1
|
Zhang S, Li GX, Fang D, Zhang X, Chen S, Cui X, Tang Z. Aerobic oxidative bromination and iodination enabled by alloxan and ascorbic acid to mimic flavin-dependent halogenases. Nat Commun 2025; 16:3680. [PMID: 40246838 PMCID: PMC12006302 DOI: 10.1038/s41467-025-58904-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
The fundamentally significant halogenation processes in organic synthesis heavily depend on the use of hazardous and toxic elemental halogens directly or indirectly, inevitably increasing environmental burden and leading to various production problems. Flavin-dependent halogenases (FDHs) in nature produce organic halides under benign conditions, yet their practical application remained elusive and difficult to scale up. Inspired by FDHs, here we develop a mild and eco-friendly biomimetic aerobic oxidative halogenation strategy that utilizes O2 from the air to oxidize non-toxic halide salts as a safer alternative. Using simple alloxan as catalyst and low-cost ascorbic acid as reductant, we successfully emulate the challenging flavoenzymatic aerobic halogenation cycle without requiring light activation. This approach yields various important monobromide and iodide products with high efficiency, excellent selectivity, and good functional group tolerance. Moreover, its successful application in late-stage bromination of complex bioactive molecules, operational ease at the gram scale, and use of cost-effective, unpurified primary raw materials all indicate significant potential for green industrial production.
Collapse
Affiliation(s)
- Shiqi Zhang
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China
| | - Guang-Xun Li
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China
| | - Dongmei Fang
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China
| | - Xia Zhang
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China
| | - Shangjing Chen
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China
| | - Xin Cui
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China
| | - Zhuo Tang
- Center for Natural Products, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
2
|
Chen J, Wang J, Yang W, Zhao L, Su J. Activity Regulation and Conformation Response of Janus Kinase 3 Mediated by Phosphorylation: Exploration from Correlation Network Analysis and Markov Model. J Chem Inf Model 2025. [PMID: 40199555 DOI: 10.1021/acs.jcim.5c00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The activity of the enzyme JAK3 is modulated by tyrosine phosphorylation, yet the underlying molecular details remain not fully understood. In this study, we employed a GaMD trajectory-based Markov model and correlation network analysis (CNA) to investigate the impact of single phosphorylation (SP) at Y980 (pY980) and double phosphorylation (DP) at Y980/Y981 (pY980/pY981) on the conformational dynamics of JAK3 bound by inhibitors IZA and MI1. The Markov model analysis indicated that both SP and DP result in fewer conformational states and significantly influence the conformational dynamics of the P-loop, αC-helix, and loop1-loop3, while maintaining the hinge region's high rigidity. The CNA findings revealed that phosphorylation alters the communication network among different structural regions of JAK3, providing a rational explanation for how phosphorylation affects the conformational dynamics of the distant P-loop and loop1-loop3. Moreover, the conformational changes mediated by SP and DP further affect the interactions between the inhibitors and the hot spots (L828, V836, E903, Y904, L905, and L956) of JAK3. This work offers valuable theoretical insights into the molecular mechanisms that regulate JAK3 activity.
Collapse
Affiliation(s)
- Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Jian Wang
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Wanchun Yang
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Lu Zhao
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Jing Su
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| |
Collapse
|
3
|
Maji L, Sengupta S, Purawarga Matada GS, Teli G, Biswas G, Das PK, Panduranga Mudgal M. Medicinal chemistry perspective of JAK inhibitors: synthesis, biological profile, selectivity, and structure activity relationship. Mol Divers 2024; 28:4467-4513. [PMID: 38236444 DOI: 10.1007/s11030-023-10794-5] [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: 10/26/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024]
Abstract
JAK-STAT signalling pathway was discovered more than quarter century ago. The JAK-STAT pathway protein is considered as one of the crucial hubs for cytokine secretion which mediates activation of different inflammatory, cellular responses and hence involved in different etiological factors. The various etiological factors involved are haematopoiesis, immune fitness, tissue repair, inflammation, apoptosis, and adipogenesis. The presence of the active mutation V617K plays a significant role in the progression of the JAK-STAT pathway-related disease. Consequently, targeting the JAK-STAT pathway could be a promising therapeutic approach for addressing a range of causative factors. In this current review, we provided a comprehensive discussion for the in-detail study of anatomy and physiology of the JAK-STAT pathway which contributes structural domain rearrangement, activation, and negative regulation associated with the downstream signaling pathway, relationship between different cytokines and diseases. This review also discussed the recent development of clinical trial entities. Additionally, this review also provides updates on FDA-approved drugs. In the current investigation, we have classified recently developed small molecule inhibitors of JAK-STAT pathway according to different chemical classes and we emphasized their synthetic routes, biological evaluation, selectivity, and structure-activity relationship.
Collapse
Affiliation(s)
- Lalmohan Maji
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, Karnataka, India
| | - Sindhuja Sengupta
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, Karnataka, India
| | | | - Ghanshyam Teli
- School of Pharmacy, Sangam University, Atoon, Bhilwara, 311001, Rajasthan, India
| | - Gourab Biswas
- Department of Pharmaceutical Technology, Brainware University, Kolkata, West Bengal, India
| | - Pronoy Kanti Das
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, Karnataka, India
| | | |
Collapse
|
4
|
Merugu SR, Selmer-Olsen S, Kaada CJ, Sundby E, Hoff BH. Synthetic Routes to 2-aryl-1 H-pyrrolo[2,3- b]pyridin-4-amines: Cross-Coupling and Challenges in SEM-Deprotection. Molecules 2024; 29:4743. [PMID: 39407670 PMCID: PMC11478076 DOI: 10.3390/molecules29194743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
7-Azaindoles are compounds of considerable medicinal interest. During development of the structure-activity relationship for inhibitors of the colony stimulated factor 1 receptor tyrosine kinase (CSF1R), a specific 2-aryl-1H-pyrrolo[2,3-b]pyridin-4-amine was needed. Two different synthetic strategies were evaluated, in which the order of the key C-C and C-N cross-coupling steps differed. The best route relied on a chemoselective Suzuki-Miyaura cross-coupling at C-2 on a 2-iodo-4-chloropyrrolopyridine intermediate, and subsequently a Buchwald-Hartwig amination with a secondary amine at C-4. Masking of hydroxyl and pyrroles proved essential to succeed with the latter transformation. The final trimethylsilylethoxymethyl (SEM) deprotection step was challenging, as release of formaldehyde gave rise to different side products, most interestingly a tricyclic eight-membered 7-azaindole. The target 2-aryl-1H-pyrrolo[2,3-b]pyridin-4-amine (compound 3c) proved to be 20-fold less potent than the reference inhibitor, confirming the importance of the N-3 in the pyrrolopyrimidine parent compound for efficient CSF1R inhibition.
