1
|
Yoo J, Choi Y, Kim H, Park SB. Revisiting Pyrimidine-Embedded Molecular Frameworks to Probe the Unexplored Chemical Space for Protein-Protein Interactions. Acc Chem Res 2024; 57:3254-3265. [PMID: 39480992 PMCID: PMC11580176 DOI: 10.1021/acs.accounts.4c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/19/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024]
Abstract
ConspectusProtein-protein interactions (PPIs) are essential in numerous biological processes and diseases, making them attractive yet challenging drug targets. While many advances have been made in traditional drug discovery, targeting PPIs has been difficult due to a lack of specialized chemical libraries designed to modulate these interactions. Current libraries mainly focus on conventional target proteins like enzymes or receptors as substrate analogs rather than small-molecule modulators targeting PPIs. These traditional drug targets behave differently from PPIs. Conventional druggable targets have relatively small surfaces and binding pockets that have allowed them to be targeted with current libraries, but PPIs behave differently than these traditional drug targets. As a result, there is an urgent need for an innovative approach to expand the druggable space.To address this, we developed a privileged substructure-based diversity-oriented synthesis (pDOS) strategy, aimed at creating maximal skeletal diversity to explore broader biochemical space. Pyrimidine serves as the privileged substructure in our approach, which employs several strategies: (i) silver-catalyzed or iodine-mediated tandem cyclizations to generate pyrimidine-embedded polyheterocycles; (ii) diverse pairing strategies to produce pyrimidodiazepine-containing polyheterocyclic skeletons with enhanced scaffold saturation; (iii) skeletal transformation to develop pyrimidine-fused medium-sized azacycles via chemoselective cleavages or migrations of N-N or C-N bond; (iv) design of small-molecule peptidomimetics that systematically mimic three pivotal protein secondary structures using pyrimidodiazepine-based scaffolds; and (v) identification of pyrimidodiazepine-based small-molecules that allosterically inhibits the interaction between human ACE2 and the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein to block viral entry into host cells.Through these approaches, we generated 39 distinct pyrimidine-embedded frameworks, demonstrating significant molecular diversity validated by chemoinformatic analyses such as Tanimoto similarity and principal moment of inertia (PMI) analysis. This molecular diversity extends pyrimidine structures beyond traditional linear or bicyclic forms, creating polyheterocycles with enhanced 3D structural diversity. These novel frameworks overcome the limitation of simpler privileged scaffolds, offering promising tools for modulating PPIs.Our pDOS approach highlights how privileged structure-embedded polyheterocycles, particularly those based on pyrimidine, can effectively target previously undruggable PPIs. This strategy provides a new direction for drug discovery, allowing for the development of small molecules that operate beyond traditional drug-like rules. In addition to expanding the chemical space for PPI modulation, our pDOS strategy enables the creation of scaffolds that are particularly suited for targeting complex and dynamic protein interfaces. This innovation could significantly impact therapeutic development, offering solutions for previously intractable drug targets. By expanding the scope of pyrimidine-based scaffolds, we have opened up new possibilities for targeting PPIs and advancing chemical biology.This perspective demonstrates the potential outlines of our pDOS strategy in creating structurally diverse frameworks, offering a platform for the discovery of PPI modulators and facilitating the exploration of untapped biochemical spaces in drug development, potentially transforming the way we approach these complex biological interactions.
Collapse
Affiliation(s)
- Jeong
Yeon Yoo
- Department of Chemistry, Seoul
National University, Seoul 08826, Korea (South)
| | - Yoona Choi
- Department of Chemistry, Seoul
National University, Seoul 08826, Korea (South)
| | - Heejun Kim
- Department of Chemistry, Seoul
National University, Seoul 08826, Korea (South)
| | - Seung Bum Park
- Department of Chemistry, Seoul
National University, Seoul 08826, Korea (South)
| |
Collapse
|
2
|
Broad-Spectrum Small-Molecule Inhibitors of the SARS-CoV-2 Spike-ACE2 Protein-Protein Interaction from a Chemical Space of Privileged Protein Binders. Pharmaceuticals (Basel) 2022; 15:ph15091084. [PMID: 36145305 PMCID: PMC9504289 DOI: 10.3390/ph15091084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022] Open
Abstract
Therapeutically useful small-molecule inhibitors (SMIs) of protein−protein interactions (PPIs) initiating the cell attachment and entry of viruses could provide novel alternative antivirals that act via mechanisms similar to that of neutralizing antibodies but retain the advantages of small-molecule drugs such as oral bioavailability and low likelihood of immunogenicity. From screening our library, which is focused around the chemical space of organic dyes to provide good protein binders, we have identified several promising SMIs of the SARS-CoV-2 spike—ACE2 interaction, which is needed for the attachment and cell entry of this coronavirus behind the COVID-19 pandemic. They included organic dyes, such as Congo red, direct violet 1, and Evans blue, which seem to be promiscuous PPI inhibitors, as well as novel drug-like compounds (e.g., DRI-C23041). Here, we show that in addition to the original SARS-CoV-2 strain, these SMIs also inhibit this PPI for variants of concern including delta (B.1.617.2) and omicron (B.1.1.529) as well as HCoV-NL63 with low- or even sub-micromolar activity. They also concentration-dependently inhibited SARS-CoV-2-S expressing pseudovirus entry into hACE2-expressing cells with low micromolar activity (IC50 < 10 μM) both for the original strain and the delta variant. DRI-C23041 showed good therapeutic (selectivity) index, i.e., separation between activity and cytotoxicity (TI > 100). Specificities and activities require further optimization; nevertheless, these results provide a promising starting point toward novel broad-spectrum small-molecule antivirals that act via blocking the interaction between the spike proteins of coronaviruses and their ACE2 receptor initiating cellular entry.
Collapse
|
3
|
Bojadzic D, Alcazar O, Chen J, Chuang ST, Capcha JMC, Shehadeh LA, Buchwald P. Small-Molecule Inhibitors of the Coronavirus Spike: ACE2 Protein-Protein Interaction as Blockers of Viral Attachment and Entry for SARS-CoV-2. ACS Infect Dis 2021; 7:1519-1534. [PMID: 33979123 PMCID: PMC8130611 DOI: 10.1021/acsinfecdis.1c00070] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Inhibitors of the protein-protein interaction (PPI) between the SARS-CoV-2 spike protein and human ACE2 (hACE2), which acts as a ligand-receptor pair that initiates the viral attachment and cellular entry of this coronavirus causing the ongoing COVID-19 pandemic, are of considerable interest as potential antiviral agents. While blockade of such PPIs with small molecules is more challenging than that with antibodies, small-molecule inhibitors (SMIs) might offer alternatives that are less strain- and mutation-sensitive, suitable for oral or inhaled administration, and more controllable/less immunogenic. Here, we report the identification of SMIs of this PPI by screening our compound library focused around the chemical space of organic dyes. Among promising candidates identified, several dyes (Congo red, direct violet 1, Evans blue) and novel druglike compounds (DRI-C23041, DRI-C91005) inhibited the interaction of hACE2 with the spike proteins of SARS-CoV-2 as well as SARS-CoV with low micromolar activity in our cell-free ELISA-type assays (IC50's of 0.2-3.0 μM), whereas control compounds, such as sunset yellow FCF, chloroquine, and suramin, showed no activity. Protein thermal shift assays indicated that the SMIs of interest identified here bind SARS-CoV-2-S and not hACE2. While dyes seemed to be promiscuous inhibitors, DRI-C23041 showed some selectivity and inhibited the entry of two different SARS-CoV-2-S expressing pseudoviruses into hACE2-expressing cells in a concentration-dependent manner with low micromolar IC50's (6-7 μM). This provides proof-of-principle evidence for the feasibility of small-molecule inhibition of PPIs critical for SARS-CoV-2 attachment/entry and serves as a first guide in the search for SMI-based alternative antiviral therapies for the prevention and treatment of diseases caused by coronaviruses in general and COVID-19 in particular.
Collapse
Affiliation(s)
- Damir Bojadzic
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Oscar Alcazar
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Jinshui Chen
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Sung-Ting Chuang
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
| | - Jose M. Condor Capcha
- Division of Cardiology, University of Miami, Miami, Florida, USA
- Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
| | - Lina A. Shehadeh
- Division of Cardiology, University of Miami, Miami, Florida, USA
- Interdisciplinary Stem Cell Institute, University of Miami, Miami, Florida, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miami, Florida, USA
| | - Peter Buchwald
- Diabetes Research Institute, University of Miami, Miami, Florida, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
4
|
Kuusk A, Boyd H, Chen H, Ottmann C. Small-molecule modulation of p53 protein-protein interactions. Biol Chem 2021; 401:921-931. [PMID: 32049643 DOI: 10.1515/hsz-2019-0405] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/30/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022]
Abstract
Small-molecule modulation of protein-protein interactions (PPIs) is a very promising but also challenging area in drug discovery. The tumor suppressor protein p53 is one of the most frequently altered proteins in human cancers, making it an attractive target in oncology. 14-3-3 proteins have been shown to bind to and positively regulate p53 activity by protecting it from MDM2-dependent degradation or activating its DNA binding affinity. PPIs can be modulated by inhibiting or stabilizing specific interactions by small molecules. Whereas inhibition has been widely explored by the pharmaceutical industry and academia, the opposite strategy of stabilizing PPIs still remains relatively underexploited. This is rather interesting considering the number of natural compounds like rapamycin, forskolin and fusicoccin that exert their activity by stabilizing specific PPIs. In this review, we give an overview of 14-3-3 interactions with p53, explain isoform specific stabilization of the tumor suppressor protein, explore the approach of stabilizing the 14-3-3σ-p53 complex and summarize some promising small molecules inhibiting the p53-MDM2 protein-protein interaction.
