1
|
Xu Z, Chu M. Advances in Immunosuppressive Agents Based on Signal Pathway. Front Pharmacol 2022; 13:917162. [PMID: 35694243 PMCID: PMC9178660 DOI: 10.3389/fphar.2022.917162] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
Immune abnormality involves in various diseases, such as infection, allergic diseases, autoimmune diseases, as well as transplantation. Several signal pathways have been demonstrated to play a central role in the immune response, including JAK/STAT, NF-κB, PI3K/AKT-mTOR, MAPK, and Keap1/Nrf2/ARE pathway, in which multiple targets have been used to develop immunosuppressive agents. In recent years, varieties of immunosuppressive agents have been approved for clinical use, such as the JAK inhibitor tofacitinib and the mTOR inhibitor everolimus, which have shown good therapeutic effects. Additionally, many immunosuppressive agents are still in clinical trials or preclinical studies. In this review, we classified the immunosuppressive agents according to the immunopharmacological mechanisms, and summarized the phase of immunosuppressive agents.
Collapse
Affiliation(s)
- Zhiqing Xu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Pharmacology, Jilin University, Changchun, China
| | - Ming Chu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
2
|
Mao Y, Li S, Gong B, Lai L, He G, Li H. ePharmer: An integrated and graphical software for pharmacophore-based virtual screening. J Comput Chem 2021; 42:2181-2195. [PMID: 34410013 DOI: 10.1002/jcc.26743] [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: 02/02/2021] [Revised: 05/03/2021] [Accepted: 06/01/2021] [Indexed: 11/09/2022]
Abstract
Pharmacophore-based virtual screening (VS) has emerged as an efficient computer-aided drug design technique when appraising multiple ligands with similar structures or targets with unknown crystal structures. Current pharmacophore modeling and analysis software suffers from inadequate integration of mainstream methods and insufficient user-friendly program interface. In this study, we propose a stand-alone, integrated, graphical software for pharmacophore-based VS, termed ePharmer. Both ligand-based and structure-based pharmacophore generation methods were integrated into a compact architecture. Fine-grained modules were carefully organized into the computing, integration, and visualization layers. Graphical design covered the global user interface and specific user operations including editing, evaluation, and task management. Metabolites prediction analysis with the chosen VS result is provided for preselection of wet experiments. Moreover, the underlying computing units largely adopted the preliminary work of our research team. The presented software is currently in client use and will be released for both professional and nonexpert users. Experimental results verified the favorable computing capability, user convenience, and case performance of the proposed software.
Collapse
Affiliation(s)
- Yuxia Mao
- School of Computer Science and Technology, East China Normal University, Shanghai, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai, China
| | - Bojie Gong
- Department of Computer Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Luhua Lai
- BNLMS, Peking-Tsinghua Center for Life Sciences at College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Gaoqi He
- School of Computer Science and Technology, East China Normal University, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science & Technology, Shanghai, China
| |
Collapse
|
3
|
Huang L, Guo Y, Liu S, Wang H, Zhu J, Ou L, Xu X. Targeting regulatory T cells for immunotherapy in melanoma. MOLECULAR BIOMEDICINE 2021; 2:11. [PMID: 34806028 PMCID: PMC8591697 DOI: 10.1186/s43556-021-00038-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are essential in the maintenance of immunity, and they are also a key to immune suppressive microenvironment in solid tumors. Many studies have revealed the biology of Tregs in various human pathologies. Here we review recent understandings of the immunophenotypes and suppressive functions of Tregs in melanoma, including Treg recruitment and expansion in a tumor. Tregs are frequently accumulated in melanoma and the ratio of CD8+ T cells versus Tregs in the melanoma is predictive for patient survival. Hence, depletion of Tregs is a promising strategy for the enhancement of anti-melanoma immunity. Many recent studies are aimed to target Tregs in melanoma. Distinguishing Tregs from other immune cells and understanding the function of different subsets of Tregs may contribute to better therapeutic efficacy. Depletion of functional Tregs from the tumor microenvironment has been tested to induce clinically relevant immune responses against melanomas. However, the lack of Treg specific therapeutic antibodies or Treg specific depleting strategies is a big hurdle that is yet to be overcome. Additional studies to fine-tune currently available therapies and more agents that specifically and selectively target tumor infiltrating Tregs in melanoma are urgently needed.