Collapse
Affiliation(s)
- Srinivas Reddy Merugu
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), N-7491 Trondheim, Norway; (S.R.M.); (S.S.-O.); (C.J.K.)
| | - Sigrid Selmer-Olsen
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), N-7491 Trondheim, Norway; (S.R.M.); (S.S.-O.); (C.J.K.)
| | - Camilla Johansen Kaada
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), N-7491 Trondheim, Norway; (S.R.M.); (S.S.-O.); (C.J.K.)
| | - Eirik Sundby
- Department of Materials Science and Engineering, Norwegian University of Science and Technology (NTNU), N-7491 Trondheim, Norway;
| | - Bård Helge Hoff
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), N-7491 Trondheim, Norway; (S.R.M.); (S.S.-O.); (C.J.K.)
| |
Collapse
|
5
|
Khorasani F, Ranjbar-Karimi R, Mohammadiannejad K. Utilizing perhalopyridine-based alkynes as suitable precursors for the synthesis of novel poly(1,2,3-triazolyl)-substituted perhalopyridines. RSC Adv 2024; 14:30873-30885. [PMID: 39346527 PMCID: PMC11427873 DOI: 10.1039/d4ra05861e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
A novel series of poly(1,2,3-triazolyl)-substituted perhalopyridines 5a-f were successfully synthesized from the click reaction of the terminal alkynes (drived from the nucleophilic substitution reactions of PFP 1a and PCP 1b with excess amounts of propargyl alcohol) with aryl azides 4a-c under ultrasonic irradiation. Likewise, the sonication of reaction mixtures containing pyridyl cores 3, alkyl bromides 6a,b, and NaN3 under one-pot conditions afforded their respective aliphatic 1,2,3-triazoles 7a-d in yields ranging from 71% to 83%. We next developed an effective method for the regioselective preparation of 2,3,4,5-tetrachloro-6-(prop-2-yn-1-yloxy)pyridine 3c through SNAr reaction of PCP with propargyl alcohol without the utilization of any catalyst. It was then used to fabricate several ((1,2,3-triazol-4-yl)methoxy)-3,4,5,6-tetrachloropyridines 8a-c under the reaction conditions. Finally, the Pd(PPh3)4-catalyzed SMC reaction of tris-triazoles 5b,e with arylboronic acids 9a-c offered a practical method for the synthesis of biaryl-embedded poly(1,2,3-triazoles) 10a-f in good yields.
Collapse
Affiliation(s)
- Fereshteh Khorasani
- Department of Chemistry, Vali-e-Asr University of Rafsanjan Rafsanjan 77176 Islamic Republic of Iran +98-343-131-2429 +98 391 320 2162
| | - Reza Ranjbar-Karimi
- Department of Chemistry, Vali-e-Asr University of Rafsanjan Rafsanjan 77176 Islamic Republic of Iran +98-343-131-2429 +98 391 320 2162
| | - Kazem Mohammadiannejad
- NMR Laboratory, Faculty of Science, Vali-e-Asr University of Rafsanjan Rafsanjan 77176 Islamic Republic of Iran
| |
Collapse
|
6
|
Lv Y, Mi P, Babon JJ, Fan G, Qi J, Cao L, Lang J, Zhang J, Wang F, Kobe B. Small molecule drug discovery targeting the JAK-STAT pathway. Pharmacol Res 2024; 204:107217. [PMID: 38777110 DOI: 10.1016/j.phrs.2024.107217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway functions as a central hub for transmitting signals from more than 50 cytokines, playing a pivotal role in maintaining hematopoiesis, immune balance, and tissue homeostasis. Dysregulation of this pathway has been implicated in various diseases, including immunodeficiency, autoimmune conditions, hematological disorders, and certain cancers. Proteins within this pathway have emerged as effective therapeutic targets for managing these conditions, with various approaches developed to modulate key nodes in the signaling process, spanning from receptor engagement to transcription factor activation. Following the success of JAK inhibitors such as tofacitinib for RA treatment and ruxolitinib for managing primary myelofibrosis, the pharmaceutical industry has obtained approvals for over 10 small molecule drugs targeting the JAK-STAT pathway and many more are at various stages of clinical trials. In this review, we consolidate key strategies employed in drug discovery efforts targeting this pathway, with the aim of contributing to the collective understanding of small molecule interventions in the context of JAK-STAT signaling. We aspire that our endeavors will contribute to advancing the development of innovative and efficacious treatments for a range of diseases linked to this pathway dysregulation.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-Communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi 710054, China; Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai 201112, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100080, China
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi 710026, China
| | - Faming Wang
- Center for Molecular Biosciences and Non-Communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi 710054, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
7
|
Demir Ğ, Valjakka J, Turunen O, Aktaş F, Binay B. Cloning and expression heterologous alanine dehydrogenase genes: Investigation of reductive amination potential of L-alanine dehydrogenases for green synthesis of alanine derivatives. Heliyon 2024; 10:e26899. [PMID: 38463761 PMCID: PMC10923667 DOI: 10.1016/j.heliyon.2024.e26899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/09/2024] [Accepted: 02/21/2024] [Indexed: 03/12/2024] Open
Abstract
Unnatural amino acids (UAAs) offer significant promise in a wide range of applications, including drug discovery, the custom design of peptides and proteins, and their utility and use as markers for monitoring molecular interactions in biological research. The synthesis of UAAs presents a formidable challenge and can be classified into two primary categories: enzymatic and chemical synthesis. Notably, the enzymatic route, specifically asymmetric synthesis, emerges as a an attractive method for procuring enantiopure UAAs with high efficiency, owing to its streamlined and concise reaction mechanism. The current study investigated the reductive amination activity mechanisms of alanine dehydrogenase (L-AlaDH), sourced from a combination of newly and previously characterized microorganisms. Our principal aim was to evaluate the catalytic efficiency of these L-AlaDH enzymes concerning a range of specific ketoacids and pyruvate to ascertain their capability for facilitating the production of both natural and unnatural amino acids. After the characterization processes, mutation points for TtAlaDH were determined and as a result of the mutations, mutants that could use ketocaproate and ketovalerate more effectively than the wild type were obtained. Among the enzymes studied, MetAlaDH exhibited the highest specific activity against pyruvate, 173 U/mg, and a KM value of 1.3 mM. VlAlaDH displayed the most favourable catalytic efficiency with a rate constant of 170 s-1mM-1. On the other hand, AfAlaDH demonstrated the highest catalytic efficiency against α-ketobutyrate (34.0 s-1mM-1) and α-ketovalerate (2.7 s-1mM-1). Of the enzymes investigated in the study, TtAlaDH exhibited the highest effectiveness among bacterial enzymes in catalyzing ketocaproate with a measured catalytic efficiency of about 0.6 s-1mM-1 and a KM value of approximately 0.3 mM. These findings provide valuable insights into the substrate specificity and catalytic performance of L-AlaDHs, enhancing our understanding of their potential applications in various biocatalytic processes.
Collapse
Affiliation(s)
- Ğarip Demir
- Department of Molecular Biology and Genetics, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Jarkko Valjakka
- Faculty of Medicine and Health Technology, Tampere University, FI-33100, Tampere, Finland
| | - Ossi Turunen
- School of Forest Sciences, University of Eastern Finland, FI-80101, Joensuu, Finland
| | - Fatih Aktaş
- Faculty of Engineering, Düzce University, 81600, Düzce, Turkey
| | - Barış Binay
- Department of Bioengineering, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
- BAUZYME Biotechnology Co., Gebze Technical University Technopark, 41400, Gebze, Kocaeli, Turkey
| |
Collapse
|
8
|
Sk MF, Samanta S, Poddar S, Kar P. Deciphering the molecular choreography of Janus kinase 2 inhibition via Gaussian accelerated molecular dynamics simulations: a dynamic odyssey. J Comput Aided Mol Des 2024; 38:8. [PMID: 38324213 DOI: 10.1007/s10822-023-00548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/24/2023] [Indexed: 02/08/2024]
Abstract
The Janus kinases (JAK) are crucial targets in drug development for several diseases. However, accounting for the impact of possible structural rearrangements on the binding of different kinase inhibitors is complicated by the extensive conformational variability of their catalytic kinase domain (KD). The dynamic KD contains mainly four prominent mobile structural motifs: the phosphate-binding loop (P-loop), the αC-helix within the N-lobe, the Asp-Phe-Gly (DFG) motif, and the activation loop (A-loop) within the C-lobe. These distinct structural orientations imply a complex signal transmission path for regulating the A-loop's flexibility and conformational preference for optimal JAK function. Nevertheless, the precise dynamical features of the JAK induced by different types of inhibitors still remain elusive. We performed comparative, microsecond-long, Gaussian accelerated molecular dynamics simulations in triplicate of three phosphorylated JAK2 systems: the KD alone, type-I ATP-competitive inhibitor (CI) bound KD in the catalytically active DFG-in conformation, and the type-II inhibitor (AI) bound KD in the catalytically inactive DFG-out conformation. Our results indicate significant conformational variations observed in the A-loop and αC helix motions upon inhibitor binding. Our studies also reveal that the DFG-out inactive conformation is characterized by the closed A-loop rearrangement, open catalytic cleft of N and C-lobe, the outward movement of the αC helix, and open P-loop states. Moreover, the outward positioning of the αC helix impacts the hallmark salt bridge formation between Lys882 and Glu898 in an inactive conformation. Finally, we compared their ligand binding poses and free energy by the MM/PBSA approach. The free energy calculations suggested that the AI's binding affinity is higher than CI against JAK2 due to an increased favorable contribution from the total non-polar interactions and the involvement of the αC helix. Overall, our study provides the structural and energetic insights crucial for developing more promising type I/II JAK2 inhibitors for treating JAK-related diseases.