Collapse
Affiliation(s)
- Ave Kuusk
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, NL-5600MB Eindhoven, The Netherlands
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, S-43183 Mölndal, Sweden
| | - Helen Boyd
- Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| | - Hongming Chen
- Guangzhou Regenerative Medicine and Health-Guangdong Laboratory, Science Park, Guangzhou 510530, China
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, NL-5600MB Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, D-45141 Essen, Germany
| |
Collapse
|
5
|
Bojadzic D, Alcazar O, Buchwald P. Methylene Blue Inhibits the SARS-CoV-2 Spike-ACE2 Protein-Protein Interaction-a Mechanism that can Contribute to its Antiviral Activity Against COVID-19. Front Pharmacol 2021; 11:600372. [PMID: 33519460 PMCID: PMC7838506 DOI: 10.3389/fphar.2020.600372] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
Due to our interest in the chemical space of organic dyes to identify potential small-molecule inhibitors (SMIs) for protein-protein interactions (PPIs), we initiated a screen of such compounds to assess their inhibitory activity against the interaction between SARS-CoV-2 spike protein and its cognate receptor ACE2, which is the first critical step initiating the viral attachment and entry of this coronavirus responsible for the ongoing COVID-19 pandemic. As part of this, we found that methylene blue, a tricyclic phenothiazine compound approved by the FDA for the treatment of methemoglobinemia and used for other medical applications (including the inactivation of viruses in blood products prior to transfusion when activated by light), inhibits this interaction. We confirmed that it does so in a concentration-dependent manner with a low micromolar half-maximal inhibitory concentration (IC50 = 3 μM) in our protein-based ELISA-type setup, while chloroquine, siramesine, and suramin showed no inhibitory activity in this assay. Erythrosine B, which we have shown before to be a promiscuous SMI of PPIs, also inhibited this interaction. Methylene blue inhibited the entry of a SARS-CoV-2 spike bearing pseudovirus into ACE2-expressing cells with similar IC50 (3.5 μM). Hence, this PPI inhibitory activity could contribute to its antiviral activity against SARS-CoV-2 even in the absence of light by blocking its attachment to ACE2-expressing cells and making this inexpensive and widely available drug potentially useful in the prevention and treatment of COVID-19 as an oral or inhaled medication.
Collapse
Affiliation(s)
- Damir Bojadzic
- Diabetes Research Institute, University of Miami, Miami, FL, United States
| | - Oscar Alcazar
- Diabetes Research Institute, University of Miami, Miami, FL, United States
| | - Peter Buchwald
- Diabetes Research Institute, University of Miami, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
6
|
Mantsyzov AB, Sokolov MN, Ivantcova PM, Bräse S, Polshakov VI, Kudryavtsev KV. Interplay of Pyrrolidine Units with Homo/Hetero Chirality and CF 3-Aryl Substituents on Secondary Structures of β-Proline Tripeptides in Solution. J Org Chem 2020; 85:8865-8871. [PMID: 32526142 DOI: 10.1021/acs.joc.0c00598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/17/2022]
Abstract
All possible variants of β-proline functionalized tripeptides consisting of homo/hetero chiral monomeric all-cis 5-arylpyrrolidine-2,4-dicarboxylate units were synthesized for the first time by a nonpeptidic coupling method based on 1,3-dipolar cycloaddition chemistry of azomethine ylides. Secondary structures of β-proline tripeptides in solution were determined using the NMR spectroscopy data. o-(Trifluoromethyl)phenyl substituent contributes to stereoselectivity of 1,3-dipolar cycloaddition and structural features of β-proline tripeptides. A β-proline CF3-tripeptide with alternating absolute chirality between adjacent pyrrolidine units mimics natural PPII helix secondary structure.
Collapse
Affiliation(s)
- Alexey B Mantsyzov
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Lomonosovsky Ave 31/5, Moscow, 119992, Russian Federation
| | - Mikhail N Sokolov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow, 119991, Russian Federation
| | - Polina M Ivantcova
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow, 119991, Russian Federation
| | - Stefan Bräse
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, Karlsruhe, 76131, Germany.,Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, D-76344, Germany
| | - Vladimir I Polshakov
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Lomonosovsky Ave 31/5, Moscow, 119992, Russian Federation
| | - Konstantin V Kudryavtsev
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow, 119991, Russian Federation.,Pirogov Russian National Research Medical University, Ostrovityanova Street 1, 117997, Moscow, Russian Federation
| |
Collapse
|
7
|
Taechalertpaisarn J, Lyu RL, Arancillo M, Lin CM, Perez LM, Ioerger TR, Burgess K. Correlations between secondary structure- and protein-protein interface-mimicry: the interface mimicry hypothesis. Org Biomol Chem 2019; 17:3267-3274. [PMID: 30847453 PMCID: PMC6863600 DOI: 10.1039/c9ob00204a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/28/2023]
Abstract
An active segment of the research community designing small molecules ("minimalist mimics" of peptide fragments) to interfere with protein-protein interactions have based their studies on an implicit hypothesis. Here we refer to this as the Secondary Structure Hypothesis, that might be defined as, "If a small molecule can orient amino acid side-chains in directions that resemble side-chains of the parent secondary structure at the interface, then that small molecule is a candidate to perturb the protein-protein interaction". Rigorous tests of this hypothesis require co-crystallization of minimalist mimics with protein receptors, and comparison of the bound conformations with the interface secondary structures they were designed to resemble. Unfortunately, to the best of our knowledge, there is no such analysis in the literature, and it is unlikely that enough examples will emerge in the near future to test the hypothesis. Research described here was designed to challenge this hypothesis from a different perspective. In a previous study, preferred conformations of a series of novel minimalist mimics were simulated then systematically overlaid on >240 000 crystallographically characterized protein-protein interfaces. Select data from that overlay procedure revealed chemotypes that overlay side chains on various PPI interfaces with a relatively high frequency of occurrence. The first aim of this work was to determine if good secondary structure mimics overlay frequently on PPI interfaces. The second aim of this work was to determine if overlays of preferred conformers at interface regions involve secondary structures. Thus situations where these conformations overlaid extremely well on PPI interfaces were analyzed to determine if secondary structures featured the PPI regions where these molecules overlaid in the previous study. Combining conclusions from these two studies enabled us to formulate a hypothesis that is complementary to the Secondary Structure Hypothesis, but, unlike this, is supported by abundant data. We call this the Interface Mimicry Hypothesis.
Collapse
Affiliation(s)
- Jaru Taechalertpaisarn
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842-3012, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
Bojadzic D, Buchwald P. Toward Small-Molecule Inhibition of Protein-Protein Interactions: General Aspects and Recent Progress in Targeting Costimulatory and Coinhibitory (Immune Checkpoint) Interactions. Curr Top Med Chem 2018; 18:674-699. [PMID: 29848279 PMCID: PMC6067980 DOI: 10.2174/1568026618666180531092503] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/01/2017] [Revised: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023]
Abstract
Protein-Protein Interactions (PPIs) that are part of the costimulatory and coinhibitory (immune checkpoint) signaling are critical for adequate T cell response and are important therapeutic targets for immunomodulation. Biologics targeting them have already achieved considerable clinical success in the treatment of autoimmune diseases or transplant recipients (e.g., abatacept, belatacept, and belimumab) as well as cancer (e.g., ipilimumab, nivolumab, pembrolizumab, atezolizumab, durvalumab, and avelumab). In view of such progress, there have been only relatively limited efforts toward developing small-molecule PPI inhibitors (SMPPIIs) targeting these cosignaling interactions, possibly because they, as all other PPIs, are difficult to target by small molecules and were not considered druggable. Nevertheless, substantial progress has been achieved during the last decade. SMPPIIs proving the feasibility of such approaches have been identified through various strategies for a number of cosignaling interactions including CD40-CD40L, OX40-OX40L, BAFFR-BAFF, CD80-CD28, and PD-1-PD-L1s. Here, after an overview of the general aspects and challenges of SMPPII-focused drug discovery, we review them briefly together with relevant structural, immune-signaling, physicochemical, and medicinal chemistry aspects. While so far only a few of these SMPPIIs have shown activity in animal models (DRI-C21045 for CD40-D40L, KR33426 for BAFFR-BAFF) or reached clinical development (RhuDex for CD80-CD28, CA-170 for PD-1-PD-L1), there is proof-of-principle evidence for the feasibility of such approaches in immunomodulation. They can result in products that are easier to develop/ manufacture and are less likely to be immunogenic or encounter postmarket safety events than corresponding biologics, and, contrary to them, can even become orally bioavailable.