Collapse
Affiliation(s)
- Lili Huang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Yeye Guo
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Huaishan Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Jinjin Zhu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Lingling Ou
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
4
|
Brotherton-Pleiss C, Yue P, Zhu Y, Nakamura K, Chen W, Fu W, Kubota C, Chen J, Alonso-Valenteen F, Mikhael S, Medina-Kauwe L, Tius MA, Lopez-Tapia F, Turkson J. Discovery of Novel Azetidine Amides as Potent Small-Molecule STAT3 Inhibitors. J Med Chem 2021; 64:695-710. [PMID: 33352047 PMCID: PMC7816766 DOI: 10.1021/acs.jmedchem.0c01705] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Indexed: 02/07/2023]
Abstract
We optimized our previously reported proline-based STAT3 inhibitors into an exciting new series of (R)-azetidine-2-carboxamide analogues that have sub-micromolar potencies. 5a, 5o, and 8i have STAT3-inhibitory potencies (IC50) of 0.55, 0.38, and 0.34 μM, respectively, compared to potencies greater than 18 μM against STAT1 or STAT5 activity. Further modifications derived analogues, including 7e, 7f, 7g, and 9k, that addressed cell membrane permeability and other physicochemical issues. Isothermal titration calorimetry analysis confirmed high-affinity binding to STAT3, with KD of 880 nM (7g) and 960 nM (9k). 7g and 9k inhibited constitutive STAT3 phosphorylation and DNA-binding activity in human breast cancer, MDA-MB-231 or MDA-MB-468 cells. Furthermore, treatment of breast cancer cells with 7e, 7f, 7g, or 9k inhibited viable cells, with an EC50 of 0.9-1.9 μM, cell growth, and colony survival, and induced apoptosis while having relatively weaker effects on normal breast epithelial, MCF-10A or breast cancer, MCF-7 cells that do not harbor constitutively active STAT3.
Collapse
Affiliation(s)
- Christine Brotherton-Pleiss
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Medicinal Chemistry Leader, Department of Chemistry,
University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu,
Hawaii 9682, United States
| | - Peibin Yue
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
| | - Yinsong Zhu
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
| | - Kayo Nakamura
- Department of Chemistry, University of
Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, Hawaii 9682, United
States
| | - Weiliang Chen
- Department of Chemistry, University of
Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, Hawaii 9682, United
States
| | - Wenzhen Fu
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Department of Chemistry, University of
Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, Hawaii 9682, United
States
| | - Casie Kubota
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
| | - Jasmine Chen
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
| | - Felix Alonso-Valenteen
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
- Department of Biomedical Sciences,
Cedars-Sinai Medical Center, Los Angeles, California 90048,
United States
| | - Simoun Mikhael
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
- Department of Biomedical Sciences,
Cedars-Sinai Medical Center, Los Angeles, California 90048,
United States
| | - Lali Medina-Kauwe
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
- Department of Biomedical Sciences,
Cedars-Sinai Medical Center, Los Angeles, California 90048,
United States
| | - Marcus A. Tius
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Medicinal Chemistry Leader, Department of Chemistry,
University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu,
Hawaii 9682, United States
| | - Francisco Lopez-Tapia
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Medicinal Chemistry Leader, Department of Chemistry,
University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu,
Hawaii 9682, United States
| | - James Turkson
- Cancer Biology Program, University of
Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii 96813,
United States
- Department of Medicine, Division of Oncology and
Cedars-Sinai Cancer, Cedars-Sinai Medical
Center, 8700 Beverly Blvd, Davis 5065, Los Angeles, California 90048,
United States
| |
Collapse
|
5
|
Wu KJ, Lei PM, Liu H, Wu C, Leung CH, Ma DL. Mimicking Strategy for Protein-Protein Interaction Inhibitor Discovery by Virtual Screening. Molecules 2019; 24:molecules24244428. [PMID: 31817099 PMCID: PMC6943618 DOI: 10.3390/molecules24244428] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
As protein–protein interactions (PPIs) are highly involved in most cellular processes, the discovery of PPI inhibitors that mimic the structure of the natural protein partners is a promising strategy toward the discovery of PPI inhibitors. In this review, we discuss recent advances in the application of virtual screening for identifying mimics of protein partners. The classification and function of the mimicking protein partner inhibitor discovery by virtual screening are described. We anticipate that this review would be of interest to medicinal chemists and chemical biologists working in the field of protein–protein interaction inhibitors or probes.
