1
|
Mi D, Li C, Li Y, Yao M, Li Y, Hong K, Xie C, Chen Y. Discovery of novel BCL6-Targeting PROTACs with effective antitumor activities against DLBCL in vitro and in vivo. Eur J Med Chem 2024; 277:116789. [PMID: 39208743 DOI: 10.1016/j.ejmech.2024.116789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
The transcriptional repressor B cell lymphoma 6 (BCL6) plays a critical role in driving tumorigenesis of diffuse large B-cell lymphoma (DLBCL). However, the therapeutic potential of inhibiting or degrading BCL6 for DLBCL has not been thoroughly understood. Herein, we reported the discovery of a series of novel BCL6-targeting PROTACs based on our previously reported N-phenyl-4-pyrimidinamine BCL6 inhibitors. The optimal compound DZ-837 degraded BCL6 with DC50 values around 600 nM and effectively inhibited the proliferation of several DLBCL cell lines. Further study indicated that DZ-837 induced significant G1 phase arrest and exhibited sustained reactivation of BCL6 downstream genes. In the SU-DHL-4 xenograft model, DZ-837 significantly inhibited tumor growth with TGI of 71.8 % at 40 mg/kg once daily. Furthermore, the combination of DZ-837 with BTK inhibitor Ibrutinib showed synergistic effects and overcame acquired resistance against DLBCL cells. Overall, our findings demonstrate that DZ-837 is an effective BCL6 degrader for DLBCL treatment as a monotherapy or in combination with Ibrutinib.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-6/metabolism
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Animals
- Cell Proliferation/drug effects
- Mice
- Structure-Activity Relationship
- Drug Discovery
- Drug Screening Assays, Antitumor
- Molecular Structure
- Dose-Response Relationship, Drug
- Cell Line, Tumor
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Proteolysis Targeting Chimera
Collapse
Affiliation(s)
- Dazhao Mi
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Cheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yuzhan Li
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | | | - Yan Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Keyu Hong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Lingang Laboratory, Shanghai, 200031, China
| | - Chengying Xie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Lingang Laboratory, Shanghai, 200031, China.
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China; School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunna, 650500, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, Yunnan, 650500, China.
| |
Collapse
|
2
|
Cushing VI, Koh AF, Feng J, Jurgaityte K, Bondke A, Kroll SHB, Barbazanges M, Scheiper B, Bahl AK, Barrett AGM, Ali S, Kotecha A, Greber BJ. High-resolution cryo-EM of the human CDK-activating kinase for structure-based drug design. Nat Commun 2024; 15:2265. [PMID: 38480681 PMCID: PMC10937634 DOI: 10.1038/s41467-024-46375-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Rational design of next-generation therapeutics can be facilitated by high-resolution structures of drug targets bound to small-molecule inhibitors. However, application of structure-based methods to macromolecules refractory to crystallization has been hampered by the often-limiting resolution and throughput of cryogenic electron microscopy (cryo-EM). Here, we use high-resolution cryo-EM to determine structures of the CDK-activating kinase, a master regulator of cell growth and division, in its free and nucleotide-bound states and in complex with 15 inhibitors at up to 1.8 Å resolution. Our structures provide detailed insight into inhibitor interactions and networks of water molecules in the active site of cyclin-dependent kinase 7 and provide insights into the mechanisms contributing to inhibitor selectivity, thereby providing the basis for rational design of next-generation therapeutics. These results establish a methodological framework for the use of high-resolution cryo-EM in structure-based drug design.
Collapse
Affiliation(s)
- Victoria I Cushing
- The Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London, SW3 6JB, UK
| | - Adrian F Koh
- Materials and Structural Analysis Division, Thermo Fisher Scientific, Achtseweg Noord 5, 5651, Eindhoven, The Netherlands
| | - Junjie Feng
- The Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London, SW3 6JB, UK
| | - Kaste Jurgaityte
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | | | | - Marion Barbazanges
- Department of Chemistry, Imperial College London, London, UK
- Institut Parisien de Chimie Moléculaire, Sorbonne Université, CNRS, 4 Place Jussieu, 75252, Paris Cedex 05, France
| | - Bodo Scheiper
- Department of Chemistry, Imperial College London, London, UK
| | - Ash K Bahl
- Carrick Therapeutics, Nova UCD, Bellfield Innovation Park, Dublin 4, Ireland
| | | | - Simak Ali
- Division of Cancer, Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK.