Collapse
Affiliation(s)
- Md Fulbabu Sk
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India
- Theoretical and Computational Biophysics Group, Beckman Institute for Advanced Science and Technology, NIH Resource for Macromolecular Modeling and Visualization, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sunanda Samanta
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India
| | - Sayan Poddar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India.
| |
Collapse
|
9
|
Liang T, Cen L, Wang J, Cheng M, Guo W, Wang W, Yu C, Zhang H, Wang Y, Hao Z, Jin J, Wu Y, Jiang T, Zhu Q, Xu Y. Discovery of novel dual Bruton's tyrosine kinase (BTK) and Janus kinase 3 (JAK3) inhibitors as a promising strategy for rheumatoid arthritis. Bioorg Med Chem 2023; 96:117354. [PMID: 37944414 DOI: 10.1016/j.bmc.2023.117354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 11/12/2023]
Abstract
Rheumatoid arthritis (RA) is a chronically systemic autoimmune disorder, which is related with various cellular signal pathways. Both BTK (Bruton's Tyrosine Kinase) and JAK3 (Janus Kinase 3) play important roles in the pathogenesis of rheumatoid arthritis. Herein, we reported the discovery of dual BTK/JAK3 inhibitors through bioisosterism and computer-aided drug design based on the structure of BTK inhibitor ibrutinib. We reported the discovery of dual BTK/JAK3 inhibitors which are based on the structure of BTK inhibitor ibrutinib via the method of bioisosterism and computer-aided drug design) Most of the target compounds exhibited moderate to strong inhibitory activities against BTK and JAK3. Among them, compound XL-12 stood out as the most promising candidate targeting BTK and JAK3 with potent inhibitory activities (IC50 = 2.0 nM and IC50 = 14.0 nM respectively). In the in vivo studies, compound XL-12 (40 mg/kg) exhibited more potent antiarthritic activity than ibrutinib (10 mg/kg) in adjuvant arthritis (AA) rat model. Furthermore, compound XL-12 (LD50 > 1600 mg/kg) exerted improved safety compared with ibrutinib (LD50 = 750 mg/kg). These results indicated that compound XL-12, the dual BTK/JAK3 inhibitor, might be a potent drug candidate for the treatment of RA.
Collapse
Affiliation(s)
- Tingting Liang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lifang Cen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Junjie Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ming Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Weibo Guo
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wenjie Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Chunqiu Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Haifeng Zhang
- Xi'an Xintong Pharmaceutical Research Co., Ltd. Xian, 710077, China
| | - Yuan Wang
- Xi'an Xintong Pharmaceutical Research Co., Ltd. Xian, 710077, China
| | - Zhongyan Hao
- Xi'an Xintong Pharmaceutical Research Co., Ltd. Xian, 710077, China
| | - Jiaming Jin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yaoyao Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Teng Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Qihua Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yungen Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
10
|
Zhang X, Xu X, Chen J, Wang G, Li Q, Li M, Lu J. Identification of HHT-9041P1: A novel potent and selective JAK1 inhibitor in a rat model of rheumatoid arthritis. Int Immunopharmacol 2023; 125:111086. [PMID: 37883818 DOI: 10.1016/j.intimp.2023.111086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/28/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic disease associated with long-term disability and premature mortality. If left untreated, it can seriously affect patients' quality of life. The JAK-STAT signal transduction process is known to affect the occurrence and development of RA, and small molecule JAK inhibitors, such as tofacitinib, have been identified as treatments for RA. However, tofacitinib is a non-selective JAK inhibitor that was found to be associated with dose-limiting tolerability and safety issues, such as anemia in phase 2 dose-ranging studies. Therefore, we developed a selective JAK1 inhibitor, HHT-9041P1, to overcome target-related adverse reactions. We used enzyme and cytokine potency assays in vitro as well as the collagen-induced arthritis (CIA) model in vivo to explore the efficacy and mechanism. In vitro, HHT-9041P1 was diluted (0.017 nM-1 mM) in DMSO) and mixed with JAK1, JAK2, JAK3 or TYK2 kinases for use in the respective assays for inhibitory activity and selectivity evaluation. Fresh human PBMCs were activated and incubated with 100 ng/mL cytokine IL-6 or 20 ng/mL GM-CSF for use in the investigation of the immune mechanism. In vivo, HHT-9041P1 (1 mg/kg, 3 mg/kg and 10 mg/kg) was administered by oral gavage twice daily to CIA model Lewis rats from Day 8 to Day 29 for paw swelling and arthritis score evaluation. At the end of the experiment, the rats were sacrificed before collection of the hind ankle joint, spleen and blood for analysis of inflammation, arthritis phenotypes, inflammatory cytokine expression and Th1 cell proportions. As expected, HHT-9041P1 showed 10-fold greater selectivity for JAK1 over JAK2, and 23-fold greater selectivity over JAK3 in cellular assays. The high selectivity of HHT-9041P1 was also validated by in vivo safety studies. HHT-9041P1 demonstrated significant efficacy in a rat model of collagen-induced arthritis (CIA) and was associated with reduced helper T Cell 1 (Th1) cell differentiation. HHT-9041P1 also exhibited excellent pharmacokinetics properties. Thus, HHT-9041P1 was identified as a candidate for clinical development with many options for the treatment of RA.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Pharmacoanalysis, School of Pharmacy, Fudan University, Shanghai, China; Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Xin Xu
- Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Jia Chen
- Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Guan Wang
- Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Qiang Li
- Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Min Li
- Member of Zhejiang Huahai Pharmaceutical, Shanghai, China.
| | - Jianzhong Lu
- Department of Pharmacoanalysis, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Sharma N, Chaudhary A, Sachdeva M. An insight into the structure-activity relationship studies of anticancer medicinal attributes of 7-azaindole derivatives: a review. Future Med Chem 2023; 15:2309-2323. [PMID: 38112047 DOI: 10.4155/fmc-2023-0216] [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: 07/24/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
In the current portfolio, there is a lot of interest in the 7-azaindole building block for drug discovery. The creation of synthetic, sophisticated methods for the modification of 7-azaindoles is a promising area of research. This review covers the structure-activity relationship of 7-azaindole analogs, which have been shown to be effective anticancer agents in the literature of the past two decades. Positions 1, 3 and 5 of the 7-azaindole ring are the most active sites. Disubstitution is used for the synthesis of a new analog of the 7-azaindole moiety. All positions are used to create novel molecules that are effective anticancer agents. The alkyl, aryl carboxamide group and heterocyclic ring are the most successful types of substitution.
Collapse
Affiliation(s)
- Neha Sharma
- Rajkumar Goel Institute of Technology (Pharmacy), NH-58, Ghaziabad, 201001, India
| | - Anurag Chaudhary
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, Meerut, 250005, India
| | - Monika Sachdeva
- Rajkumar Goel Institute of Technology (Pharmacy), NH-58, Ghaziabad, 201001, India
| |
Collapse
|
12
|
Palamini P, Allouche EMD, Waser J. Iron-Catalyzed Synthesis of α-Azido α-Amino Esters via the Alkylazidation of Alkenes. Org Lett 2023; 25:6791-6795. [PMID: 37684011 PMCID: PMC10521020 DOI: 10.1021/acs.orglett.3c02153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 09/10/2023]
Abstract
An iron-catalyzed alkylazidation of dehydroamino acids using peroxides as alkyl radical precursors is described. Non-natural azidated amino esters bearing an α-alkyl chain could be obtained in 18-94% yields using TMSN3 as an azide source. The obtained α-alkyl-α-azide α-amino esters could be further functionalized through cycloaddition or azide reduction with amide couplings to afford aminal-type peptides, α-triazolo amino acids, and tetrahydro-triazolopyridine, showing the great versatility of this now easily accessible class of amino acids.