Collapse
Affiliation(s)
- Ke-Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (K.-J.W.); (P.-M.L.)
| | - Pui-Man Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (K.-J.W.); (P.-M.L.)
| | - Hao Liu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China; (H.L.); (C.W.)
| | - Chun Wu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China; (H.L.); (C.W.)
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; (K.-J.W.); (P.-M.L.)
- Correspondence: (C.-H.L.); (D.-L.M.); Tel.: +(853)-8822-4688 (C.-H.L.); +(852)-3411-7075 (D.-L.M.)
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China; (H.L.); (C.W.)
- Correspondence: (C.-H.L.); (D.-L.M.); Tel.: +(853)-8822-4688 (C.-H.L.); +(852)-3411-7075 (D.-L.M.)
| |
Collapse
|
6
|
Orlova A, Wagner C, de Araujo ED, Bajusz D, Neubauer HA, Herling M, Gunning PT, Keserű GM, Moriggl R. Direct Targeting Options for STAT3 and STAT5 in Cancer. Cancers (Basel) 2019; 11:E1930. [PMID: 31817042 PMCID: PMC6966570 DOI: 10.3390/cancers11121930] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/22/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Signal transducer and activator of transcription (STAT)3 and STAT5 are important transcription factors that are able to mediate or even drive cancer progression through hyperactivation or gain-of-function mutations. Mutated STAT3 is mainly associated with large granular lymphocytic T-cell leukemia, whereas mutated STAT5B is associated with T-cell prolymphocytic leukemia, T-cell acute lymphoblastic leukemia and γδ T-cell-derived lymphomas. Hyperactive STAT3 and STAT5 are also implicated in various hematopoietic and solid malignancies, such as chronic and acute myeloid leukemia, melanoma or prostate cancer. Classical understanding of STAT functions is linked to their phosphorylated parallel dimer conformation, in which they induce gene transcription. However, the functions of STAT proteins are not limited to their phosphorylated dimerization form. In this review, we discuss the functions and the roles of unphosphorylated STAT3/5 in the context of chromatin remodeling, as well as the impact of STAT5 oligomerization on differential gene expression in hematopoietic neoplasms. The central involvement of STAT3/5 in cancer has made these molecules attractive targets for small-molecule drug development, but currently there are no direct STAT3/5 inhibitors of clinical grade available. We summarize the development of inhibitors against the SH2 domains of STAT3/5 and discuss their applicability as cancer therapeutics.
Collapse
Affiliation(s)
- Anna Orlova
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Christina Wagner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Elvin D. de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (E.D.d.A.); (P.T.G.)
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases (CECAD), and Center for Molecular Medicine Cologne (CMMC), Cologne University, 50937 Cologne, Germany;
| | - Patrick T. Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (E.D.d.A.); (P.T.G.)
- Centre for Medicinal Chemistry, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - György M. Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (D.B.); (G.M.K.)