Collapse
Affiliation(s)
- Damir Bojadzic
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
9
|
Targeting Intramembrane Protein-Protein Interactions: Novel Therapeutic Strategy of Millions Years Old. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:61-99. [PMID: 29459036 PMCID: PMC7102818 DOI: 10.1016/bs.apcsb.2017.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Abstract
Intramembrane protein-protein interactions (PPIs) are involved in transmembrane signal transduction mediated by cell surface receptors and play an important role in health and disease. Recently, receptor-specific modulatory peptides rationally designed using a general platform of transmembrane signaling, the signaling chain homooligomerization (SCHOOL) model, have been proposed to therapeutically target these interactions in a variety of serious diseases with unmet needs including cancer, sepsis, arthritis, retinopathy, and thrombosis. These peptide drug candidates use ligand-independent mechanisms of action (SCHOOL mechanisms) and demonstrate potent efficacy in vitro and in vivo. Recent studies surprisingly revealed that in order to modify and/or escape the host immune response, human viruses use similar mechanisms and modulate cell surface receptors by targeting intramembrane PPIs in a ligand-independent manner. Here, I review these intriguing mechanistic similarities and discuss how the viral strategies optimized over a billion years of the coevolution of viruses and their hosts can help to revolutionize drug discovery science and develop new, disruptive therapies. Examples are given.
Collapse
|
10
|
Chen J, Song Y, Bojadzic D, Tamayo-Garcia A, Landin AM, Blomberg BB, Buchwald P. Small-Molecule Inhibitors of the CD40-CD40L Costimulatory Protein-Protein Interaction. J Med Chem 2017; 60:8906-8922. [PMID: 29024591 PMCID: PMC5823691 DOI: 10.1021/acs.jmedchem.7b01154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023]
Abstract
Costimulatory interactions are required for T cell activation and development of an effective immune response; hence, they are valuable therapeutic targets for immunomodulation. However, they, as all other protein-protein interactions, are difficult to target by small molecules. Here, we report the identification of novel small-molecule inhibitors of the CD40-CD40L interaction designed starting from the chemical space of organic dyes. For the most promising compounds such as DRI-C21045, activity (IC50) in the low micromolar range has been confirmed in cell assays including inhibition of CD40L-induced activation in NF-κB sensor cells, THP-1 myeloid cells, and primary human B cells as well as in murine allogeneic skin transplant and alloantigen-induced T cell expansion in draining lymph node experiments. Specificity versus other TNF-superfamily interactions (TNF-R1-TNF-α) and lack of cytotoxicity have also been confirmed at these concentrations. These novel compounds provide proof-of-principle evidence for the possibility of small-molecule inhibition of costimulatory protein-protein interactions, establish the structural requirements needed for efficient CD40-CD40L inhibition, and serve to guide the search for such immune therapeutics.
Collapse
Affiliation(s)
- Jinshui Chen
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Yun Song
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
- Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Damir Bojadzic
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Alejandro Tamayo-Garcia
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Ana Marie Landin
- Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Bonnie B. Blomberg
- Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
- Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA
| |
Collapse
|
11
|
Bosc N, Kuenemann MA, Bécot J, Vavrusa M, Cerdan AH, Sperandio O. Privileged Substructures to Modulate Protein-Protein Interactions. J Chem Inf Model 2017; 57:2448-2462. [PMID: 28922596 DOI: 10.1021/acs.jcim.7b00435] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022]
Abstract
Given the difficulties to identify chemical probes that can modulate protein-protein interactions (PPIs), actors in the field have started to agree on the necessity to use PPI-tailored screening chemical collections. However, which type of scaffolds may promote the binding of compounds to PPI targets remains unclear. In this big data analysis, we have identified a list of privileged chemical substructures that are most often observed within inhibitors of PPIs. Using molecular frameworks as a way to perceive chemical substructures with the combination of an experimental and a machine-learning based predicted data set of iPPI compounds, we propose a list of privileged substructures in the form of scaffolds and chemical moieties that can be substantially chemically functionalized and do not present any toxicophore nor pan-assay interference (PAINS) alerts. We think that such chemical guidance will be valuable for medicinal chemists in their attempt to identify initial quality chemical probes on PPI targets.
Collapse
Affiliation(s)
- Nicolas Bosc
- Inserm, U973 , Paris 75013, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm , Paris 75013, France.,Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, Institut Pasteur , 25-28 rue du Dr Roux, Paris 75015, France.,CNRS UMR3528, Institut Pasteur , 25-28 rue du Dr Roux, Paris 75015, France
| | - Mélaine A Kuenemann
- Inserm, U973 , Paris 75013, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm , Paris 75013, France
| | - Jerome Bécot
- Inserm, U973 , Paris 75013, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm , Paris 75013, France
| | - Marek Vavrusa
- Inserm, U973 , Paris 75013, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm , Paris 75013, France
| | - Adrien H Cerdan
- Inserm, U973 , Paris 75013, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm , Paris 75013, France
| | - Olivier Sperandio
- Inserm, U973 , Paris 75013, France.,Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm , Paris 75013, France.,Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, Institut Pasteur , 25-28 rue du Dr Roux, Paris 75015, France.,CNRS UMR3528, Institut Pasteur , 25-28 rue du Dr Roux, Paris 75015, France
| |
Collapse
|
12
|
Affiliation(s)
- Garland R. Marshall
- Department of Biochemistry and Molecular Biophysics; Washington University School of Medicine; St. Louis Missouri 63110
| | - Flavio Ballante
- Department of Biochemistry and Molecular Biophysics; Washington University School of Medicine; St. Louis Missouri 63110
| |
Collapse
|
13
|
Metrano A, Abascal NC, Mercado BQ, Paulson EK, Hurtley AE, Miller SJ. Diversity of Secondary Structure in Catalytic Peptides with β-Turn-Biased Sequences. J Am Chem Soc 2017; 139:492-516. [PMID: 28029251 PMCID: PMC5312972 DOI: 10.1021/jacs.6b11348] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/01/2016] [Indexed: 11/30/2022]
Abstract
X-ray crystallography has been applied to the structural analysis of a series of tetrapeptides that were previously assessed for catalytic activity in an atroposelective bromination reaction. Common to the series is a central Pro-Xaa sequence, where Pro is either l- or d-proline, which was chosen to favor nucleation of canonical β-turn secondary structures. Crystallographic analysis of 35 different peptide sequences revealed a range of conformational states. The observed differences appear not only in cases where the Pro-Xaa loop-region is altered, but also when seemingly subtle alterations to the flanking residues are introduced. In many instances, distinct conformers of the same sequence were observed, either as symmetry-independent molecules within the same unit cell or as polymorphs. Computational studies using DFT provided additional insight into the analysis of solid-state structural features. Select X-ray crystal structures were compared to the corresponding solution structures derived from measured proton chemical shifts, 3J-values, and 1H-1H-NOESY contacts. These findings imply that the conformational space available to simple peptide-based catalysts is more diverse than precedent might suggest. The direct observation of multiple ground state conformations for peptides of this family, as well as the dynamic processes associated with conformational equilibria, underscore not only the challenge of designing peptide-based catalysts, but also the difficulty in predicting their accessible transition states. These findings implicate the advantages of low-barrier interconversions between conformations of peptide-based catalysts for multistep, enantioselective reactions.
Collapse
Affiliation(s)
- Anthony
J. Metrano
- Department of Chemistry, Yale University, P.O.
Box 208107, New Haven, Connecticut 06520-8107, United States
| | - Nadia C. Abascal
- Department of Chemistry, Yale University, P.O.
Box 208107, New Haven, Connecticut 06520-8107, United States
| | - Brandon Q. Mercado
- Department of Chemistry, Yale University, P.O.
Box 208107, New Haven, Connecticut 06520-8107, United States
| | - Eric K. Paulson
- Department of Chemistry, Yale University, P.O.
Box 208107, New Haven, Connecticut 06520-8107, United States
| | - Anna E. Hurtley
- Department of Chemistry, Yale University, P.O.
Box 208107, New Haven, Connecticut 06520-8107, United States
| | - Scott J. Miller
- Department of Chemistry, Yale University, P.O.
