shaped 2H-Imidazolines. ACS COMBINATORIAL SCIENCE 2015; 17:682-90. [PMID: 26402035 DOI: 10.1021/acscombsci.5b00107] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A library of star-shaped 2H-imidazolines has been synthesized via Debus-Radziszewski condensation from 1,2-diketones and ketone starting materials. Selective reduction of one imine group of the 2H-imidazole intermediate with LiAlH4 or catalytic flow hydrogenation furnished 2H-imidazolines, which could be conveniently diversified by reacting the amine N with electrophiles, resulting in a set of 21 amide-, carbamate-, urea-, and allylamine-containing products. In total, five points of diversification could be used, which allow the production of a set of functionally diverse compounds. The synthesis of acylated 2H-imidazolidines resulted in intrinsically labile compounds, which spontaneously degraded to acyclic derivatives, as shown for the reaction of 2H-imidazolidine with hexylisocyanate.
Collapse
Affiliation(s)
- Xuepu Yu
- Institute of Organic Chemistry, Graz University of Technology, A-8010 Graz, Austria
| | | | - Elisabeth Fuchs
- Institute of Organic Chemistry, Graz University of Technology, A-8010 Graz, Austria
| | - Martin Peters
- Institute of Organic Chemistry, Graz University of Technology, A-8010 Graz, Austria
| | - Hansjörg Weber
- Institute of Organic Chemistry, Graz University of Technology, A-8010 Graz, Austria
| | - Rolf Breinbauer
- Institute of Organic Chemistry, Graz University of Technology, A-8010 Graz, Austria
| |
Collapse
|
191
|
Doak BC, Zheng J, Dobritzsch D, Kihlberg J. How Beyond Rule of 5 Drugs and Clinical Candidates Bind to Their Targets. J Med Chem 2015; 59:2312-27. [DOI: 10.1021/acs.jmedchem.5b01286] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Bradley C. Doak
- Department of Chemistry—BMC, Uppsala University, Box
576, SE-751 23 Uppsala, Sweden
| | - Jie Zheng
- Department of Chemistry—BMC, Uppsala University, Box
576, SE-751 23 Uppsala, Sweden
| | - Doreen Dobritzsch
- Department of Chemistry—BMC, Uppsala University, Box
576, SE-751 23 Uppsala, Sweden
| | - Jan Kihlberg
- Department of Chemistry—BMC, Uppsala University, Box
576, SE-751 23 Uppsala, Sweden
| |
Collapse
|
192
|
Fernández A, Scott LR. Drug leads for interactive protein targets with unknown structure. Drug Discov Today 2015; 21:531-5. [PMID: 26484433 DOI: 10.1016/j.drudis.2015.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/29/2015] [Accepted: 10/12/2015] [Indexed: 11/24/2022]
Abstract
The disruption of protein-protein interfaces (PPIs) remains a challenge in drug discovery. The problem becomes daunting when the structure of the target protein is unknown and is even further complicated when the interface is susceptible to disruptive phosphorylation. Based solely on protein sequence and information about phosphorylation-susceptible sites within the PPI, a new technology has been developed to identify drug leads to inhibit protein associations. Here we reveal this technology and contrast it with current structure-based technologies for the generation of drug leads. The novel technology is illustrated by a patented invention to treat heart failure. The success of this technology shows that it is possible to generate drug leads in the absence of target structure.
Collapse
Affiliation(s)
- Ariel Fernández
- Argentine Institute of Mathematics (IAM), National Research Council (CONICET), Buenos Aires 1083, Argentina; AF Innovation, Avenida del Libertador 1092, Buenos Aires 1112, Argentina.
| | - L Ridgway Scott
- Department of Computer Science, The University of Chicago, Chicago, IL 60637, USA; Department of Mathematics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
193
|
Loh J, Asad N, Samarakoon TB, Hanson PR. Modular, One-Pot, Sequential Aziridine Ring Opening-S(N)Ar Strategy to 7-, 10-, and 11-Membered Benzo-Fused Sultams. J Org Chem 2015; 80:9926-41. [PMID: 26446396 PMCID: PMC4943336 DOI: 10.1021/acs.joc.5b01429] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Indexed: 12/12/2022]
Abstract
The generation of common and stereochemically rich medium-sized benzo-fused sultams via complementary pairing of heretofore-unknown (o-fluoroaryl)sulfonyl aziridine building blocks with an array of amino alcohols/amines in a modular one-pot, sequential protocol using an aziridine ring opening and intramolecular nucleophilic aromatic substitution is reported. The strategy employs a variety of amino alcohols/amines and proceeds with 6 + 4/6 + 5 and 6 + 1 cycloetherification pathways in a highly chemo- and regioselective fashion to obtain skeletally and structurally diverse, polycyclic, 10- to 11- and 7-membered benzo-fused sultams for broad-scale screening.