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, 1210 Vienna, Austria; (A.O.); (C.W.); (H.A.N.)
| |
Collapse
|
7
|
Gelain A, Mori M, Meneghetti F, Villa S. Signal Transducer and Activator of Transcription Protein 3 (STAT3): An Update on its Direct Inhibitors as Promising Anticancer Agents. Curr Med Chem 2019; 26:5165-5206. [PMID: 30027840 DOI: 10.2174/0929867325666180719122729] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/08/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Since Signal Transducer and Activator of Transcription 3 (STAT3) is a transcription factor which plays an important role in multiple aspects of cancer, including progression and migration, and it is constitutively activated in various human tumors, STAT3 inhibition has emerged as a validated strategy for the treatment of several malignancies. The aim of this review is to provide an update on the identification of new promising direct inhibitors targeting STAT3 domains, as potential anticancer agents. METHODS A thorough literature search focused on recently reported STAT3 direct inhibitors was undertaken. We considered the relevant developments regarding the STAT3 domains, which have been identified as potential drug targets. RESULTS In detail, 135 peer-reviewed papers and 7 patents were cited; the inhibitors we took into account targeted the DNA binding domain (compounds were grouped into natural derivatives, small molecules, peptides, aptamers and oligonucleotides), the SH2 binding domain (natural, semi-synthetic and synthetic compounds) and specific residues, like cysteines (natural, semi-synthetic, synthetic compounds and dual inhibitors) and tyrosine 705. CONCLUSION The huge number of direct STAT3 inhibitors recently identified demonstrates a strong interest in the investigation of this target, although it represents a challenging task considering that no drug targeting this enzyme is currently available for anticancer therapy. Notably, many studies on the available inhibitors evidenced that some of them possess a dual mechanism of action.
Collapse
Affiliation(s)
- Arianna Gelain
- Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy
| | - Matteo Mori
- Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy
| | - Fiorella Meneghetti
- Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy
| | - Stefania Villa
- Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Milano, via L. Mangiagalli 25, 20133 Milano, Italy
| |
Collapse
|
8
|
Cryptotanshinone ameliorates the pathogenesis of systemic lupus erythematosus by blocking T cell proliferation. Int Immunopharmacol 2019; 74:105677. [PMID: 31177018 DOI: 10.1016/j.intimp.2019.105677] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 11/20/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, devastating autoimmune disorder associated with severe organ damage. Recently, the role of Signal Transducer and Activator of Transcription 3 (STAT3) in murine lupus has been described, suggesting the involvement of STAT3 signaling in the development of SLE. Cryptotanshinone (CTS) is an effective inhibitor of STAT3; however its potential as a SLE treatment remains to be explored. To determine the function of CTS in SLE, we treated MRL/lpr female mice with CTS. Firstly, we found CTS treatment reversed the elevated STAT3 signaling of spleens in lupus-prone MRL/lpr mice, accompanying with a dramatically decreased number of T cells, especially double-negative (DN) T cells. Further research showed that CTS inhibited T cell proliferation via suppressing of STAT3 activation in vitro and in vivo. Consistently, we also proved that CTS treatment significantly alleviated autoimmune response including notably diminished skin lesions, reduced spleen size and increased life span. In addition, CTS treatment decreased the levels of auto-antibodies and pro-inflammatory cytokines, as well as normalized structure and function of kidneys. All these data suggested that CTS treatment depressed STAT3 phosphorylation, which resulted in blocked DN T cell proliferation and finally attenuated the spontaneous SLE development. Taken together, our data identify CTS as a potential therapeutic drug for SLE patients.