Collapse
Affiliation(s)
- Pierre Palamini
- Laboratory of Catalysis and
Organic
Synthesis, Institut des Sciences et Ingénierie
Chimique, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Emmanuelle M. D. Allouche
- Laboratory of Catalysis and
Organic
Synthesis, Institut des Sciences et Ingénierie
Chimique, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jerome Waser
- Laboratory of Catalysis and
Organic
Synthesis, Institut des Sciences et Ingénierie
Chimique, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
13
|
Li T, Yang X, Zhu J, Liu Y, Jin X, Chen G, Ye L. Current application status and structure-activity relationship of selective and non-selective JAK inhibitors in diseases. Int Immunopharmacol 2023; 122:110660. [PMID: 37478665 DOI: 10.1016/j.intimp.2023.110660] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
JAK kinase includes four family members: JAK1, JAK2, JAK3, and TYK2. It forms the JAK-STAT pathway with signal transmitters and activators of subscription (STAT). This pathway is one of the main mechanisms by which many cytokine receptors transduce intracellular signals, it is associated with the occurrence of various immune, inflammatory, and tumor diseases. JAK inhibitors block the signal transduction of the JAK-STAT pathway by targeting JAK kinase. Based on whether they target multiple subtypes of JAK kinase, JAK inhibitors are categorized into pan-JAK inhibitors and selective JAK inhibitors. Compared with pan JAK inhibitors, selective JAK inhibitors are associated with a specific member, thus more targeted in therapy, with improved efficacy and reduced side effects. Currently, a number of JAK inhibitors have been approval for disease treatment. This review summarized the current application status of JAK inhibitors that have been marketed, advances of JAK inhibitors currently in phase Ш clinical trials, and the structure-activity relationship of them, with an intention to provide references for the development of novel JAK inhibitors.
Collapse
Affiliation(s)
- Tong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xianjing Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Juan Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ying Liu
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaobao Jin
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Gong Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lianbao Ye
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
14
|
Feng M, Fernandes AJ, Meyrelles R, Maulide N. Direct enantioselective α-amination of amides guided by DFT prediction of E/Z selectivity in a sulfonium intermediate. Chem 2023. [DOI: 10.1016/j.chempr.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
15
|
Zhang H, He F, Gao G, Lu S, Wei Q, Hu H, Wu Z, Fang M, Wang X. Approved Small-Molecule ATP-Competitive Kinases Drugs Containing Indole/Azaindole/Oxindole Scaffolds: R&D and Binding Patterns Profiling. Molecules 2023; 28:molecules28030943. [PMID: 36770611 PMCID: PMC9920796 DOI: 10.3390/molecules28030943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
Kinases are among the most important families of biomolecules and play an essential role in the regulation of cell proliferation, apoptosis, metabolism, and other critical physiological processes. The dysregulation and gene mutation of kinases are linked to the occurrence and development of various human diseases, especially cancer. As a result, a growing number of small-molecule drugs based on kinase targets are being successfully developed and approved for the treatment of many diseases. The indole/azaindole/oxindole moieties are important key pharmacophores of many bioactive compounds and are generally used as excellent scaffolds for drug discovery in medicinal chemistry. To date, 30 ATP-competitive kinase inhibitors bearing the indole/azaindole/oxindole scaffold have been approved for the treatment of diseases. Herein, we summarize their research and development (R&D) process and describe their binding models to the ATP-binding sites of the target kinases. Moreover, we discuss the significant role of the indole/azaindole/oxindole skeletons in the interaction of their parent drug and target kinases, providing new medicinal chemistry inspiration and ideas for the subsequent development and optimization of kinase inhibitors.
Collapse
Affiliation(s)
- Haofan Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Fengming He
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Guiping Gao
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Sheng Lu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Qiaochu Wei
- School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongyu Hu
- Xingzhi College, Zhejiang Normal University, Lanxi 321004, China
| | - Zhen Wu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Meijuan Fang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (M.F.); (X.W.)
| | - Xiumin Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (M.F.); (X.W.)
| |
Collapse
|
16
|
Marcos Santos L, da Silveira NJF. Current Fragment-to-lead Approaches Starting from the 7-azaindole: The Pharmacological Versatility of a Privileged Molecular Fragment. Curr Top Med Chem 2023; 23:2116-2130. [PMID: 37461366 DOI: 10.2174/1568026623666230718100541] [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: 03/26/2023] [Revised: 06/03/2023] [Accepted: 06/15/2023] [Indexed: 09/09/2023]
Abstract
Fragment-based drug discovery is one of the most powerful paradigms in the recent context of medicinal chemistry and is being widely practiced by academic and industrial researchers. Currently, azaindoles are among the most exploited molecular fragments in pharmaceutical innovation projects inspired by fragment-to-lead strategies. The 7-azaindole is the most prominent representative within this remarkable family of pyrrolopyridine fragments, as it is present in the chemical structure of several approved antitumor drugs and also of numerous therapeutic candidates. In this paper, a brief overview on existing proofs of concept in the literature will be presented, as well as some recent works that corroborate 7-azaindole as a privileged and pharmacologically versatile molecular fragment.
Collapse
Affiliation(s)
- Leandro Marcos Santos
- Laboratory of Molecular Modeling and Computer Simulation / MolMod-CS (D311-F), Institute of Chemistry, Federal University of Alfenas / UNIFAL-MG, Alfenas, Minas Gerais, 37130-001, Brazil
- Pharmaceutical Chemistry Research Laboratory / LQFar (D202A), Department of Food and Medicines, Faculty of Pharmaceutical Sciences, Federal University of Alfenas / UNIFAL-MG, Alfenas, Minas Gerais, 37130-001, Brazil
| | - Nelson José Freitas da Silveira
- Laboratory of Molecular Modeling and Computer Simulation / MolMod-CS (D311-F), Institute of Chemistry, Federal University of Alfenas / UNIFAL-MG, Alfenas, Minas Gerais, 37130-001, Brazil
| |
Collapse
|
17
|
Zhou S, Mao W, Su Y, Zheng X, Qian W, Shen M, Shan N, Li Y, Wang D, Wu S, Sun T, Mu L. Identification of TUL01101: A Novel Potent and Selective JAK1 Inhibitor for the Treatment of Rheumatoid Arthritis. J Med Chem 2022; 65:16716-16740. [PMID: 36512734 DOI: 10.1021/acs.jmedchem.2c01550] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Janus kinase 1 (JAK1) is a potential target for the treatment of rheumatoid arthritis (RA). In this study, the introduction of a spiro ring with a difluoro-substituted cyclopropionamide resulted in the identification of TUL01101 (compound 36) based on a triazolo[1,5-a]pyridine core of filgotinib. It showed excellent potency on JAK1 with an IC50 value of 3 nM and exhibited more than 12-fold selectivity for JAK2 and TYK2. Whole blood assay also demonstrated the high activity and selectivity (37-fold for JAK2). At the same time, TUL01101 also demonstrated excellent metabolic stability and pharmacokinetics (PK) profiles were assayed in three species (mouse, rat, and dog). Moreover, it has been validated for effective activity in the treatment of RA both in collagen-induced arthritis (CIA) and adjuvant-induced arthritis (AIA) models, with low dose and low toxicity. Now, TUL01101 has progressed into phase I clinical trials.