Collapse
Affiliation(s)
- Joanna
K. Loh
- Department of Chemistry, University
of Kansas, 1251 Wescoe
Hall Drive, Lawrence, Kansas 66045, United
States
- Center
for Chemical Methodologies
and Library Development (KU-CMLD), Delbert M. Shankel Structural Biology
Center, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Naeem Asad
- Department of Chemistry, University
of Kansas, 1251 Wescoe
Hall Drive, Lawrence, Kansas 66045, United
States
- Center
for Chemical Methodologies
and Library Development (KU-CMLD), Delbert M. Shankel Structural Biology
Center, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Thiwanka B. Samarakoon
- Department of Chemistry, University
of Kansas, 1251 Wescoe
Hall Drive, Lawrence, Kansas 66045, United
States
- Center
for Chemical Methodologies
and Library Development (KU-CMLD), Delbert M. Shankel Structural Biology
Center, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | | |
Collapse
|
194
|
Giorgio C, Russo S, Incerti M, Bugatti A, Vacondio F, Barocelli E, Mor M, Pala D, Hassan-Mohamed I, Gioiello A, Rusnati M, Lodola A, Tognolini M. Biochemical characterization of EphA2 antagonists with improved physico-chemical properties by cell-based assays and surface plasmon resonance analysis. Biochem Pharmacol 2015; 99:18-30. [PMID: 26462575 DOI: 10.1016/j.bcp.2015.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/06/2015] [Indexed: 12/28/2022]
Abstract
Amino acid conjugates of lithocholic acid (LCA) have been recently described as effective disruptors of the EphA2-ephrin-A1 interaction able to inhibit EphA2 phosphorylation in intact cells and thus able to block prometastatic responses such as cellular retraction and angiogenesis. However, these LCA-based compounds were significantly more potent at disrupting the EphA2-ephrin-A1 interaction than at blocking phenotype responses in cells, which might reflect an unclear mechanism of action or a metabolic issue responsible for a reduction of the compound concentration at the cell's surface. Through the synthesis of new compounds and their examination by a combination of cell-based assays and real-time interaction analysis by surface plasmon resonance, we showed at molecular level that l-tryptophan conjugates of lithocholic acid disrupt EphA2-ephrin-A1 interaction by targeting the EphA 2 receptor and that the presence of a polar group in position 3 of steroid scaffold is a key factor to increase the effective concentration of the compounds in cancer cell lines.
Collapse
Affiliation(s)
- Carmine Giorgio
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| | - Simonetta Russo
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| | - Matteo Incerti
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| | - Antonella Bugatti
- Dipartimento di Medicina Molecolare Traslazionale, Università degli Studi di Brescia, Brescia, Italy
| | - Federica Vacondio
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| | | | - Marco Mor
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| | - Daniele Pala
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| | | | - Antimo Gioiello
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, Perugia, Italy
| | - Marco Rusnati
- Dipartimento di Medicina Molecolare Traslazionale, Università degli Studi di Brescia, Brescia, Italy
| | - Alessio Lodola
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy; Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, UK.
| | | |
Collapse
|
195
|
Dailing A, Luchini A, Liotta L. Unlocking the secrets to protein-protein interface drug targets using structural mass spectrometry techniques. Expert Rev Proteomics 2015; 12:457-67. [PMID: 26400464 DOI: 10.1586/14789450.2015.1079487] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Protein-protein interactions (PPIs) drive all biologic systems at the subcellular and extracellular level. Changes in the specificity and affinity of these interactions can lead to cellular malfunctions and disease. Consequently, the binding interfaces between interacting protein partners are important drug targets for the next generation of therapies that block such interactions. Unfortunately, protein-protein contact points have proven to be very difficult pharmacological targets because they are hidden within complex 3D interfaces. For the vast majority of characterized binary PPIs, the specific amino acid sequence of their close contact regions remains unknown. There has been an important need for an experimental technology that can rapidly reveal the functionally important contact points of native protein complexes in solution. In this review, experimental techniques employing mass spectrometry to explore protein interaction binding sites are discussed. Hydrogen-deuterium exchange, hydroxyl radical footprinting, crosslinking and the newest technology protein painting are compared and contrasted.