Box 208107, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
14
|
Jouanne M, Voisin-Chiret AS, Legay R, Coufourier S, Rault S, Sopkova-de Oliveira Santos J. β-Strand Mimicry: Exploring Oligothienylpyridine Foldamers. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600882] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Affiliation(s)
- Marie Jouanne
- Université Caen Normandie; Caen France
- UNICAEN; CERMN - EA 4258; FR CNRS 3038 INC3M, SF 4206 ICORE; bd Becquerel 14032 Caen France
| | - Anne Sophie Voisin-Chiret
- Université Caen Normandie; Caen France
- UNICAEN; CERMN - EA 4258; FR CNRS 3038 INC3M, SF 4206 ICORE; bd Becquerel 14032 Caen France
| | - Rémi Legay
- Université Caen Normandie; Caen France
- UNICAEN; CERMN - EA 4258; FR CNRS 3038 INC3M, SF 4206 ICORE; bd Becquerel 14032 Caen France
| | - Sébastien Coufourier
- Université Caen Normandie; Caen France
- UNICAEN; CERMN - EA 4258; FR CNRS 3038 INC3M, SF 4206 ICORE; bd Becquerel 14032 Caen France
| | - Sylvain Rault
- Université Caen Normandie; Caen France
- UNICAEN; CERMN - EA 4258; FR CNRS 3038 INC3M, SF 4206 ICORE; bd Becquerel 14032 Caen France
| | - Jana Sopkova-de Oliveira Santos
- Université Caen Normandie; Caen France
- UNICAEN; CERMN - EA 4258; FR CNRS 3038 INC3M, SF 4206 ICORE; bd Becquerel 14032 Caen France
| |
Collapse
|
15
|
Kudryavtsev KV, Mantsyzov AB, Ivantcova PM, Sokolov MN, Churakov AV, Bräse S, Zefirov NS, Polshakov VI. Control of Azomethine Cycloaddition Stereochemistry by CF3 Group: Structural Diversity of Fluorinated β-Proline Dimers. Org Lett 2016; 18:4698-701. [PMID: 27574905 DOI: 10.1021/acs.orglett.6b02327] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
Abstract
β-Proline-functionalized dimers consisting of homochiral monomeric units were synthesized by a non-peptidic coupling method for the first time. The applied synthetic methodology is based on 1,3-dipolar cycloaddition chemistry of azomethine ylides and provides absolute control over the β-proline backbone stereogenic centers. An o-(trifluoromethyl)phenyl substituent contributes to appropriate stabilization of the definite acrylamide chiral cis conformation and to achieve the dipole reactivity that is not observed for aryl groups lacking strong electronegative character.
Collapse
Affiliation(s)
- Konstantin V Kudryavtsev
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory 1/3, Moscow 119991, Russian Federation.,Institute of Physiologically Active Compounds, Russian Academy of Sciences , Chernogolovka, Moscow Region 142432, Russian Federation
| | - Alexey B Mantsyzov
- Faculty of Fundamental Medicine, Lomonosov Moscow State University , Lomonosovsky Ave 31/5, Moscow 119992, Russian Federation
| | - Polina M Ivantcova
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory 1/3, Moscow 119991, Russian Federation
| | - Mikhail N Sokolov
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory 1/3, Moscow 119991, Russian Federation
| | - Andrei V Churakov
- Institute of General and Inorganic Chemistry, Russian Academy of Sciences , Leninskii prosp. 31, Moscow 119991, Russian Federation
| | - Stefan Bräse
- Institute of Organic Chemistry, Karlsruhe Institute of Technology , Fritz-Haber-Weg 6, Karlsruhe 76131, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology , Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen 76344, Germany
| | - Nikolay S Zefirov
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory 1/3, Moscow 119991, Russian Federation.,Institute of Physiologically Active Compounds, Russian Academy of Sciences , Chernogolovka, Moscow Region 142432, Russian Federation
| | - Vladimir I Polshakov
- Faculty of Fundamental Medicine, Lomonosov Moscow State University , Lomonosovsky Ave 31/5, Moscow 119992, Russian Federation.,NMR Laboratory, Institute of Physics, Kazan Federal University , Kremlevskaya 18, Kazan 420008, Russian Federation
| |
Collapse
|
16
|
Gopalakrishnan R, Frolov AI, Knerr L, Drury WJ, Valeur E. Therapeutic Potential of Foldamers: From Chemical Biology Tools To Drug Candidates? J Med Chem 2016; 59:9599-9621. [PMID: 27362955 DOI: 10.1021/acs.jmedchem.6b00376] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
Over the past decade, foldamers have progressively emerged as useful architectures to mimic secondary structures of proteins. Peptidic foldamers, consisting of various amino acid based backbones, have been the most studied from a therapeutic perspective, while polyaromatic foldamers have barely evolved from their nascency and remain perplexing for medicinal chemists due to their poor drug-like nature. Despite these limitations, this compound class may still offer opportunities to study challenging targets or provide chemical biology tools. The potential of foldamer drug candidates reaching the clinic is still a stretch. Nevertheless, advances in the field have demonstrated their potential for the discovery of next generation therapeutics. In this perspective, the current knowledge of foldamers is reviewed in a drug discovery context. Recent advances in the early phases of drug discovery including hit finding, target validation, and optimization and molecular modeling are discussed. In addition, challenges and focus areas are debated and gaps highlighted.
Collapse
Affiliation(s)
- Ranganath Gopalakrishnan
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, 431 83, Sweden.,AstraZeneca MPI Satellite Unit, Department of Chemical Biology, Max Planck Institute of Molecular Physiology , Dortmund 44202, Germany
| | - Andrey I Frolov
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Laurent Knerr
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - William J Drury
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Eric Valeur
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, 431 83, Sweden
| |
Collapse
|
17
|
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] [Academic Contribution 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
|
18
|
Jakobi S, Nguyen PTX, Debaene F, Cianférani S, Reuter K, Klebe G. What Glues a Homodimer Together: Systematic Analysis of the Stabilizing Effect of an Aromatic Hot Spot in the Protein-Protein Interface of the tRNA-Modifying Enzyme Tgt. ACS Chem Biol 2015; 10:1897-907. [PMID: 25951081 DOI: 10.1021/acschembio.5b00028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022]
Abstract
Shigella bacteria constitute the causative agent of bacillary dysentery, an acute inflammatory disease causing the death of more than one million humans per year. A null mutation in the tgt gene encoding the tRNA-modifying enzyme tRNA-guanine transglycosylase (Tgt) was found to drastically decrease the pathogenicity of Shigella bacteria, suggesting the use of Tgt as putative target for selective antibiotics. The enzyme is only functionally active as a homodimer; thus, interference with the formation of its protein-protein interface is an attractive opportunity for therapeutic intervention. To better understand the driving forces responsible for the assembly, stability, and formation of the homodimer, we studied the properties of the residues that establish the dimer interface in detail. We performed site-directed mutagenesis and controlled shifts in the monomer/dimer equilibrium ratio in solution in a concentration-dependent manner by native mass spectrometry and used crystal structure analysis to elucidate the geometrical modulations resulting from mutational variations. The wild-type enzyme exhibits nearly exclusive dimer geometry. A patch of four aromatic amino acids, embedded into a ring of hydrophobic residues and further stabilized by a network of H-bonds, is essential for the stability of the dimer's contact. Accordingly, any perturbance in the constitution of this aromatic patch by nonaromatic residues reduces dimer stability significantly, with some of these exchanges resulting in a nearly exclusively monomeric state. Apart from the aromatic hot spot, the interface comprises an extended loop-helix motif that exhibits remarkable flexibility. In the destabilized mutated variants, the loop-helix motif adopts deviating conformations in the interface region, and a number of water molecules, penetrating into the interface, are observed.