Collapse
Affiliation(s)
- Shuhao Zhou
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Weiwei Mao
- WuXi AppTec (Shanghai) Co., Ltd., 288 FuTe Zhong Road, Shanghai 200131, P. R. China
| | - Yuan Su
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Xuejian Zheng
- WuXi AppTec (Shanghai) Co., Ltd., 288 FuTe Zhong Road, Shanghai 200131, P. R. China
| | - Wenyuan Qian
- WuXi AppTec (Shanghai) Co., Ltd., 288 FuTe Zhong Road, Shanghai 200131, P. R. China
| | - Meiyue Shen
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Ningli Shan
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Yaoshuang Li
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Degang Wang
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Shouting Wu
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| | - Tiemin Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Ministry of Education, Shenyang 110016, P. R. China
| | - Liwei Mu
- Zhuhai United Laboratories Co., Ltd., NO.12 Jialian Road, Tanzhou Township, Zhongshan, Guangdong 528467, P. R. China
| |
Collapse
|
18
|
Okoye-Okafor UC, Javarappa KK, Tsallos D, Saad J, Yang D, Zhang C, Benard L, Thiruthuvanathan VJ, Cole S, Ruiz S, Tatiparthy M, Choudhary G, DeFronzo S, Bartholdy BA, Pallaud C, Ramos PM, Shastri A, Verma A, Heckman CA, Will B. Megakaryopoiesis impairment through acute innate immune signaling activation by azacitidine. J Exp Med 2022; 219:e20212228. [PMID: 36053753 PMCID: PMC9441716 DOI: 10.1084/jem.20212228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/02/2022] [Accepted: 07/22/2022] [Indexed: 11/04/2022] Open
Abstract
Thrombocytopenia, prevalent in the majority of patients with myeloid malignancies, such as myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML), is an independent adverse prognostic factor. Azacitidine (AZA), a mainstay therapeutic agent for stem cell transplant-ineligible patients with MDS/AML, often transiently induces or further aggravates disease-associated thrombocytopenia by an unknown mechanism. Here, we uncover the critical role of an acute type-I interferon (IFN-I) signaling activation in suppressing megakaryopoiesis in AZA-mediated thrombocytopenia. We demonstrate that megakaryocytic lineage-primed progenitors present IFN-I receptors and, upon AZA exposure, engage STAT1/SOCS1-dependent downstream signaling prematurely attenuating thrombopoietin receptor (TPO-R) signaling and constraining megakaryocytic progenitor cell growth and differentiation following TPO-R stimulation. Our findings directly implicate RNA demethylation and IFN-I signal activation as a root cause for AZA-mediated thrombocytopenia and suggest mitigation of TPO-R inhibitory innate immune signaling as a suitable therapeutic strategy to support platelet production, particularly during the early phases of AZA therapy.
Collapse
Affiliation(s)
- Ujunwa Cynthia Okoye-Okafor
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
| | - Komal K. Javarappa
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Dimitrios Tsallos
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Joseph Saad
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Daozheng Yang
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
| | - Chi Zhang
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
| | - Lumie Benard
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
| | - Victor J. Thiruthuvanathan
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
| | - Sally Cole
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
| | - Stephen Ruiz
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
| | - Madhuri Tatiparthy
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
| | - Gaurav Choudhary
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Medicine (Oncology), Bronx, NY
| | - Stefanie DeFronzo
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
| | - Boris A. Bartholdy
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
| | | | | | - Aditi Shastri
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Medicine (Oncology), Bronx, NY
| | - Amit Verma
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Medicine (Oncology), Bronx, NY
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Britta Will
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Cell Biology, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Cancer Stem Cell Pharmacodynamics Unit, Bronx, NY
- Albert Einstein College of Medicine/Montefiore Medical Center, Department of Medicine (Oncology), Bronx, NY
| |
Collapse
|
19
|
Dammeijer F, van Gulijk M, Klaase L, van Nimwegen M, Bouzid R, Hoogenboom R, Joosse ME, Hendriks RW, van Hall T, Aerts JG. Low-dose JAK3-inhibition improves anti-tumor T-cell immunity and immunotherapy efficacy. Mol Cancer Ther 2022; 21:1393-1405. [PMID: 35732501 DOI: 10.1158/1535-7163.mct-21-0943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/20/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022]
Abstract
Terminal T-cell exhaustion poses a significant barrier to effective anti-cancer immunotherapy efficacy with current drugs aimed at reversing exhaustion being limited. Recent investigations into the molecular drivers of T-cell exhaustion have led to the identification of chronic IL-2 receptor (IL-2R) - STAT5 pathway signaling in mediating T-cell exhaustion. We targeted the key downstream IL-2R-intermediate Janus kinase (JAK) 3 using a clinically relevant highly specific JAK3-inhibitor (JAK3i; PF-06651600) which potently inhibited STAT5-phosphorylation in vitro. Whereas pulsed high-dose JAK3i administration inhibited anti-tumor T-cell effector function, low-dose chronic JAK3i significantly improved T-cell responses and decreased tumor load in mouse models of solid cancer. Low-dose JAK3i combined with cellular and peptide vaccine strategies further decreased tumor load compared to both monotherapies alone. Collectively, these results identify JAK3 as a novel and promising target for combination immunotherapy.
Collapse
|
20
|
Mai NT, Lan NT, Vu TY, Tung NT, Phung HTT. A computationally affordable approach for accurate prediction of the binding affinity of JAK2 inhibitors. J Mol Model 2022; 28:163. [DOI: 10.1007/s00894-022-05149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/06/2022] [Indexed: 11/24/2022]
|
21
|
Patel J, Vazquez T, Chin F, Keyes E, Yan D, Diaz D, Grinnell M, Sharma M, Li Y, Feng R, Sprow G, Dan J, Werth VP. Multidimensional immune profiling of cutaneous lupus erythematosus in vivo stratified by patient responses to antimalarials. Arthritis Rheumatol 2022; 74:1687-1698. [PMID: 35583812 DOI: 10.1002/art.42235] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/14/2022] [Accepted: 05/12/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The pathogenesis of cutaneous lupus erythematous (CLE) is multifactorial and CLE is difficult to treat due to heterogeneity of inflammatory processes between patients. Antimalarials such as hydroxychloroquine (HCQ) and quinacrine (QC) have long been first-line systemic therapy; however, many patients do not respond and require systemic immunosuppressants with undesirable side effects. Given the complexity and unpredictable responses in CLE, we sought to identify the immunologic landscape of CLE patients stratified by subsequent treatment outcomes to identify potential biomarkers of inducible response. METHOD We performed imaging mass cytometry with 48 treatment-naïve skin biopsies of HCQ responders, QC responders, and non-responders (NR) to analyze multiple immune cell types and inflammatory markers in their native environment in CLE skin. Patients were stratified according to their subsequent response to antimalarials to identify baseline immunophenotypes which may predict response to therapy. RESULTS HCQ responders demonstrated increased CD4 T cells compared to QC. NR had decreased Tregs compared to QC and increased central memory T cells compared to HCQ. QC responders expressed increased phosphorylated (p) STING and IFNκ compared to HCQ. pSTING and IFNκ localized to conventional dendritic cells and positively correlated on a tissue and cellular level. Neighborhood analysis revealed decreased regulatory cell interactions in NR patients. Hierarchical clustering revealed NR groups separated based on pSTAT2/3/4/5, pIRF3, Granzyme B, pJAK2, IL4, IL17, and IFNγ. CONCLUSION These findings demonstrate differential immune compositions between CLE patients, guiding the future for precision-based medicine and treatment response.