Collapse
Affiliation(s)
| | - Alessandra Luchini
- a Center for Applied Proteomics and Molecular Medicine, Life Sciences Lab Building, George Mason University, 10920 University Boulevard, Manassas, Virginia 20110, USA
| | - Lance Liotta
- a Center for Applied Proteomics and Molecular Medicine, Life Sciences Lab Building, George Mason University, 10920 University Boulevard, Manassas, Virginia 20110, USA
| |
Collapse
|
196
|
Bartolowits M, Davisson VJ. Considerations of Protein Subpockets in Fragment-Based Drug Design. Chem Biol Drug Des 2015; 87:5-20. [PMID: 26307335 DOI: 10.1111/cbdd.12631] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While the fragment-based drug design approach continues to gain importance, gaps in the tools and methods available in the identification and accurate utilization of protein subpockets have limited the scope. The importance of these features of small molecule-protein recognition is highlighted with several examples. A generalized solution for the identification of subpockets and corresponding chemical fragments remains elusive, but there are numerous advancements in methods that can be used in combination to address subpockets. Finally, additional examples of approaches that consider the relative importance of small-molecule co-dependence of protein conformations are highlighted to emphasize an increased significance of subpockets, especially at protein interfaces.
Collapse
Affiliation(s)
- Matthew Bartolowits
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47907, USA
| | - V Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47907, USA
| |
Collapse
|
197
|
Cierpicki T, Grembecka J. Targeting protein-protein interactions in hematologic malignancies: still a challenge or a great opportunity for future therapies? Immunol Rev 2015; 263:279-301. [PMID: 25510283 DOI: 10.1111/imr.12244] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over the past several years, there has been an increasing research effort focused on inhibition of protein-protein interactions (PPIs) to develop novel therapeutic approaches for cancer, including hematologic malignancies. These efforts have led to development of small molecule inhibitors of PPIs, some of which already advanced to the stage of clinical trials while others are at different stages of preclinical optimization, emphasizing PPIs as an emerging and attractive class of drug targets. Here, we review several examples of recently developed inhibitors of PPIs highly relevant to hematologic cancers. We address the existing skepticism about feasibility of targeting PPIs and emphasize potential therapeutic benefit from blocking PPIs in hematologic malignancies. We then use these examples to discuss the approaches for successful identification of PPI inhibitors and provide analysis of the protein-protein interfaces, with the goal to address 'druggability' of new PPIs relevant to hematology. We discuss lessons learned to improve the success of targeting new PPIs and evaluate prospects and limits of the research in this field. We conclude that not all PPIs are equally tractable for blocking by small molecules, and detailed analysis of PPI interfaces is critical for selection of those with the highest chance of success. Together, our analysis uncovers patterns that should help to advance drug discovery in hematologic malignancies by successful targeting of new PPIs.
Collapse
Affiliation(s)
- Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
198
|
Rooklin D, Wang C, Katigbak J, Arora PS, Zhang Y. AlphaSpace: Fragment-Centric Topographical Mapping To Target Protein-Protein Interaction Interfaces. J Chem Inf Model 2015. [PMID: 26225450 PMCID: PMC4550072 DOI: 10.1021/acs.jcim.5b00103] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Inhibition
of protein–protein interactions (PPIs) is emerging
as a promising therapeutic strategy despite the difficulty in targeting
such interfaces with drug-like small molecules. PPIs generally feature
large and flat binding surfaces as compared to typical drug targets.
These features pose a challenge for structural characterization of
the surface using geometry-based pocket-detection methods. An attractive
mapping strategy—that builds on the principles of fragment-based
drug discovery (FBDD)—is to detect the fragment-centric modularity
at the protein surface and then characterize the large PPI interface
as a set of localized, fragment-targetable interaction regions. Here,
we introduce AlphaSpace, a computational analysis tool designed for
fragment-centric topographical mapping (FCTM) of PPI interfaces. Our
approach uses the alpha sphere construct, a geometric feature of a
protein’s Voronoi diagram, to map out concave interaction space
at the protein surface. We introduce two new features—alpha-atom
and alpha-space—and the concept of the alpha-atom/alpha-space
pair to rank pockets for fragment-targetability and to facilitate
the evaluation of pocket/fragment complementarity. The resulting high-resolution
interfacial map of targetable pocket space can be used to guide the
rational design and optimization of small molecule or biomimetic PPI
inhibitors.