| | - Abhay Kotecha
- Materials and Structural Analysis Division, Thermo Fisher Scientific, Achtseweg Noord 5, 5651, Eindhoven, The Netherlands.
| | - Basil J Greber
- The Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London, SW3 6JB, UK.
| |
Collapse
|
3
|
de Groot AP, de Haan G. How CBX proteins regulate normal and leukemic blood cells. FEBS Lett 2024. [PMID: 38426219 DOI: 10.1002/1873-3468.14839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Hematopoietic stem cell (HSC) fate decisions are dictated by epigenetic landscapes. The Polycomb Repressive Complex 1 (PRC1) represses genes that induce differentiation, thereby maintaining HSC self-renewal. Depending on which chromobox (CBX) protein (CBX2, CBX4, CBX6, CBX7, or CBX8) is part of the PRC1 complex, HSC fate decisions differ. Here, we review how this occurs. We describe how CBX proteins dictate age-related changes in HSCs and stimulate oncogenic HSC fate decisions, either as canonical PRC1 members or by alternative interactions, including non-epigenetic regulation. CBX2, CBX7, and CBX8 enhance leukemia progression. To target, reprogram, and kill leukemic cells, we suggest and describe multiple therapeutic strategies to interfere with the epigenetic functions of oncogenic CBX proteins. Future studies should clarify to what extent the non-epigenetic function of cytoplasmic CBX proteins is important for normal, aged, and leukemic blood cells.
Collapse
Affiliation(s)
- Anne P de Groot
- European Research Institute for Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), The Netherlands
- Sanquin Research, Landsteiner Laboratory, Sanquin Blood Supply, Amsterdam, The Netherlands
| | - Gerald de Haan
- European Research Institute for Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), The Netherlands
- Sanquin Research, Landsteiner Laboratory, Sanquin Blood Supply, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC, University of Amsterdam, The Netherlands
| |
Collapse
|
4
|
Harnden A, Davis OA, Box GM, Hayes A, Johnson LD, Henley AT, de Haven Brandon AK, Valenti M, Cheung KMJ, Brennan A, Huckvale R, Pierrat OA, Talbot R, Bright MD, Akpinar HA, Miller DSJ, Tarantino D, Gowan S, de Klerk S, McAndrew PC, Le Bihan YV, Meniconi M, Burke R, Kirkin V, van Montfort RLM, Raynaud FI, Rossanese OW, Bellenie BR, Hoelder S. Discovery of an In Vivo Chemical Probe for BCL6 Inhibition by Optimization of Tricyclic Quinolinones. J Med Chem 2023; 66:5892-5906. [PMID: 37026591 PMCID: PMC10150366 DOI: 10.1021/acs.jmedchem.3c00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Indexed: 04/08/2023]
Abstract
B-cell lymphoma 6 (BCL6) is a transcriptional repressor and oncogenic driver of diffuse large B-cell lymphoma (DLBCL). Here, we report the optimization of our previously reported tricyclic quinolinone series for the inhibition of BCL6. We sought to improve the cellular potency and in vivo exposure of the non-degrading isomer, CCT373567, of our recently published degrader, CCT373566. The major limitation of our inhibitors was their high topological polar surface areas (TPSA), leading to increased efflux ratios. Reducing the molecular weight allowed us to remove polarity and decrease TPSA without considerably reducing solubility. Careful optimization of these properties, as guided by pharmacokinetic studies, led to the discovery of CCT374705, a potent inhibitor of BCL6 with a good in vivo profile. Modest in vivo efficacy was achieved in a lymphoma xenograft mouse model after oral dosing.