Collapse
Affiliation(s)
- Stephan Jakobi
- Institut
für Pharmazeutische Chemie, Philipps-Universität Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Phong T. X. Nguyen
- Institut
für Pharmazeutische Chemie, Philipps-Universität Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - François Debaene
- BioOrganic
Mass Spectrometry Laboratory (LSMBO), Université de Strasbourg, IPHC,
25 rue Becquerel, 67087 Strasbourg, France
- CNRS, UMR7178, 67087 Strasbourg, France
| | - Sarah Cianférani
- BioOrganic
Mass Spectrometry Laboratory (LSMBO), Université de Strasbourg, IPHC,
25 rue Becquerel, 67087 Strasbourg, France
- CNRS, UMR7178, 67087 Strasbourg, France
| | - Klaus Reuter
- Institut
für Pharmazeutische Chemie, Philipps-Universität Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| | - Gerhard Klebe
- Institut
für Pharmazeutische Chemie, Philipps-Universität Marburg, Marbacher Weg
6, D-35032 Marburg, Germany
| |
Collapse
|
19
|
Fahs S, Patil-Sen Y, Snape TJ. Foldamers as Anticancer Therapeutics: Targeting Protein-Protein Interactions and the Cell Membrane. Chembiochem 2015; 16:1840-1853. [DOI: 10.1002/cbic.201500188] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2015] [Indexed: 01/10/2023]
|
20
|
Gloaguen C, Voisin-Chiret AS, Sopkova-de Oliveira Santos J, Fogha J, Gautier F, De Giorgi M, Burzicki G, Perato S, Pétigny-Lechartier C, Simonin-Le Jeune K, Brotin E, Goux D, N'Diaye M, Lambert B, Louis MH, Ligat L, Lopez F, Juin P, Bureau R, Rault S, Poulain L. First evidence that oligopyridines, α-helix foldamers, inhibit Mcl-1 and sensitize ovarian carcinoma cells to Bcl-xL-targeting strategies. J Med Chem 2015; 58:1644-68. [PMID: 25585174 DOI: 10.1021/jm500672y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
Apoptosis control defects such as the deregulation of Bcl-2 family member expression are frequently involved in chemoresistance. In ovarian carcinoma, we previously demonstrated that Bcl-xL and Mcl-1 cooperate to protect cancer cells against apoptosis and their concomitant inhibition leads to massive apoptosis even in the absence of chemotherapy. Whereas Bcl-xL inhibitors are now available, Mcl-1 inhibition, required to sensitize cells to Bcl-xL-targeting strategies, remains problematic. In this context, we designed and synthesized oligopyridines potentially targeting the Mcl-1 hydrophobic pocket, evaluated their capacity to inhibit Mcl-1 in live cells, and implemented a functional screening assay to evaluate their ability to sensitize ovarian carcinoma cells to Bcl-xL-targeting strategies. We established structure-activity relationships and focused our attention on MR29072, named Pyridoclax. Surface plasmon resonance assay demonstrated that pyridoclax directly binds to Mcl-1. Without cytotoxic activity when administered as a single agent, pyridoclax induced apoptosis in combination with Bcl-xL-targeting siRNA or with ABT-737 in ovarian, lung, and mesothelioma cancer cells.
Collapse
|
21
|
Song Y, Buchwald P. TNF superfamily protein-protein interactions: feasibility of small- molecule modulation. Curr Drug Targets 2015; 16:393-408. [PMID: 25706111 PMCID: PMC4408546 DOI: 10.2174/1389450116666150223115628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 01/09/2023]
Abstract
The tumor necrosis factor (TNF) superfamily (TNFSF) contains about thirty structurally related receptors (TNFSFRs) and about twenty protein ligands that bind to one or more of these receptors. Almost all of these cell surface protein-protein interactions (PPIs) represent high-value therapeutic targets for inflammatory or immune modulation in autoimmune diseases, transplant recipients, or cancers, and there are several biologics including antibodies and fusion proteins targeting them that are in various phases of clinical development. Small-molecule inhibitors or activators could represent possible alternatives if the difficulties related to the targeting of protein-protein interactions by small molecules can be addressed. Compounds proving the feasibility of such approaches have been identified through different drug discovery approaches for a number of these TNFSFR-TNFSF type PPIs including CD40-CD40L, BAFFR-BAFF, TRAIL-DR5, and OX40-OX40L. Corresponding structural, signaling, and medicinal chemistry aspects are briefly reviewed here. While none of these small-molecule modulators identified so far seems promising enough to be pursued for clinical development, they provide proof-of-principle evidence that these interactions are susceptible to small-molecule modulation and can serve as starting points toward the identification of more potent and selective candidates.
Collapse
Affiliation(s)
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, 1450 NW 10 Ave (R-134), Miami, FL 33136, USA.
| |
Collapse
|
22
|
Jakobi S, Nguyen TXP, Debaene F, Metz A, Sanglier-Cianférani S, Reuter K, Klebe G. Hot-spot analysis to dissect the functional protein-protein interface of a tRNA-modifying enzyme. Proteins 2014; 82:2713-32. [DOI: 10.1002/prot.24637] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2014] [Revised: 05/24/2014] [Accepted: 06/18/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Stephan Jakobi
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg; Marbacher Weg 6 D-35032 Marburg Germany
| | - Tran Xuan Phong Nguyen
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg; Marbacher Weg 6 D-35032 Marburg Germany
| | - François Debaene
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg; CNRS UMR7178; 25 rue Becquerel 67087 Strasbourg France
| | - Alexander Metz
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg; Marbacher Weg 6 D-35032 Marburg Germany
| | - Sarah Sanglier-Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg; CNRS UMR7178; 25 rue Becquerel 67087 Strasbourg France
| | - Klaus Reuter
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg; Marbacher Weg 6 D-35032 Marburg Germany
| | - Gerhard Klebe
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg; Marbacher Weg 6 D-35032 Marburg Germany
| |
Collapse
|
23
|
Fayne D. De-peptidising protein-protein interactions - big jobs for small molecules. DRUG DISCOVERY TODAY. TECHNOLOGIES 2013; 10:e467-e474. [PMID: 24451636 DOI: 10.1016/j.ddtec.2013.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/03/2023]
Abstract
Virtually all biological processes rely on protein-protein interactions (PPIs) for signal propagation, therefore representing a vast array of potentially viable therapeutic intervention points. Targeting PPIs is a relatively novel drug development strategy so computational approaches towards analysing the interface between protein partners and predicting the likelihood of developing a small molecule inhibitor are still progressing. This review provides an overview of recent successful examples of computational methodologies used to predict druggable PPIs and small molecules designed to inhibit them.
Collapse
|
24
|
Perato S, Fogha J, Sebban M, Voisin-Chiret AS, Sopkova-de Oliveira Santos J, Oulyadi H, Rault S. Conformation Control of Abiotic α-Helical Foldamers. J Chem Inf Model 2013; 53:2671-80. [DOI: 10.1021/ci400365y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
Affiliation(s)
- Serge Perato
- Normandie Université, France
- UNICAEN, CERMN (Centre d’Etudes
et de Recherche sur
le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M,
Bd Becquerel), F-14032 Caen, France
| | - Jade Fogha
- Normandie Université, France
- UNICAEN, CERMN (Centre d’Etudes
et de Recherche sur
le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M,
Bd Becquerel), F-14032 Caen, France
| | - Muriel Sebban
- Université de Rouen, COBRA-UMR 6014, Rue Lucien
Tesnières, F-76821 Mont-Saint-Aignan, France
| | - Anne Sophie Voisin-Chiret
- Normandie Université, France
- UNICAEN, CERMN (Centre d’Etudes
et de Recherche sur
le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M,
Bd Becquerel), F-14032 Caen, France
| | - Jana Sopkova-de Oliveira Santos
- Normandie Université, France
- UNICAEN, CERMN (Centre d’Etudes
et de Recherche sur
le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M,
Bd Becquerel), F-14032 Caen, France
| | - Hassan Oulyadi
- Université de Rouen, COBRA-UMR 6014, Rue Lucien
Tesnières, F-76821 Mont-Saint-Aignan, France
| | - Sylvain Rault
- Normandie Université, France
- UNICAEN, CERMN (Centre d’Etudes
et de Recherche sur
le Médicament de Normandie UPRES EA 4258-FR CNRS 3038 INC3M,
Bd Becquerel), F-14032 Caen, France
| |
Collapse
|
25
|
Wawrzyńska A, Kurzyk A, Mierzwińska M, Płochocka D, Wieczorek G, Sirko A. Direct targeting of Arabidopsis cysteine synthase complexes with synthetic polypeptides to selectively deregulate cysteine synthesis. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2013; 207:148-157. [PMID: 23602110 DOI: 10.1016/j.plantsci.2013.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/27/2012] [Revised: 02/22/2013] [Accepted: 02/24/2013] [Indexed: 06/02/2023]
Abstract
Biosynthesis of cysteine is one of the fundamental processes in plants providing the reduced sulfur for cell metabolism. It is accomplished by the sequential action of two enzymes, serine acetyltransferase (SAT) and O-acetylserine (thiol) lyase (OAS-TL). Together they constitute the hetero-oligomeric cysteine synthase (CS) complex through specific protein-protein interactions influencing the rate of cysteine production. The aim of our studies was to deregulate the CS complex formation in order to investigate its function in the control of sulfur homeostasis and optimize cysteine synthesis. Computational modeling was used to build a model of the Arabidopsis thaliana mitochondrial CS complex. Several polypeptides based on OAS-TL C amino-acid sequence found at SAT-OASTL interaction sites were designed as probable competitors for SAT3 binding. After verification of the binding in a yeast two-hybrid assay, the most strongly interacting polypeptide was introduced to different cellular compartments of Arabidopsis cell via genetic transformation. Moderate increase in total SAT and OAS-TL activities, but not thiols content, was observed dependent on the transgenic line and sulfur availability in the hydroponic medium. Though our studies demonstrate the proof of principle, they also suggest more complex interaction of both enzymes underlying the mechanism of their reciprocal regulation.