Collapse
Affiliation(s)
- Jay Patel
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas Vazquez
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104
| | - Felix Chin
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Keyes
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daisy Yan
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - DeAnna Diaz
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison Grinnell
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meena Sharma
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yubin Li
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Feng
- Department of Biostatistics and Epidemiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Grant Sprow
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josh Dan
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria P Werth
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
22
|
Sk MF, Jonniya NA, Roy R, Kar P. Phosphorylation-Induced Conformational Dynamics and Inhibition of Janus Kinase 1 by Suppressors of Cytokine Signaling 1. J Phys Chem B 2022; 126:3224-3239. [PMID: 35443129 DOI: 10.1021/acs.jpcb.1c10733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The dysfunction of the JAK/STAT (Janus kinase/signal transducers and activators of transcription) pathway results in several pathophysiological conditions, including autoimmune disorders. The negative feedback regulators of the JAK/STAT signaling pathway, suppressors of cytokine signaling (SOCS), act as a natural inhibitor of JAK and inhibit aberrant activity. SOCS1 is the most potent member of the SOCS family, whose kinase inhibitory region targets the substrate-binding groove of JAK with high affinity and blocks the phosphorylation of JAK kinases. Overall, we performed an aggregate of 13 μs molecular dynamics simulations on the activation loop's three different phosphorylation (double and single) states. Results from our simulations show that the single Tyr1034 phosphorylation could stabilize the JAK1/SOCS1 complex as well as the flexible activation segment. The phosphate-binding loop (P-loop) shows conformational variability at dual and single phosphorylated states. Principal component analysis and protein structure network (PSN) analysis reveal that the different phosphorylation states and SOCS1 binding induce intermediate inactive conformations of JAK1, which could be a better target for future JAK1 selective drug design. PSN analysis suggests that the com-pY1034 system is stabilized due to higher values of network hubs than the other two complex systems. Moreover, the binding free energy calculations suggest that pTyr1034 states show a higher affinity toward SOCS1 than the dual and pTyr1035 states. We believe that the mechanistic understanding of JAK1/SOCS1 complexation will aid future studies related to peptide inhibitors based on SOCS1.
Collapse
Affiliation(s)
- Md Fulbabu Sk
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| | - Nisha Amarnath Jonniya
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| | - Rajarshi Roy
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| |
Collapse
|
23
|
Zhao MY, Zhang W, Rao GW. Targeting Janus Kinase (JAK) for Fighting Diseases: The Research of JAK Inhibitor Drugs. Curr Med Chem 2022; 29:5010-5040. [PMID: 35255783 DOI: 10.2174/1568026622666220307124142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/11/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
Janus Kinase (JAK), a nonreceptor protein tyrosine kinase, has emerged as an excellent target through research and development since its discovery in the 1990s. As novel small-molecule targeted drugs, JAK inhibitor drugs have been successfully used in the treatment of rheumatoid arthritis (RA), myofibrosis (MF) and ulcerative colitis (UC). With the gradual development of JAK targets in the market, JAK inhibitors have also received very considerable feedback in the treatment of autoimmune diseases such as atopic dermatitis (AD), Crohn's disease (CD) and graft-versus host disease (GVHD). This article reviews the research progress of JAK inhibitor drugs: introducing the existing JAK inhibitors on the market and some JAK inhibitors in clinical trials currently. In addition, the synthesis of various types of JAK inhibitors were summarized, and the effects of different drug structures on drug inhibition and selectivity.
Collapse
Affiliation(s)
- Min-Yan Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wen Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| |
Collapse
|
24
|
Taldaev A, Rudnev VR, Nikolsky KS, Kulikova LI, Kaysheva AL. Molecular Modeling Insights into Upadacitinib Selectivity upon Binding to JAK Protein Family. Pharmaceuticals (Basel) 2021; 15:ph15010030. [PMID: 35056087 PMCID: PMC8778839 DOI: 10.3390/ph15010030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 01/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic disease characterized by bone joint damage and incapacitation. The mechanism underlying RA pathogenesis is autoimmunity in the connective tissue. Cytokines play an important role in the human immune system for signal transduction and in the development of inflammatory responses. Janus kinases (JAK) participate in the JAK/STAT pathway, which mediates cytokine effects, in particular interleukin 6 and IFNγ. The discovery of small molecule inhibitors of the JAK protein family has led to a revolution in RA therapy. The novel JAK inhibitor upadacitinib (RinvoqTM) has a higher selectivity for JAK1 compared to JAK2 and JAK3 in vivo. Currently, details on the molecular recognition of JAK1 by upadacitinib are not available. We found that characteristics of hydrogen bond formation with the glycine loop and hinge in JAKs define the selectivity. Our molecular modeling study could provide insight into the drug action mechanism and pharmacophore model differences in JAK isoforms.
Collapse
Affiliation(s)
- Amir Taldaev
- Biobanking Group, V.N. Orekhovich Institute of Biomedical Chemistry, 109028 Moscow, Russia; (A.T.); (V.R.R.); (K.S.N.); (L.I.K.)
- Department of Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Vladimir R. Rudnev
- Biobanking Group, V.N. Orekhovich Institute of Biomedical Chemistry, 109028 Moscow, Russia; (A.T.); (V.R.R.); (K.S.N.); (L.I.K.)
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Kirill S. Nikolsky
- Biobanking Group, V.N. Orekhovich Institute of Biomedical Chemistry, 109028 Moscow, Russia; (A.T.); (V.R.R.); (K.S.N.); (L.I.K.)
| | - Liudmila I. Kulikova
- Biobanking Group, V.N. Orekhovich Institute of Biomedical Chemistry, 109028 Moscow, Russia; (A.T.); (V.R.R.); (K.S.N.); (L.I.K.)
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Institute of Mathematical Problems of Biology RAS—The Branch of Keldysh Institute of Applied Mathematics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Anna L. Kaysheva
- Biobanking Group, V.N. Orekhovich Institute of Biomedical Chemistry, 109028 Moscow, Russia; (A.T.); (V.R.R.); (K.S.N.); (L.I.K.)
- Correspondence:
| |
Collapse
|
25
|
Li SF, Liang X, Wu XK, Gao X, Zhang LW. Discovering the Mechanisms of Wikstroelide E as a Potential HIV-Latency-Reversing Agent by Transcriptome Profiling. JOURNAL OF NATURAL PRODUCTS 2021; 84:1022-1033. [PMID: 33721994 DOI: 10.1021/acs.jnatprod.0c01039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The discovery of efficient and specific HIV-latency-reversing agents is critical for HIV therapy. Here, we developed wikstroelide E, a daphnane diterpene from the buds of Wikstroemia chamaedaphne, as a potential HIV-latency-reversing agent that is 2500-fold more potent than the drug prostratin. Based on transcriptome analysis, the underlying mechanism was that wikstroelide E regulated the MAPK, PI3K-Akt, JAK-Stat, TNF, and NF-κB signaling pathways. We clearly demonstrated that wikstroelide E reversed latent HIV infection by activating PKC-NF-κB signals, serving as a proxy for verifying the transcriptome data. Strikingly, the Tat protein contributes to the robust activation of latent HIV in wikstroelide-E-treated cells, producing an unexpected latency-reversing effect against latent HIV. This study provides the basis for the potential development of wikstroelide E as an effective HIV-latency-reversing agent.
Collapse
Affiliation(s)
- Shi-Fei Li
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, People's Republic of China
| | - Xue Liang
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, People's Republic of China
| | - Xing-Kang Wu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, People's Republic of China
| | - Xiang Gao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, People's Republic of China
| | - Li-Wei Zhang
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan 030006, People's Republic of China
| |
Collapse
|
26
|
Al-Zaqri N, Pooventhiran T, Rao DJ, Alsalme A, Warad I, Thomas R. Structure, conformational dynamics, quantum mechanical studies and potential biological activity analysis of multiple sclerosis medicine ozanimod. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129685] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Wątły J, Miller A, Kozłowski H, Rowińska-Żyrek M. Peptidomimetics - An infinite reservoir of metal binding motifs in metabolically stable and biologically active molecules. J Inorg Biochem 2021; 217:111386. [PMID: 33610030 DOI: 10.1016/j.jinorgbio.2021.111386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/14/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022]
Abstract
The involvement of metal ions in interactions with therapeutic peptides is inevitable. They are one of the factors able to fine-tune the biological properties of antimicrobial peptides, a promising group of drugs with one large drawback - a problematic metabolic stability. Appropriately chosen, proteolytically stable peptidomimetics seem to be a reasonable solution of the problem, and the use of D-, β-, γ-amino acids, unnatural amino acids, azapeptides, peptoids, cyclopeptides and dehydropeptides is an infinite reservoir of metal binding motifs in metabolically stable, well-designed, biologically active molecules. Below, their specific structural features, metal-chelating abilities and antimicrobial potential are discussed.