Collapse
Affiliation(s)
- Alice
C. Harnden
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Owen A. Davis
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Gary M. Box
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Angela Hayes
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Louise D. Johnson
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alan T. Henley
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alexis K. de Haven Brandon
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Melanie Valenti
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Kwai-Ming J. Cheung
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alfie Brennan
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rosemary Huckvale
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Olivier A. Pierrat
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rachel Talbot
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Michael D. Bright
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Hafize Aysin Akpinar
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Daniel S. J. Miller
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Dalia Tarantino
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Sharon Gowan
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Selby de Klerk
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Peter Craig McAndrew
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Yann-Vaï Le Bihan
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Mirco Meniconi
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rosemary Burke
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Vladimir Kirkin
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rob L. M. van Montfort
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Florence I. Raynaud
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Olivia W. Rossanese
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Benjamin R. Bellenie
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| | - Swen Hoelder
- Centre
for Cancer Drug Discovery and Division of Structural Biology, The Institute of Cancer Research, London SM2 5NG, U.K.
| |
Collapse
|
5
|
Gu H, He J, Li Y, Mi D, Guan T, Guo W, Liu B, Chen Y. B-cell Lymphoma 6 Inhibitors: Current Advances and Prospects of Drug Development for Diffuse Large B-cell Lymphomas. J Med Chem 2022; 65:15559-15583. [PMID: 36441945 DOI: 10.1021/acs.jmedchem.2c01433] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
B-cell lymphoma 6 (BCL6) is a transcriptional repressor that regulates the differentiation of B lymphocytes and mediates the formation of germinal centers (GCs) by recruiting corepressors through the BTB domain of BCL6. Physiological processes regulated by BCL6 involve cell activation, differentiation, DNA damage, and apoptosis. BCL6 is highly expressed when the gene is mutated, leading to the malignant proliferation of cells and drives tumorigenesis. BCL6 overexpression is closely correlated with tumorigenesis in diffuse large B-cell lymphoma (DLBCL) and other lymphomas, and BCL6 inhibitors can effectively inhibit some lymphomas and overcome resistance. Therefore, targeting BCL6 might be a promising therapeutic strategy for treating lymphomas. Herein, we comprehensively review the latest development of BCL6 inhibitors in diffuse large B-cell lymphoma and discuss the overview of the pharmacophores of BCL6 inhibitors and their efficacies in vitro and in vivo. Additionally, the current advances in BCL6 degraders are provided.
Collapse
Affiliation(s)
- Haijun Gu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jia He
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuzhan Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dazhao Mi
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Tian Guan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Weikai Guo
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Bo Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yihua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
6
|
Karimi S, Shahabi F, Mubarak SMH, Arjmandi H, Hashemi ZS, Pourzardosht N, Zakeri A, Mahboobi M, Jahangiri A, Rahbar MR, Khalili S. Impact of SNPs, off-targets, and passive permeability on efficacy of BCL6 degrading drugs assigned by virtual screening and 3D-QSAR approach. Sci Rep 2022; 12:21091. [PMID: 36473934 PMCID: PMC9726907 DOI: 10.1038/s41598-022-25587-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
B-cell lymphoma 6 (BCL6) regulates various genes and is reported to be overexpressed in lymphomas and other malignancies. Thus, BCL6 inhibition or its tagging for degradation would be an amenable therapeutic approach. A library of 2500 approved drugs was employed to find BCL6 inhibitory molecules via virtual screening. Moreover, the 3D core structure of 170 BCL6 inhibitors was used to build a 3D QSAR model and predict the biological activity. The SNP database was analyzed to study the impact on the destabilization of BCL6/drug interactions. Structural similarity search and molecular docking analyses were used to assess the interaction between possible off-targets and BCL6 inhibitors. The tendency of drugs for passive membrane permeability was also analyzed. Lifitegrast (DB11611) had favorable binding properties and biological activity compared to the BI-3802. Missense SNPs were located at the essential interaction sites of the BCL6. Structural similarity search resulted in five BTB-domain containing off-target proteins. BI-3802 and Lifitegrast had similar chemical behavior and binding properties against off-target candidates. More interestingly, the binding affinity of BI-3802 (against off-targets) was higher than Lifitegrast. Energetically, Lifitegrast was less favorable for passive membrane permeability. The interaction between BCL6 and BI-3802 is more prone to SNP-derived variations. On the other hand, higher nonspecific binding of BI-3802 to off-target proteins could bring about higher undesirable properties. It should also be noted that energetically less desirable passive membrane translocation of Lifitegrast would demand drug delivery vehicles. However, further empirical evaluation of Lifitegrast would unveil its true potential.