Collapse
|
9
|
Zhou Y, Huang L, Ji S, Hou S, Luo L, Li C, Liu M, Liu Y, Jiang L. Structural Basis for the Inhibition of the Autophosphorylation Activity of HK853 by Luteolin. Molecules 2019; 24:molecules24050933. [PMID: 30866470 PMCID: PMC6429454 DOI: 10.3390/molecules24050933] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/21/2019] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
Abstract
The two-component system (TCS) is a significant signal transduction system for bacteria to adapt to complicated and variable environments, and thus has recently been regarded as a novel target for developing antibacterial agents. The natural product luteolin (Lut) can inhibit the autophosphorylation activity of the typical histidine kinase (HK) HK853 from Thermotoga maritime, but the inhibition mechanism is not known. Herein, we report on the binding mechanism of a typical flavone with HK853 by using solution NMR spectroscopy, isothermal titration calorimetry (ITC), and molecular docking. We show that luteolin inhibits the activity of HK853 by occupying the binding pocket of adenosine diphosphate (ADP) through hydrogen bonds and π-π stacking interaction structurally. Our results reveal a detailed mechanism for the inhibition of flavones and observe the conformational and dynamics changes of HK. These results should provide a feasible approach for antibacterial agent design from the view of the histidine kinases.
Collapse
Affiliation(s)
- Yuan Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Liqun Huang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Shixia Ji
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Shi Hou
- Laboratory of Computer-Aided Drug Design and Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Liang Luo
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Conggang Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Maili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Yixiang Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Ling Jiang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
10
|
Zhang W, Yu W, Cai G, Zhu J, Zhang C, Li S, Guo J, Yin G, Chen C, Kong L. A new synthetic derivative of cryptotanshinone KYZ3 as STAT3 inhibitor for triple-negative breast cancer therapy. Cell Death Dis 2018; 9:1098. [PMID: 30368518 PMCID: PMC6204138 DOI: 10.1038/s41419-018-1139-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 01/10/2023]
Abstract
Silencing STAT3 is confirmed as a promising therapeutic strategy for triple-negative breast cancer (TNBC) therapy to address the issue of its poor prognosis. In this study, the natural product cryptotanshinone was firstly remodeled and modified as a more effective STAT3 inhibitor by structure-based strategy. The synthetic derivative KYZ3 had 22-24-fold increase in antitumor activity than cryptotanshinone on two TNBC cell lines but had little effect on normal breast epithelial MCF-10A cells. Further investigation showed that KYZ3 inhibited persistent STAT3 phosphorylation. It also prevented the STAT3 protein nuclear translocation to regulate the expressions of the target oncogenes including Bax and Bcl-2. Furthermore, KYZ3 inhibited TNBC cell metastasis by decreasing the levels of MMP-9 which were directly regulated by activated STAT3. A STAT3 plasmid transfecting assay suggested that KYZ3 induced tumor cell apoptosis mainly by targeting STAT3. Finally, KYZ3 suppressed the growth of tumors resulting from subcutaneous implantation of MDA-MB-231 cells in vivo. Taken together, KYZ3 may be a promising cancer therapeutic agent for TNBC.
Collapse
Affiliation(s)
- Wenda Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Wenying Yu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China.
| | - Guiping Cai
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Jiawen Zhu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Chao Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Shanshan Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Jianpeng Guo
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Guoping Yin
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Chen Chen
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, 210009, Nanjing, China.