Collapse
Affiliation(s)
- David Rooklin
- Department of Chemistry, New York University , New York, New York 10003, United States
| | - Cheng Wang
- Department of Chemistry, New York University , New York, New York 10003, United States
| | - Joseph Katigbak
- Department of Chemistry, New York University , New York, New York 10003, United States
| | - Paramjit S Arora
- Department of Chemistry, New York University , New York, New York 10003, United States
| | - Yingkai Zhang
- Department of Chemistry, New York University , New York, New York 10003, United States.,NYU-ECNU Center for Computational Chemistry at NYU Shanghai , Shanghai 200062, China
| |
Collapse
|
199
|
Raffeiner P, Röck R, Schraffl A, Hartl M, Hart JR, Janda KD, Vogt PK, Stefan E, Bister K. In vivo quantification and perturbation of Myc-Max interactions and the impact on oncogenic potential. Oncotarget 2015; 5:8869-78. [PMID: 25326649 PMCID: PMC4253403 DOI: 10.18632/oncotarget.2588] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The oncogenic bHLH-LZ transcription factor Myc forms binary complexes with its binding partner Max. These and other bHLH-LZ-based protein-protein interactions (PPI) in the Myc-Max network are essential for the physiological and oncogenic activities of Myc. We have generated a genetically determined and highly specific protein-fragment complementation assay based on Renilla luciferase to analyze the dynamic interplay of bHLH-LZ transcription factors Myc, Max, and Mxd1 in vivo. We also applied this PPI reporter to quantify alterations of nuclear Myc-Max complexes in response to mutational events, competitive binding by the transcriptional repressor Mxd1, or perturbations by small-molecule Myc inhibitors, including recently identified potent PPI inhibitors from a Kröhnke pyridine library. We show that the specificity of Myc-Max PPI reduction by the pyridine inhibitors directly correlates with their efficient and highly specific potential to interfere with the proliferation of human and avian tumor cells displaying deregulated Myc expression. In a direct comparison with known Myc inhibitors using human and avian cell systems, the pyridine compounds reveal a unique inhibitory potential even at sub-micromolar concentrations combined with remarkable specificity for the inhibition of Myc-driven tumor cell proliferation. Furthermore, we show in direct comparisons using defined avian cell systems that different Max PPI profiles for the variant members of the Myc protein family (c-Myc, v-Myc, N-Myc, L-Myc) correlate with their diverse oncogenic potential and their variable sensitivity to the novel pyridine inhibitors.
Collapse
Affiliation(s)
- Philipp Raffeiner
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Ruth Röck
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Andrea Schraffl
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Markus Hartl
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Jonathan R Hart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - Peter K Vogt
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Klaus Bister
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
200
|
Sheng C, Dong G, Miao Z, Zhang W, Wang W. State-of-the-art strategies for targeting protein-protein interactions by small-molecule inhibitors. Chem Soc Rev 2015; 44:8238-59. [PMID: 26248294 DOI: 10.1039/c5cs00252d] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Targeting protein-protein interactions (PPIs) has emerged as a viable approach in modern drug discovery. However, the identification of small molecules enabling us to effectively interrupt their interactions presents significant challenges. In the recent past, significant advances have been made in the development of new biological and chemical strategies to facilitate the discovery process of small-molecule PPI inhibitors. This review aims to highlight the state-of-the-art technologies and the achievements made recently in this field. The "hot spots" of PPIs have been proved to be critical for small molecules to bind. Three strategies including screening, designing, and synthetic approaches have been explored for discovering PPI inhibitors by targeting the "hot spots". Although the classic high throughput screening approach can be used, fragment screening, fragment-based drug design and newly improved virtual screening are demonstrated to be more effective in the discovery of PPI inhibitors. In addition to screening approaches, design strategies including anchor-based and small molecule mimetics of secondary structures involved in PPIs have become powerful tools as well. Finally, constructing new chemically spaced libraries with high diversity and complexity is becoming an important area of interest for PPI inhibitors. The successful cases from the recent five year studies are used to illustrate how these approaches are implemented to uncover and optimize small molecule PPI inhibitors and notably some of them have become promising therapeutics.
Collapse
Affiliation(s)
- Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, P. R. China.
| | | | | | | | | |
Collapse
|