Collapse
Affiliation(s)
- Solmaz Karimi
- grid.419305.a0000 0001 1943 2944Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Farzaneh Shahabi
- grid.411747.00000 0004 0418 0096Faculty of Advanced Technologies in Medical Sciences, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shaden M. H. Mubarak
- grid.442852.d0000 0000 9836 5198Department of Clinical Laboratory Science, Faculty of Pharmacy, University of Kufa, Najaf, Iraq
| | - Hanie Arjmandi
- grid.467532.10000 0004 4912 2930Faculty of Pharmacy, Islamic Azad University of Amol Branch, Amol, Iran
| | - Zahra Sadat Hashemi
- grid.417689.5ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Navid Pourzardosht
- grid.411874.f0000 0004 0571 1549Biochemistry Department, Guilan University of Medical Sciences, Rasht, Iran
| | - Alireza Zakeri
- grid.440791.f0000 0004 0385 049XDepartment of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mahdieh Mahboobi
- grid.411521.20000 0000 9975 294XApplied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abolfazl Jahangiri
- grid.411521.20000 0000 9975 294XApplied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rahbar
- grid.412571.40000 0000 8819 4698Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Khalili
- grid.440791.f0000 0004 0385 049XDepartment of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
7
|
Wang Z, Ren J, Jia K, Zhao Y, Liang L, Cheng Z, Huang F, Zhao X, Cheng J, Song S, Sheng T, Wan W, Shu Q, Wu D, Zhang J, Lu T, Chen Y, Ran T, Lu S. Identification and structural analysis of a selective tropomyosin receptor kinase C (TRKC) inhibitor. Eur J Med Chem 2022; 241:114601. [PMID: 35872544 DOI: 10.1016/j.ejmech.2022.114601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 07/07/2022] [Indexed: 11/04/2022]
Abstract
Tropomyosin receptor kinases (TRKs) are a family of TRKA, TRKB and TRKC isoforms. It has been widely reported that TRKs are implicated in a variety of tumors with several Pan-TRK inhibitors currently being used or evaluated in clinical treatment. However, off-target adverse events frequently occur in the clinical use of Pan-TRK inhibitors, which result in poor patient compliance, even drug discontinuation. Although a subtype-selectivity TRK inhibitor may avert the potential off-target adverse events and can act as a more powerful tool compound in the biochemical studies on TRKs, the high sequence similarities of TRKs hinder the development of subtype-selectivity TRK inhibitors. For example, no selective TRKC inhibitor has been reported. Herein, a selective TRKC inhibitor (L13) was disclosed, with potent TRKC inhibitory activity and 107.5-/34.9-fold selectivity over TRKA/B (IC50 TRKA/B/C = 1400 nM, 454 nM, 13 nM, respectively). Extensive molecular dynamics simulations illustrated that key interactions of L13 with the residues and diversely conserved water molecules in the ribose regions of different TRKs may be the structural basis of selectivity. This will provide inspiring insights into the development of subtype-selectivity TRK inhibitors. Moreover, L13 could serve as a tool compound to investigate the distinct biological functions of TRKC and a starting point for further research on drugs specifically targeting TRKC.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jiwei Ren
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Kun Jia
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yuming Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, PR China
| | - Li Liang
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Zitian Cheng
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Fei Huang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Xiaofei Zhao
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Shiyu Song
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, 210038, PR China
| | - Tiancheng Sheng
- School of Engineering, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Weiqi Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Qingqing Shu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Donglin Wu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Junhao Zhang
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Ting Ran
- Drug and Vaccine Research Center, Guangzhou Laboratory, Guangzhou, 510005, PR China.