| |
Collapse
|
11
|
Chen L, Lv D, Chen X, Liu M, Wang D, Liu Y, Hong Z, Zhu Z, Hu X, Cao Y, Yang J, Chai Y. Biosensor-Based Active Ingredients Recognition System for Screening STAT3 Ligands from Medical Herbs. Anal Chem 2018; 90:8936-8945. [PMID: 29953204 DOI: 10.1021/acs.analchem.8b01103] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Langdong Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Diya Lv
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Xiaofei Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Mingdong Liu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Yue Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Zhanying Hong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Zhenyu Zhu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Xiaoxia Hu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Yan Cao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Jianmin Yang
- Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| |
Collapse
|
12
|
Ran X, Gestwicki JE. Inhibitors of protein-protein interactions (PPIs): an analysis of scaffold choices and buried surface area. Curr Opin Chem Biol 2018; 44:75-86. [PMID: 29908451 DOI: 10.1016/j.cbpa.2018.06.004] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
Protein-protein interactions (PPI) were once considered 'undruggable', but clinical successes, driven by advanced methods in drug discovery, have challenged that notion. Here, we review the last three years of literature on PPI inhibitors to understand what is working and why. From the 66 recently reported PPI inhibitors, we found that the average molecular weight was significantly greater than 500Da, but that this trend was driven, in large part, by the contribution of peptide-based compounds. Despite differences in average molecular weight, we found that compounds based on small molecules or peptides were almost equally likely to be potent inhibitors (KD<1μM). Finally, we found PPIs with buried surface area (BSA) less than 2000Å2 were more likely to be inhibited by small molecules, while PPIs with larger BSA values were typically inhibited by peptides. PPIs with BSA values over 4000Å2 seemed to create a particular challenge, especially for orthosteric small molecules. Thus, it seems important to choose the inhibitor scaffold based on the properties of the target interaction. Moreover, this survey suggests a (more nuanced) conclusion to the question of whether PPIs are good drug targets; namely, that some PPIs are readily 'druggable' given the right choice of scaffold, while others still seem to deserve the 'undruggable' moniker.
Collapse
Affiliation(s)
- Xu Ran
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States
| | - Jason E Gestwicki
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States.
| |
Collapse
|
13
|
Structural Biology of STAT3 and Its Implications for Anticancer Therapies Development. Int J Mol Sci 2018; 19:ijms19061591. [PMID: 29843450 PMCID: PMC6032208 DOI: 10.3390/ijms19061591] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/21/2018] [Accepted: 05/25/2018] [Indexed: 12/16/2022] Open
Abstract
Transcription factors are proteins able to bind DNA and induce the transcription of specific genes. Consequently, they play a pivotal role in multiple cellular pathways and are frequently over-expressed or dysregulated in cancer. Here, we will focus on a specific “signal transducer and activator of transcription” (STAT3) factor that is involved in several pathologies, including cancer. For long time, the mechanism by which STAT3 exerts its cellular functions has been summarized by a three steps process: (1) Protein phosphorylation by specific kinases, (2) dimerization promoted by phosphorylation, (3) activation of gene expression by the phosphorylated dimer. Consequently, most of the inhibitors reported in literature aimed at blocking phosphorylation and dimerization. However, recent observations reopened the debate and the entire functional mechanism has been revisited stimulating the scientific community to pursue new inhibition strategies. In particular, the dimerization of the unphosphorylated species has been experimentally demonstrated and specific roles proposed also for these dimers. Despite difficulties in the expression and purification of the full length STAT3, structural biology investigations allowed the determination of atomistic structures of STAT3 dimers and several protein domains. Starting from this information, computational methods have been used both to improve the understanding of the STAT3 functional mechanism and to design new inhibitors to be used as anticancer drugs. In this review, we will focus on the contribution of structural biology to understand the roles of STAT3, to design new inhibitors and to suggest new strategies of pharmacological intervention.
Collapse
|
14
|
Xu D, Bum-Erdene K, Si Y, Zhou D, Ghozayel MK, Meroueh SO. Mimicking Intermolecular Interactions of Tight Protein-Protein Complexes for Small-Molecule Antagonists. ChemMedChem 2017; 12:1794-1809. [PMID: 28960868 DOI: 10.1002/cmdc.201700572] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Indexed: 01/12/2023]
Abstract
Tight protein-protein interactions (Kd <100 nm) that occur over a large binding interface (>1000 Å2 ) are highly challenging to disrupt with small molecules. Historically, the design of small molecules to inhibit protein-protein interactions has focused on mimicking the position of interface protein ligand side chains. Here, we explore mimicry of the pairwise intermolecular interactions of the native protein ligand with residues of the protein receptor to enrich commercial libraries for small-molecule inhibitors of tight protein-protein interactions. We use the high-affinity interaction (Kd =1 nm) between the urokinase receptor (uPAR) and its ligand urokinase (uPA) to test our methods. We introduce three methods for rank-ordering small molecules docked to uPAR: 1) a new fingerprint approach that represents uPA's pairwise interaction energies with uPAR residues; 2) a pharmacophore approach to identify small molecules that mimic the position of uPA interface residues; and 3) a combined fingerprint and pharmacophore approach. Our work led to small molecules with novel chemotypes that inhibited a tight uPAR⋅uPA protein-protein interaction with single-digit micromolar IC50 values. We also report the extensive work that identified several of the hits as either lacking stability, thiol reactive, or redox active. This work suggests that mimicking the binding profile of the native ligand and the position of interface residues can be an effective strategy to enrich commercial libraries for small-molecule inhibitors of tight protein-protein interactions.