Collapse
Affiliation(s)
- Joanna Wątły
- Faculty of Chemistry, University of Wroclaw, Joliot - Curie 14, Wroclaw 50-383, Poland.
| | - Adriana Miller
- Faculty of Chemistry, University of Wroclaw, Joliot - Curie 14, Wroclaw 50-383, Poland
| | - Henryk Kozłowski
- Faculty of Chemistry, University of Wroclaw, Joliot - Curie 14, Wroclaw 50-383, Poland; Department of Health Sciences, University of Opole, Katowicka 68, Opole 45-060, Poland
| | | |
Collapse
|
28
|
Moura RA, Fonseca JE. JAK Inhibitors and Modulation of B Cell Immune Responses in Rheumatoid Arthritis. Front Med (Lausanne) 2021; 7:607725. [PMID: 33614673 PMCID: PMC7892604 DOI: 10.3389/fmed.2020.607725] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic immune-mediated inflammatory disease that can lead to joint destruction, functional disability and substantial comorbidity due to the involvement of multiple organs and systems. B cells have several important roles in RA pathogenesis, namely through autoantibody production, antigen presentation, T cell activation, cytokine release and ectopic lymphoid neogenesis. The success of B cell depletion therapy with rituximab, a monoclonal antibody directed against CD20 expressed by B cells, has further supported B cell intervention in RA development. Despite the efficacy of synthetic and biologic disease modifying anti-rheumatic drugs (DMARDs) in the treatment of RA, few patients reach sustained remission and refractory disease is a concern that needs critical evaluation and close monitoring. Janus kinase (JAK) inhibitors or JAKi are a new class of oral medications recently approved for the treatment of RA. JAK inhibitors suppress the activity of one or more of the JAK family of tyrosine kinases, thus interfering with the JAK-Signal Transducer and Activator of Transcription (STAT) signaling pathway. To date, there are five JAK inhibitors (tofacitinib, baricitinib, upadacitinib, peficitinib and filgotinib) approved in the USA, Europe and/ or Japan for RA treatment. Evidence from the literature indicates that JAK inhibitors interfere with B cell functions. In this review, the main results obtained in clinical trials, pharmacokinetic, in vitro and in vivo studies concerning the effects of JAK inhibitors on B cell immune responses in RA are summarized.
Collapse
Affiliation(s)
- Rita A Moura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| |
Collapse
|
29
|
Xie Z, Yang X, Duan Y, Han J, Liao C. Small-Molecule Kinase Inhibitors for the Treatment of Nononcologic Diseases. J Med Chem 2021; 64:1283-1345. [PMID: 33481605 DOI: 10.1021/acs.jmedchem.0c01511] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Great successes have been achieved in developing small-molecule kinase inhibitors as anticancer therapeutic agents. However, kinase deregulation plays essential roles not only in cancer but also in almost all major disease areas. Accumulating evidence has revealed that kinases are promising drug targets for different diseases, including cancer, autoimmune diseases, inflammatory diseases, cardiovascular diseases, central nervous system disorders, viral infections, and malaria. Indeed, the first small-molecule kinase inhibitor for treatment of a nononcologic disease was approved in 2011 by the U.S. FDA. To date, 10 such inhibitors have been approved, and more are in clinical trials for applications other than cancer. This Perspective discusses a number of kinases and their small-molecule inhibitors for the treatment of diseases in nononcologic therapeutic fields. The opportunities and challenges in developing such inhibitors are also highlighted.
Collapse
Affiliation(s)
- Zhouling Xie
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xiaoxiao Yang
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yajun Duan
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Jihong Han
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Chenzhong Liao
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| |
Collapse
|
30
|
Alim K, Bruyère A, Lescoat A, Jouan E, Lecureur V, Le Vée M, Fardel O. Interactions of janus kinase inhibitors with drug transporters and consequences for pharmacokinetics and toxicity. Expert Opin Drug Metab Toxicol 2021; 17:259-271. [PMID: 33292029 DOI: 10.1080/17425255.2021.1862084] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Janus kinase inhibitors (JAKinibs) constitute an emerging and promising pharmacological class of anti-inflammatory or anti-cancer drugs, used notably for the treatment of rheumatoid arthritis and some myeloproliferative neoplasms.Areas covered: This review provides an overview of the interactions between marketed JAKinibs and major uptake and efflux drug transporters. Consequences regarding pharmacokinetics, drug-drug interactions and toxicity are summarized.Expert opinion: JAKinibs interact in vitro with transporters in various ways, as inhibitors or as substrates of transporters or as regulators of transporter expression. This may theoretically result in drug-drug interactions (DDIs), with JAKinibs acting as perpetrators or as victims, or in toxicity, via impairment of thiamine transport. Clinical significance in terms of DDIs for JAKinib-transporter interactions remains however poorly documented. In this context, the in vivo unbound concentration of JAKinibs is likely a key parameter to consider for evaluating the clinical relevance of JAKinibs-mediated transporter inhibition. Additionally, the interplay with drug metabolism as well as possible interactions with transporters of emerging importance and time-dependent inhibition have to be taken into account. The role drug transporters may play in controlling cellular JAKinib concentrations and efficacy in target cells is also an issue of interest.
Collapse
Affiliation(s)
- Karima Alim
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Alain Lescoat
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Elodie Jouan
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Valérie Lecureur
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Marc Le Vée
- Univ Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| | - Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut De Recherche En Santé, Environnement Et Travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
31
|
Blackwell JH, Kumar R, Gaunt MJ. Visible-Light-Mediated Carbonyl Alkylative Amination to All-Alkyl α-Tertiary Amino Acid Derivatives. J Am Chem Soc 2021; 143:1598-1609. [PMID: 33428383 DOI: 10.1021/jacs.0c12162] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The all-alkyl α-tertiary amino acid scaffold represents an important structural feature in many biologically and pharmaceutically relevant molecules. Syntheses of this class of molecule, however, often involve multiple steps and require activating auxiliary groups on the nitrogen atom or tailored building blocks. Here, we report a straightforward, single-step, and modular methodology for the synthesis of all-alkyl α-tertiary amino esters. This new strategy uses visible light and a silane reductant to bring about a carbonyl alkylative amination reaction that combines a wide range of primary amines, α-ketoesters, and alkyl iodides to form functionally diverse all-alkyl α-tertiary amino esters. Brønsted acid-mediated in situ condensation of primary amine and α-ketoester delivers the corresponding ketiminium species, which undergoes rapid 1,2-addition of an alkyl radical (generated from an alkyl iodide by the action of visible light and silane reductant) to form an aminium radical cation. Upon a polarity-matched and irreversible hydrogen atom transfer from electron rich silane, the electrophilic aminium radical cation is converted to an all-alkyl α-tertiary amino ester product. The benign nature of this process allows for broad scope in all three components and generates structurally and functionally diverse suite of α-tertiary amino esters that will likely have widespread use in academic and industrial settings.