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
8
|
Davis OA, Cheung KMJ, Brennan A, Lloyd MG, Rodrigues MJ, Pierrat OA, Collie GW, Le Bihan YV, Huckvale R, Harnden AC, Varela A, Bright MD, Eve P, Hayes A, Henley AT, Carter MD, McAndrew PC, Talbot R, Burke R, van Montfort RLM, Raynaud FI, Rossanese OW, Meniconi M, Bellenie BR, Hoelder S. Optimizing Shape Complementarity Enables the Discovery of Potent Tricyclic BCL6 Inhibitors. J Med Chem 2022; 65:8169-8190. [PMID: 35657291 PMCID: PMC9234963 DOI: 10.1021/acs.jmedchem.1c02174] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Indexed: 11/30/2022]
Abstract
To identify new chemical series with enhanced binding affinity to the BTB domain of B-cell lymphoma 6 protein, we targeted a subpocket adjacent to Val18. With no opportunities for strong polar interactions, we focused on attaining close shape complementarity by ring fusion onto our quinolinone lead series. Following exploration of different sized rings, we identified a conformationally restricted core which optimally filled the available space, leading to potent BCL6 inhibitors. Through X-ray structure-guided design, combined with efficient synthetic chemistry to make the resulting novel core structures, a >300-fold improvement in activity was obtained by the addition of seven heavy atoms.
Collapse
Affiliation(s)
- Owen A. Davis
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Kwai-Ming J. Cheung
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Alfie Brennan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Matthew G. Lloyd
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Matthew J. Rodrigues
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K..
| | - Olivier A. Pierrat
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Gavin W. Collie
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K..
| | - Yann-Vaï Le Bihan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K..
| | - Rosemary Huckvale
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Alice C. Harnden
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Ana Varela
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Michael D. Bright
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Paul Eve
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Angela Hayes
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Alan T. Henley
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Michael D. Carter
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - P. Craig McAndrew
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Rachel Talbot
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Rob L. M. van Montfort
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K..
| | - Florence I. Raynaud
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Olivia W. Rossanese
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Mirco Meniconi
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Benjamin R. Bellenie
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| | - Swen Hoelder
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K..
| |
Collapse
|
9
|
Huckvale R, Harnden AC, Cheung KMJ, Pierrat OA, Talbot R, Box GM, Henley AT, de Haven Brandon AK, Hallsworth AE, Bright MD, Akpinar HA, Miller DSJ, Tarantino D, Gowan S, Hayes A, Gunnell EA, Brennan A, Davis OA, Johnson LD, de Klerk S, McAndrew C, Le Bihan YV, Meniconi M, Burke R, Kirkin V, van Montfort RLM, Raynaud FI, Rossanese OW, Bellenie BR, Hoelder S. Improved Binding Affinity and Pharmacokinetics Enable Sustained Degradation of BCL6 In Vivo. J Med Chem 2022; 65:8191-8207. [PMID: 35653645 PMCID: PMC9234961 DOI: 10.1021/acs.jmedchem.1c02175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Indexed: 11/30/2022]
Abstract
The transcriptional repressor BCL6 is an oncogenic driver found to be deregulated in lymphoid malignancies. Herein, we report the optimization of our previously reported benzimidazolone molecular glue-type degrader CCT369260 to CCT373566, a highly potent probe suitable for sustained depletion of BCL6 in vivo. We observed a sharp degradation SAR, where subtle structural changes conveyed the ability to induce degradation of BCL6. CCT373566 showed modest in vivo efficacy in a lymphoma xenograft mouse model following oral dosing.
Collapse
Affiliation(s)
- Rosemary Huckvale
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alice C. Harnden
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Kwai-Ming J. Cheung
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Olivier A. Pierrat
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rachel Talbot
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Gary M. Box
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Alan T. Henley
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | | | - Albert E. Hallsworth
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Michael D. Bright
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Hafize Aysin Akpinar
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Daniel S. J. Miller
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Dalia Tarantino
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Sharon Gowan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Angela Hayes
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Emma A. Gunnell
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K.
| | - Alfie Brennan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Owen A. Davis
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Louise D. Johnson
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Selby de Klerk
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Craig McAndrew
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Yann-Vaï Le Bihan
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K.
| | - Mirco Meniconi
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Vladimir Kirkin
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rob L. M. van Montfort
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
- Division
of Structural Biology, The Institute of
Cancer Research, London SM2 5NG, U.K.
| | - Florence I. Raynaud
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Olivia W. Rossanese
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Benjamin R. Bellenie
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| | - Swen Hoelder
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London SM2 5NG, U.K.
| |
Collapse
|