Collapse
Affiliation(s)
- Anna Wawrzyńska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5A St, 02-106 Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
26
|
Immekus F, Barandun LJ, Betz M, Debaene F, Petiot S, Sanglier-Cianferani S, Reuter K, Diederich F, Klebe G. Launching spiking ligands into a protein-protein interface: a promising strategy to destabilize and break interface formation in a tRNA modifying enzyme. ACS Chem Biol 2013; 8:1163-78. [PMID: 23534552 DOI: 10.1021/cb400020b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
Apart from competitive active-site inhibition of protein function, perturbance of protein-protein interactions by small molecules in oligodomain enzymes opens new perspectives for innovative therapeutics. tRNA-guanine transglycosylase (TGT), a potential target to treat shigellosis, is active only as the homodimer. Consequently, disruption of the dimer interface by small molecules provides a novel inhibition mode. A special feature of this enzyme is the short distance between active site and rim of the dimer interface. This suggests design of expanded active-site inhibitors decorated with rigid, needle-type substituents to spike into potential hot spots of the interaction interface. Ligands with attached ethinyl-type substituents have been synthesized and characterized by Kd measurements, crystallography, noncovalent mass spectrometry, and computer simulations. In contrast to previously determined crystal structures with nonextended active-site inhibitors, a well-defined loop-helix motif, involved in several contacts across the dimer interface, falls apart and suggests enhanced flexibility once the spiking ligands are bound. Mass spectrometry indicates significant destabilization but not full disruption of the complexed TGT homodimer in solution. As directed interactions of the loop-helix motif obviously do not determine dimer stability, a structurally conserved hydrophobic patch composed of several aromatic amino acids is suggested as interaction hot spot. The residues of this patch reside on a structurally highly conserved helix-turn-helix motif, which remains unaffected by the bound spiking ligands. Nevertheless, it is shielded from solvent access by the loop-helix motif that becomes perturbed upon binding of the spiking ligands, which serves as a possible explanation for reduced interface stability.
Collapse
Affiliation(s)
- Florian Immekus
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Luzi Jakob Barandun
- Laboratorium für Organische
Chemie, ETH Zurich Hönggerberg, HCI, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | - Michael Betz
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - François Debaene
- Laboratoire de Spectrométrie de Masse Bio-Organique de Strasbourg, 25, rue Becquerel,
67087 Strasbourg Cedex 2, France
| | - Stéphanie Petiot
- Laboratoire de Spectrométrie de Masse Bio-Organique de Strasbourg, 25, rue Becquerel,
67087 Strasbourg Cedex 2, France
| | - Sarah Sanglier-Cianferani
- Laboratoire de Spectrométrie de Masse Bio-Organique de Strasbourg, 25, rue Becquerel,
67087 Strasbourg Cedex 2, France
| | - Klaus Reuter
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - François Diederich
- Laboratorium für Organische
Chemie, ETH Zurich Hönggerberg, HCI, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | - Gerhard Klebe
- Institut für Pharmazeutische
Chemie, Philipps-Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| |
Collapse
|
27
|
Using halo (het) arylboronic species to achieve synthesis of foldamers as protein–protein interaction disruptors. PURE APPL CHEM 2012. [DOI: 10.1351/pac-con-11-10-30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
Protein–protein interactions (PPIs) play a central role in all biological processes and have been the focus of intense investigations from structural molecular biology to cell biology for the majority of the last two decades and, more recently, are emerging as important targets for pharmaceuticals. A common motif found at the interface of PPIs is the α-helix, and apart from the peptidic structures, numerous nonpeptidic small molecules have been developed to mimic α-helices. The first-generation terphenyl scaffold is able to successfully mimic key helix residues and disrupt relevant interactions, including Bcl-xL-Bak interactions that are implicated in apoptosis mechanism. These scaffolds were designed and evaluated in silico. Analysis revealed that substituents on aromatic scaffolds can efficiently mimic side-chain surfaces. Unfortunately, the literature describes a long and difficult procedure to access these aromatic-based scaffolds. The search for new simpler methodology is the aim of the research of our medicinal chemistry team. On the basis of structural requirements, we developed a program concerning the synthesis of new oligo(het)aryl scaffolds produced by iterative couplings of boronic species (garlanding) in which substituents on rings project functionality in spatial orientations that mimic residues of an α-helix.
Collapse
|
28
|
Voisin-Chiret AS, Burzicki G, Perato S, De Giorgi M, Franchini C, Sopková-de Oliveira Santos J, Rault S. Aromatic garlands, as new foldamers, to mimic protein secondary structure. Tetrahedron 2012. [DOI: 10.1016/j.tet.2012.02.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
|
29
|
|
30
|
Patil NT, Lakshmi PGVV, Sridhar B, Patra S, Pal Bhadra M, Patra CR. New Linearly and Angularly Fused Quinazolinones: Synthesis through Gold(I)-Catalyzed Cascade Reactions and Anticancer Activities. European J Org Chem 2012. [DOI: 10.1002/ejoc.201101822] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
|
31
|
Sopkova-de Oliveira Santos J, Voisin-Chiret AS, Burzicki G, Sebaoun L, Sebban M, Lohier JF, Legay R, Oulyadi H, Bureau R, Rault S. Structural Characterizations of Oligopyridyl Foldamers, α-Helix Mimetics. J Chem Inf Model 2012; 52:429-39. [DOI: 10.1021/ci200424a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jana Sopkova-de Oliveira Santos
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| | - Anne Sophie Voisin-Chiret
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| | - Gregory Burzicki
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| | - Laure Sebaoun
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| | - Muriel Sebban
- Equipe Analyse et Modélisation
Moléculaire, IRCOF UMR6014 CNRS-COBRA, Université de
Rouen, rue Tesnière, 76821 Mont-Saint-Aignan, France
| | - Jean-François Lohier
- Laboratoire de Chimie Moléculaire
et Thio-organique, UMR CNRS 6507, FR CNRS 3038 INC3M, ENSICAEN - Université
de Caen, 14050 Caen, France
| | - Rémi Legay
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| | - Hassan Oulyadi
- Equipe Analyse et Modélisation
Moléculaire, IRCOF UMR6014 CNRS-COBRA, Université de
Rouen, rue Tesnière, 76821 Mont-Saint-Aignan, France
| | - Ronan Bureau
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| | - Sylvain Rault
- Centre d’Etudes et de
Recherche sur le Médicament de Normandie, UPRES EA-4258, FR
CNRS INC3M, UFR des Sciences Pharmaceutiques, Université de
Caen, bd Becquerel,
14032 Caen Cedex, France
| |
Collapse
|
32
|
Seddon G, Lounnas V, McGuire R, van den Bergh T, Bywater RP, Oliveira L, Vriend G. Drug design for ever, from hype to hope. J Comput Aided Mol Des 2012; 26:137-50. [PMID: 22252446 PMCID: PMC3268973 DOI: 10.1007/s10822-011-9519-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/22/2011] [Accepted: 12/05/2011] [Indexed: 01/28/2023]
Abstract
In its first 25 years JCAMD has been disseminating a large number of techniques aimed at finding better medicines faster. These include genetic algorithms, COMFA, QSAR, structure based techniques, homology modelling, high throughput screening, combichem, and dozens more that were a hype in their time and that now are just a useful addition to the drug-designers toolbox. Despite massive efforts throughout academic and industrial drug design research departments, the number of FDA-approved new molecular entities per year stagnates, and the pharmaceutical industry is reorganising accordingly. The recent spate of industrial consolidations and the concomitant move towards outsourcing of research activities requires better integration of all activities along the chain from bench to bedside. The next 25 years will undoubtedly show a series of translational science activities that are aimed at a better communication between all parties involved, from quantum chemistry to bedside and from academia to industry. This will above all include understanding the underlying biological problem and optimal use of all available data.
Collapse
Affiliation(s)
| | - V. Lounnas
- CMBI, Radboud University Nijmegen Medical Centre, Geert Grooteplein 26–28, 6525 GA Nijmegen, The Netherlands
| | - R. McGuire
- BioAxis Research, Bergse Heihoek 56, Berghem, 5351 SL The Netherlands
| | - T. van den Bergh
- Bio-Prodict, Dreijenplein 10, 6703 HB Wageningen, The Netherlands
| | | | - L. Oliveira
- Sao Paulo Federal University (UNIFESP), Sao Paulo, Brazil
| | - G. Vriend
- CMBI, Radboud University Nijmegen Medical Centre, Geert Grooteplein 26–28, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
33
|
Gordo S, Martos V, Vilaseca M, Menéndez M, de Mendoza J, Giralt E. On the role of flexibility in protein-ligand interactions: the example of p53 tetramerization domain. Chem Asian J 2011; 6:1463-9. [PMID: 21626703 DOI: 10.1002/asia.201000938] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022]
Abstract
The recognition of protein surfaces by designed ligands has become an attractive approach in drug discovery. However, the variable nature and irregular behavior of protein surfaces defy this new area of research. The easy to understand "lock-and-key" model is far from being the ideal paradigm in biomolecular interactions and, hence, any new finding on how proteins and ligands behave in recognition events paves a step of the way. Herein, we illustrate a clear example on how an increase in flexibility of both protein and ligand can result in an increase in the stability of the macromolecular complex. The biophysical study of the interaction between a designed flexible tetraguanidinium-calix[4]arene and the tetramerization domain of protein p53 (p53TD) and its natural mutant p53TD-R337H shows how the floppy mutant domain interacts more tightly with the ligand than the well-packed wild-type protein. Moreover, the flexible calixarene ligand interacts with higher affinity to both wild-type and mutated protein domains than a conformationally rigid calixarene analog previously reported. These findings underscore the crucial role of flexibility in molecular recognition processes, for both small ligands and large biomolecular surfaces.