Collapse
Affiliation(s)
- David Xu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indianapolis, IN, 46202, USA
| | - Khuchtumur Bum-Erdene
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 4023, Indianapolis, IN, 46202, USA
| | - Yubing Si
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Donghui Zhou
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 4023, Indianapolis, IN, 46202, USA
| | - Mona K Ghozayel
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 4023, Indianapolis, IN, 46202, USA
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 4023, Indianapolis, IN, 46202, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
15
|
Gielen F, Butz M, Rees EJ, Erdelyi M, Moschetti T, Hyvönen M, Edel JB, Kaminski CF, Hollfelder F. Quantitative Affinity Determination by Fluorescence Anisotropy Measurements of Individual Nanoliter Droplets. Anal Chem 2017; 89:1092-1101. [PMID: 28192993 PMCID: PMC5287478 DOI: 10.1021/acs.analchem.6b02528] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Fluorescence anisotropy measurements of reagents compartmentalized into individual nanoliter droplets are shown to yield high-resolution binding curves from which precise dissociation constants (Kd) for protein-peptide interactions can be inferred. With the current platform, four titrations can be obtained per minute (based on ∼100 data points each), with stoichiometries spanning more than 2 orders of magnitude and requiring only tens of microliters of reagents. In addition to affinity measurements with purified components, Kd values for unpurified proteins in crude cell lysates can be obtained without prior knowledge of the concentration of the expressed protein, so that protein purification can be avoided. Finally, we show how a competition assay can be set up to perform focused library screens, so that compound labeling is not required anymore. These data demonstrate the utility of droplet compartments for the quantitative characterization of biomolecular interactions and establish fluorescence anisotropy imaging as a quantitative technique in a miniaturized droplet format, which is shown to be as reliable as its macroscopic test tube equivalent.
Collapse
Affiliation(s)
- Fabrice Gielen
- Department of Biochemistry, University of Cambridge , 80 Tennis Court Road, Cambridge, CB2 1GA, United Kingdom.,Living Systems Institute, University of Exeter , Stocker Road, Exeter, EX4 4QD, United Kingdom
| | - Maren Butz
- Department of Biochemistry, University of Cambridge , 80 Tennis Court Road, Cambridge, CB2 1GA, United Kingdom
| | - Eric J Rees
- Department of Chemical Engineering and Biotechnology, New Museums Site , Pembroke Street, Cambridge, CB2 3RA, United Kingdom
| | - Miklos Erdelyi
- Department of Chemical Engineering and Biotechnology, New Museums Site , Pembroke Street, Cambridge, CB2 3RA, United Kingdom.,Department of Optics and Quantum Electronics, University of Szeged , Dom ter 9, Szeged, Hungary
| | - Tommaso Moschetti
- Department of Biochemistry, University of Cambridge , 80 Tennis Court Road, Cambridge, CB2 1GA, United Kingdom
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge , 80 Tennis Court Road, Cambridge, CB2 1GA, United Kingdom
| | - Joshua B Edel
- Department of Chemistry, Imperial College London , South Kensington, London, SW7 2AZ, United Kingdom
| | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, New Museums Site , Pembroke Street, Cambridge, CB2 3RA, United Kingdom
| | - Florian Hollfelder
- Department of Biochemistry, University of Cambridge , 80 Tennis Court Road, Cambridge, CB2 1GA, United Kingdom
| |
Collapse
|