Collapse
Affiliation(s)
- Susana Gordo
- Institute for Research in Biomedicine, Baldiri Reixac 10, 08028 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Hardy S, Martin SF. Multicomponent assembly and diversification of novel heterocyclic scaffolds derived from 2-arylpiperidines. Org Lett 2011; 13:3102-5. [PMID: 21598984 PMCID: PMC3116656 DOI: 10.1021/ol201010s] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
A collection of structurally diverse, polyheterocyclic scaffolds comprising a 2-arylpiperidine subunit were synthesized using a Mannich-type multicomponent assembly process, followed by appropriately sequenced ring-forming reactions. An improved procedure for removal of N-4-pentenoyl groups was developed; one-pot sequences for tandem urea/thiourea formation and cyclization and tandem enolate arylation/alkylation were discovered. A novel entry to bridged tetrahydroquinoline scaffolds exploiting A(1,3) strain was also invented. Derivatization of several scaffolds was achieved by cross-coupling and N-functionalization.
Collapse
Affiliation(s)
- Simon Hardy
- Department of Chemistry and Biochemistry, The Texas Institute for Drug and Diagnostic Development, The University of Texas at Austin, Austin Texas 78712
| | - Stephen F. Martin
- Department of Chemistry and Biochemistry, The Texas Institute for Drug and Diagnostic Development, The University of Texas at Austin, Austin Texas 78712
| |
Collapse
|
35
|
Whitby LR, Ando Y, Setola V, Vogt PK, Roth BL, Boger DL. Design, synthesis, and validation of a β-turn mimetic library targeting protein-protein and peptide-receptor interactions. J Am Chem Soc 2011; 133:10184-94. [PMID: 21609016 DOI: 10.1021/ja201878v] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/26/2022]
Abstract
The design and synthesis of a β-turn mimetic library as a key component of a small-molecule library targeting the major recognition motifs involved in protein-protein interactions is described. Analysis of a geometric characterization of 10,245 β-turns in the protein data bank (PDB) suggested that trans-pyrrolidine-3,4-dicarboxamide could serve as an effective and synthetically accessible library template. This was confirmed by initially screening select compounds against a series of peptide-activated GPCRs that recognize a β-turn structure in their endogenous ligands. This validation study was highlighted by identification of both nonbasic and basic small molecules with high affinities (K(i) = 390 and 23 nM, respectively) for the κ-opioid receptor (KOR). Consistent with the screening capabilities of collaborators and following the design validation, the complete library was assembled as 210 mixtures of 20 compounds, providing a total of 4200 compounds designed to mimic all possible permutations of 3 of the 4 residues in a naturally occurring β-turn. Unique to the design and because of the C(2) symmetry of the template, a typical 20 × 20 × 20-mix (8000 compounds prepared as 400 mixtures of 20 compounds) needed to represent 20 variations in the side chains of three amino acid residues reduces to a 210 × 20-mix, thereby simplifying the library synthesis and subsequent screening. The library was prepared using a solution-phase synthetic protocol with liquid-liquid or liquid-solid extractions for purification and conducted on a scale that insures its long-term availability for screening campaigns. Screening the library against the human opioid receptors (KOR, MOR, and DOR) identified not only the activity of library members expected to mimic the opioid receptor peptide ligands but also additional side-chain combinations that provided enhanced receptor binding selectivities (>100-fold) and affinities (as low as K(i) = 80 nM for KOR). A key insight to emerge from the studies is that the phenol of Tyr in endogenous ligands bearing the H-Tyr-Pro-Trp/Phe-Phe-NH(2) β-turn is important for MOR binding but may not be important for KOR (accommodated, but not preferred) and that the resulting selectivity for KOR observed with its removal can be increased by replacing the phenol OH with a chlorine substituent, further enhancing KOR affinity.
Collapse
Affiliation(s)
- Landon R Whitby
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Many "new generation" peptidomimetics are designed to present amino acid side chains only; they do not have structural features that resemble peptide main chains. These types of molecules have frequently been presented in the literature as mimics of specific secondary structures. However, many "side-chain only" peptidomimetics do not rest in single conformational states, but exist in a limited number of freely interconverting forms. These different conformations may resemble different secondary structures, so referring to them as, for instance, turn- or helical-mimics understates the ways they could adapt to various binding situations. Sets of scaffolds that can be used to mimic aspects of nearly every secondary structure, i.e. universal peptidomimetics, can be constructed. These may assume a privileged place in library design, particularly in high throughput screening for pharmacological probes for which binding conformations, or even the target itself, is unknown at the time the library is designed (critical review, 101 references).
Collapse
Affiliation(s)
- Eunhwa Ko
- Texas A & M University, Chemistry Department, P.O. Box 30012, College Station, Texas 77842, USA
| | | | | |
Collapse
|
37
|
|
38
|
Abstract
This paper concerns peptidomimetic scaffolds that can present side chains in conformations resembling those of amino acids in secondary structures without incurring excessive entropic or enthalpic penalties. Compounds of this type are referred to here as minimalist mimics. The core hypothesis of this paper is that small sets of such scaffolds can be designed to analogue local pairs of amino acids (including noncontiguous ones) in any secondary structure; i.e., they are universal peptidomimetics. To illustrate this concept, we designed a set of four peptidomimetic scaffolds. Libraries based on them were made bearing side chains corresponding to many of the protein-derived amino acids. Modeling experiments were performed to give an indication of kinetic and thermodynamic accessibilities of conformations that can mimic secondary structures. Together, peptidomimetics based on these four scaffolds can adopt conformations that resemble almost any combination of local amino acid side chains in any secondary structure. Universal peptidomimetics of this kind are likely to be most useful in the design of libraries for high-throughput screening against diverse targets. Consequently, data arising from submission of these molecules to the NIH Molecular Libraries Small Molecule Repository (MLSMR) are outlined.
Collapse
Affiliation(s)
- Eunhwa Ko
- Department of Chemistry and Laboratory for Molecular Simulation, Texas A&M University, Box 30012, College Station, Texas 77842, United States
| | | | | | | | | | | |
Collapse
|
39
|
Regazzoni L, Bertoletti L, Vistoli G, Colombo R, Aldini G, Serra M, Carini M, Caccialanza G, De Lorenzi E. A combined high-resolution mass spectrometric and in silico approach for the characterisation of small ligands of beta2-microglobulin. ChemMedChem 2010; 5:1015-25. [PMID: 20544784 DOI: 10.1002/cmdc.201000082] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/07/2022]
Abstract
Beta(2)-microglobulin (beta(2)-m) is a protein responsible for a severe complication of long-term hemodialysis, known as dialysis-related amyloidosis, in which initial beta(2)-m misfolding leads to amyloid fibril deposition, mainly in the skeletal tissue. Whereas much attention is paid to understanding the complex mechanism of amyloid formation, the evaluation of small molecules that may bind beta(2)-m and possibly inhibit the aggregation process is still largely unexplored mainly because the protein lacks a specific active site. Based on our previous findings, we selected a pilot set of sulfonated molecules that are known to either bind or not to the protein, including binders that are anti-amyloidogenic. We show how a complementary approach, using high-resolution mass spectrometry and in silico studies, can offer rapid and precise information on affinity, as well as insight into the structural requisites that favour or disfavour the inhibitory activity. Overall, this approach can be used for predictive purposes and for a rapid screening of fibrillogenesis inhibitors.
Collapse
Affiliation(s)
- Luca Regazzoni
- Dipartimento di Scienze Farmaceutiche Pietro Pratesi, Università degli Studi di Milano, Via Mangiagalli 25, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sigalov AB. The SCHOOL of nature: III. From mechanistic understanding to novel therapies. SELF/NONSELF 2010; 1:192-224. [PMID: 21487477 PMCID: PMC3047783 DOI: 10.4161/self.1.3.12794] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/27/2010] [Revised: 06/10/2010] [Accepted: 06/11/2010] [Indexed: 11/19/2022]
Abstract
Protein-protein interactions play a central role in biological processes and thus represent an appealing target for innovative drug design and development. They can be targeted by small molecule inhibitors, modulatory peptides and peptidomimetics, which represent a superior alternative to protein therapeutics that carry many disadvantages. Considering that transmembrane signal transduction is an attractive process to therapeutically control multiple diseases, it is fundamentally and clinically important to mechanistically understand how signal transduction occurs. Uncovering specific protein-protein interactions critical for signal transduction, a general platform for receptor-mediated signaling, the signaling chain homooligomerization (SCHOOL) platform, suggests these interactions as universal therapeutic targets. Within the platform, the general principles of signaling are similar for a variety of functionally unrelated receptors. This suggests that global therapeutic strategies targeting key protein-protein interactions involved in receptor triggering and transmembrane signal transduction may be used to treat a diverse set of diseases. This also assumes that clinical knowledge and therapeutic strategies can be transferred between seemingly disparate disorders, such as T cell-mediated skin diseases and platelet disorders or combined to develop novel pharmacological approaches. Intriguingly, human viruses use the SCHOOL-like strategies to modulate and/or escape the host immune response. These viral mechanisms are highly optimized over the millennia, and the lessons learned from viral pathogenesis can be used practically for rational drug design. Proof of the SCHOOL concept in the development of novel therapies for atopic dermatitis, rheumatoid arthritis, cancer, platelet disorders and other multiple indications with unmet needs opens new horizons in therapeutics.
Collapse
|
41
|
Buchwald P, Margolles-Clark E, Kenyon NS, Ricordi C. Organic dyes as small molecule protein-protein interaction inhibitors for the CD40-CD154 costimulatory interaction. J Mol Recognit 2010; 23:65-73. [PMID: 19621420 DOI: 10.1002/jmr.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
It is becoming increasingly clear that small molecules can often act as effective protein-protein interaction (PPI) inhibitors, an area of increasing interest for its many possible therapeutic applications. We have identified several organic dyes and related small molecules that (i) concentration-dependently inhibit the important CD40-CD154 costimulatory interaction with activities in the low micromolar (microM) range, (ii) show selectivity toward this particular PPI, (iii) seem to bind on the surface of CD154, and (iv) concentration-dependently inhibit the CD154-induced B cell proliferation. They were identified through an iterative activity screening/structural similarity search procedure starting with suramin as lead, and the best smaller compounds, the main focus of the present work, achieved an almost 3-fold increase in ligand efficiency (DeltaG(0)/nonhydrogen atom = 0.8 kJ/N(nHa)) approaching the average of known promising small-molecule PPI inhibitors (approximately 1.0 kJ/N(nHa)). Since CD154 is a member of the tumor necrosis factor (TNF) superfamily of cell surface interaction molecules, inhibitory activities on the TNF-R1-TNF-alpha interactions were also determined to test for specificity, and the compounds selected here all showed more than 30-fold selectivity toward the CD40-CD154 interaction. Because of their easy availability in various structural scaffolds and because of their good protein-binding ability, often explored for tissue-specific staining and other purposes, such organic dyes can provide a valuable addition to the chemical space searched to identify small molecule PPI inhibitors in general.
Collapse
Affiliation(s)
- Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA.
| | | | | | | |
Collapse
|
42
|
Sigalov AB. The SCHOOL of nature: I. Transmembrane signaling. SELF/NONSELF 2010; 1:4-39. [PMID: 21559175 PMCID: PMC3091606 DOI: 10.4161/self.1.1.10832] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/05/2009] [Revised: 11/30/2009] [Accepted: 12/01/2009] [Indexed: 11/19/2022]
Abstract
Receptor-mediated transmembrane signaling plays an important role in health and disease. Recent significant advances in our understanding of the molecular mechanisms linking ligand binding to receptor activation revealed previously unrecognized striking similarities in the basic structural principles of function of numerous cell surface receptors. In this work, I demonstrate that the Signaling Chain Homooligomerization (SCHOOL)-based mechanism represents a general biological mechanism of transmembrane signal transduction mediated by a variety of functionally unrelated single- and multichain activating receptors. within the SCHOOL platform, ligand binding-induced receptor clustering is translated across the membrane into protein oligomerization in cytoplasmic milieu. This platform resolves a long-standing puzzle in transmembrane signal transduction and reveals the major driving forces coupling recognition and activation functions at the level of protein-protein interactions-biochemical processes that can be influenced and controlled. The basic principles of transmembrane signaling learned from the SCHOOL model can be used in different fields of immunology, virology, molecular and cell biology and others to describe, explain and predict various phenomena and processes mediated by a variety of functionally diverse and unrelated receptors. Beyond providing novel perspectives for fundamental research, the platform opens new avenues for drug discovery and development.
Collapse
Affiliation(s)
- Alexander B Sigalov
- Department of Pathology; University of Massachusetts Medical School; Worcester, MA USA
| |
Collapse
|
43
|
|
44
|
|
45
|
Marimganti S, Cheemala MN, Ahn JM. Novel Amphiphilic α-Helix Mimetics Based on a Bis-benzamide Scaffold. Org Lett 2009; 11:4418-21. [PMID: 19719090 DOI: 10.1021/ol901785v] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/25/2022]
Affiliation(s)
- Srinivasa Marimganti
- Department of Chemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080
| | - Murthy N. Cheemala
- Department of Chemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080
| | - Jung-Mo Ahn
- Department of Chemistry, University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080
| |
Collapse
|
46
|
Small-molecule costimulatory blockade: organic dye inhibitors of the CD40–CD154 interaction. J Mol Med (Berl) 2009; 87:1133-43. [DOI: 10.1007/s00109-009-0519-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2009] [Revised: 07/29/2009] [Accepted: 08/06/2009] [Indexed: 02/08/2023]
|
47
|
Verma V, Larsen BD, Coombs W, Lin X, Spagnol G, Sorgen PL, Taffet SM, Delmar M. Novel pharmacophores of connexin43 based on the "RXP" series of Cx43-binding peptides. Circ Res 2009; 105:176-84. [PMID: 19556520 DOI: 10.1161/circresaha.109.200576] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
Gap junction pharmacology is a nascent field. Previous studies have identified molecules that enhance intercellular communication, and may offer potential for innovative antiarrhythmic therapy. However, their specific molecular target(s) and mechanism(s) of action remain unknown. Previously, we identified a 34-aa peptide (RXP-E) that binds the carboxyl terminal domain of Cx43 (Cx43CT) and prevents cardiac gap junction closure and action potential propagation block. These results supported the feasibility of a peptide-based pharmacology to Cx43, but the structure of the core active element in RXP-E, an essential step for pharmacological development, remained undefined. Here, we used a combination of molecular modeling, surface plasmon resonance, nuclear magnetic resonance and patch-clamp strategies to define, for the first time, a unique ensemble of pharmacophores that bind Cx43CT and prevent closure of Cx43 channels. Two particular molecules are best representatives of this family: a cyclized heptapeptide (called CyRP-71) and a linear octapeptide of sequence RRNYRRNY. These 2 small compounds offer the first structural platform for the design of Cx43-interacting gap junction openers. Moreover, the structure of these compounds offers an imprint of a region of Cx43CT that is fundamental to gap junction channel function.
Collapse
Affiliation(s)
- Vandana Verma
- Center for Arrhythmia Research, University of Michigan Medical School, 5025 Venture Dr, Ann Arbor MI 48104, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Fustero S, Mateu N, Albert L, Aceña JL. Straightforward Stereoselective Access to Cyclic Peptidomimetics. J Org Chem 2009; 74:4429-32. [DOI: 10.1021/jo900679c] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/03/2023]
Affiliation(s)
- Santos Fustero
- Departamento de Química Orgánica, Universidad de Valencia, E-46100 Burjassot, Spain, and Laboratorio de Moléculas Orgánicas, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - Natalia Mateu
- Departamento de Química Orgánica, Universidad de Valencia, E-46100 Burjassot, Spain, and Laboratorio de Moléculas Orgánicas, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - Laia Albert
- Departamento de Química Orgánica, Universidad de Valencia, E-46100 Burjassot, Spain, and Laboratorio de Moléculas Orgánicas, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - José Luis Aceña
- Departamento de Química Orgánica, Universidad de Valencia, E-46100 Burjassot, Spain, and Laboratorio de Moléculas Orgánicas, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| |
Collapse
|
49
|
Abstract
Unique properties of metal complexes, such as structural diversity, adjustable ligand exchange kinetics, fine-tuned redox activities, and distinct spectroscopic signatures, make them exciting scaffolds not only for binding to nucleic acids but increasingly also to proteins as non-traditional targets. This feature article discusses recent trends in this field. These include the use of chemically inert metal complexes as structural scaffolds for the design of enzyme inhibitors, new strategies for inducing selective coordination chemistry at the protein binding site, recent advances in the development of catalytic enzyme inhibitors, and the design of metal complexes that can inject electrons or holes into redox enzymes. A common theme in many of the discussed examples is that binding selectivity is at least in part achieved through weak interactions between the ligand sphere and the protein binding site. These examples hint to an exciting future in which "organic-like" molecular recognition principles are combined with properties that are unique to metals and thus promise to yield compounds with novel and unprecedented properties.
Collapse
Affiliation(s)
- Eric Meggers
- Fachbereich Chemie, Philipps-Universität Marburg, Hans-Meerwein-Strasse, 35043, Marburg, Germany.
| |
Collapse
|
50
|
|