16
|
Yesylevskyy SO, Ramseyer C, Pudlo M, Pallandre JR, Borg C. Selective Inhibition of STAT3 with Respect to STAT1: Insights from Molecular Dynamics and Ensemble Docking Simulations. J Chem Inf Model 2016; 56:1588-96. [PMID: 27479469 DOI: 10.1021/acs.jcim.6b00198] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
STAT3 protein, which is known to be involved in cancer development, is a promising target for anticancer therapy. Successful inhibitors of STAT3 should not affect an activity of closely related protein STAT1, which makes their development challenging. The mechanisms of selectivity of several existing STAT3 inhibitors are not clear. In this work, we studied molecular mechanisms of selectivity of 13 experimentally tested STAT3 inhibitors by means of extensive molecular dynamics and ensemble docking simulations. It is shown that all studied inhibitors bind to the large part of the protein surface in an unspecific statistical manner. The binding to the dimerization interface of the SH2 domain, which is usually considered as the main target region, is not energetically preferable. Binding in this region is remarkably similar for STAT1 and STAT3 proteins and cannot explain experimentally observed selectivity toward STAT3. We propose a new mechanism of selectivity called "selectivity by distraction" for existing STAT3 inhibitors. This mechanism is based on equilibrium statistical partitioning of inhibitor molecules between protein domains. The unspecific binding of inhibitors to the DNA-binding and the coil-coil domains is stronger in STAT1 in comparison to STAT3 while the energies of their binding to SH2 domains are comparable. This "distracts" inhibitor molecules from the SH2 domain of STAT1 and leads to higher effective concentration of inhibitors in the vicinity of the SH2 domain of STAT3.
Collapse
Affiliation(s)
- Semen O Yesylevskyy
- Institute of Physics, National Academy of Sciences of Ukraine , Prospect Nauki, 46, Kyiv, 03028, Ukraine
| | - Christophe Ramseyer
- Laboratoire Chrono Environnement UMR CNRS 6249, Faculté des Sciences et Techniques, La Bouloie, Université Bourgogne Franche-Comté , 25030, Besançon Cedex, France
| | - Marc Pudlo
- Fonctions et Dysfonctions Epitheliales - EA 4267, Universite de Bourgogne Franche-Comté, UFR Sciences Medicales et Pharmaceutiques , 19 rue Ambroise Pare, 25030 BESANCON cedex, France
| | - Jean-René Pallandre
- Inserm UMR 1098, EFS Bourgogne Franche Comté, Université Bourgogne Franche-Comté , IFR133, 8 rue du Dr Girod, 25020 Besançon, France
| | - Christophe Borg
- Inserm UMR 1098, EFS Bourgogne Franche Comté, Université Bourgogne Franche-Comté , IFR133, 8 rue du Dr Girod, 25020 Besançon, France
| |
Collapse
|
17
|
Poli G, Gelain A, Porta F, Asai A, Martinelli A, Tuccinardi T. Identification of a new STAT3 dimerization inhibitor through a pharmacophore-based virtual screening approach. J Enzyme Inhib Med Chem 2015; 31:1011-7. [DOI: 10.3109/14756366.2015.1079184] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy,
| | - Arianna Gelain
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy, and
| | - Federica Porta
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy, and
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | | | |
Collapse
|
18
|
Caldwell GW. In silico tools used for compound selection during target-based drug discovery and development. Expert Opin Drug Discov 2015; 10:901-23. [DOI: 10.1517/17460441.2015.1043885] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Gary W Caldwell
- Janssen Research & Development LLC, Discovery Sciences, Spring House, PA, USA
| |
Collapse
|