Collapse
Affiliation(s)
- J Henry Blackwell
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Roopender Kumar
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Matthew J Gaunt
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
32
|
Raina G, Kannaboina P, Ahmed QN, Mondal K, Das P. Palladium‐Catalyzed Barluenga‐Valdes Type Cross‐Coupling Reaction: Alkenylation of 7‐Azaindole
s. ASIAN J ORG CHEM 2020. [DOI: 10.1002/ajoc.202000516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Gaurav Raina
- Medicinal Chemistry Division CSIR-Indian Institute of Integrative Medicine (IIIM) Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Uttar Pradesh 201002 India
| | - Prakash Kannaboina
- Medicinal Chemistry Division CSIR-Indian Institute of Integrative Medicine (IIIM) Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Uttar Pradesh 201002 India
| | - Qazi Naveed Ahmed
- Medicinal Chemistry Division CSIR-Indian Institute of Integrative Medicine (IIIM) Jammu 180001 India
- Academy of Scientific and Innovative Research (AcSIR) Uttar Pradesh 201002 India
| | - Krishanu Mondal
- Department of Chemistry Indian Institute of Technology (ISM) Dhanbad 826004 India
| | - Parthasarathi Das
- Department of Chemistry Indian Institute of Technology (ISM) Dhanbad 826004 India
| |
Collapse
|
33
|
Tosstorff A, Cole JC, Taylor R, Harris SF, Kuhn B. Identification of Noncompetitive Protein–Ligand Interactions for Structural Optimization. J Chem Inf Model 2020; 60:6595-6611. [DOI: 10.1021/acs.jcim.0c00858] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Andreas Tosstorff
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
- Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, U.K
| | - Jason C. Cole
- Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, U.K
| | - Robin Taylor
- Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, U.K
| | - Seth F. Harris
- Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Bernd Kuhn
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| |
Collapse
|
34
|
El Jammal T, Sève P, Gerfaud-Valentin M, Jamilloux Y. State of the art: approved and emerging JAK inhibitors for rheumatoid arthritis. Expert Opin Pharmacother 2020; 22:205-218. [PMID: 32967471 DOI: 10.1080/14656566.2020.1822325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is the most common autoimmune inflammatory arthritis in adults. In the past decade, many treatments have emerged to expand the therapeutic armamentarium of rheumatologists. Among emerging treatments, Janus Kinase inhibitors (JAKi) are promising in treating RA and several other inflammatory conditions, such as psoriatic arthritis (PsA). The JAK/STAT signaling pathway is located downstream certain cytokines receptors that are known to be involved in RA pathogenesis. So far, three JAKi are approved for the treatment of RA, while other JAKi, are under investigation. AREAS COVERED Herein, the authors review those JAKi approved and emerging for the treatment of RA and provide their expert perspectives on the subject area. EXPERT OPINION JAKi represent an interesting alternative to other DMARDs when MTX has failed. Long-term extension studies are still ongoing, but one can assume that most of the major safety concerns have already come out. Switching from one JAKi to another DMARD has been little studied, but in such cases, preferring a treatment which does not interfere with the JAK/STAT pathway seems to be a reasonable choice.
Collapse
Affiliation(s)
- Thomas El Jammal
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| | - Pascal Sève
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| | | | - Yvan Jamilloux
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| |
Collapse
|
35
|
Robinson MF, Damjanov N, Stamenkovic B, Radunovic G, Kivitz A, Cox L, Manukyan Z, Banfield C, Saunders M, Chandra D, Vincent MS, Mancuso J, Peeva E, Beebe JS. Efficacy and Safety of PF-06651600 (Ritlecitinib), a Novel JAK3/TEC Inhibitor, in Patients With Moderate-to-Severe Rheumatoid Arthritis and an Inadequate Response to Methotrexate. Arthritis Rheumatol 2020; 72:1621-1631. [PMID: 32419304 PMCID: PMC7589242 DOI: 10.1002/art.41316] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 05/13/2020] [Indexed: 12/20/2022]
Abstract
Objective To evaluate the efficacy and safety of PF‐06651600 (ritlecitinib), an irreversible inhibitor of JAK3 and the tyrosine kinase expressed in hepatocellular carcinoma (TEC) kinase family, in comparison with placebo in patients with rheumatoid arthritis (RA). Methods An 8‐week, phase II, double‐blind, parallel‐group study was conducted. Seventy patients who were seropositive for anti–citrullinated protein antibodies and/or rheumatoid factor were randomized 3:2 to receive oral PF‐06651600 (200 mg once daily) or placebo for 8 weeks. Eligible patients had an inadequate response to methotrexate, and the study design allowed up to 50% of patients to have previously received 1 tumor necrosis factor inhibitor that was inadequately effective and/or not tolerated. The primary end point was change from baseline in the Simplified Disease Activity Index (SDAI) score at week 8, assessed by Bayesian analysis using an informative prior distribution for placebo response. Results Mean change from baseline in the SDAI score at week 8 was greater in the PF‐06651600 group (−26.1 [95% credible interval −29.7, −22.4]) than in the placebo group (−16.8 [95% credible interval −20.9, −12.7]; P < 0.001). Most adverse events (AEs) were mild in severity, and no treatment‐related serious AEs, severe AEs, or deaths were reported. The most common classes of AE were infections and infestations as well as skin and subcutaneous tissue disorders; there was 1 mild case of herpes simplex in the PF‐06651600 group that was considered to be treatment related, which resolved within 3 days without study treatment discontinuation or antiviral therapy. Conclusion Treatment with the oral JAK3/TEC inhibitor PF‐06651600 (200 mg once daily) was associated with significant improvements in RA disease activity and was generally well‐tolerated in this small 8‐week study.
Collapse
Affiliation(s)
| | | | - Bojana Stamenkovic
- Institute for Treatment and Rehabilitation Niska Banja and Nis University School of Medicine, Nis, Serbia
| | | | - Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, Pennsylvania
| | - Lori Cox
- Pfizer, Inc., New York, New York
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nimje RY, Vytla D, Kuppusamy P, Velayuthaperumal R, Jarugu LB, Reddy CA, Chikkananjaiah NK, Rampulla RA, Cavallaro CL, Li J, Mathur A, Gupta A, Roy A. Synthesis of Differentially Protected Azatryptophan Analogs via Pd 2(dba) 3/XPhos Catalyzed Negishi Coupling of N-Ts Azaindole Halides with Zinc Derivative from Fmoc-Protected tert-Butyl ( R)-2-Amino-3-iodopropanoate. J Org Chem 2020; 85:11519-11530. [PMID: 32786620 DOI: 10.1021/acs.joc.0c00973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Unnatural amino acids play an important role in peptide based drug discovery. Herein, we report a class of differentially protected azatryptophan derivatives synthesized from N-tosyl-3-haloazaindoles 1 and Fmoc-protected tert-butyl iodoalanine 2 via a Negishi coupling. Through ligand screening, Pd2(dba)3/XPhos was found to be a superior catalyst for the coupling of 1 with the zinc derivative of 2 to give tert-butyl (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-(1-tosyl-1H-pyrrolo[2,3-b]pyridin-3-yl)propanoate derivatives 3 in 69-91% isolated yields. In addition, we have demonstrated that the protecting groups, namely, Ts, Fmoc, and tBu, can be easily removed selectively.
Collapse
Affiliation(s)
- Roshan Y Nimje
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Devaiah Vytla
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Prakasam Kuppusamy
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Rajeswari Velayuthaperumal
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Lokesh Babu Jarugu
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - China Anki Reddy
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Nanjundaswamy Kanikahalli Chikkananjaiah
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Richard A Rampulla
- Small Molecule Drug Discovery, Bristol-Myers Squibb Research and Development, P.O. Box 5400, Princeton, New Jersey 08543-4000, United States
| | - Cullen L Cavallaro
- Small Molecule Drug Discovery, Bristol-Myers Squibb Research and Development, P.O. Box 5400, Princeton, New Jersey 08543-4000, United States
| | - Jianqing Li
- Small Molecule Drug Discovery, Bristol-Myers Squibb Research and Development, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Arvind Mathur
- Small Molecule Drug Discovery, Bristol-Myers Squibb Research and Development, P.O. Box 5400, Princeton, New Jersey 08543-4000, United States
| | - Anuradha Gupta
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| | - Amrita Roy
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb R&D Center, Syngene International Ltd., Biocon Park, Plot No. 2 and 3, Bommasandra-Jigani Road, Bangalore-560 100, India
| |
Collapse
|
37
|
Angelini J, Talotta R, Roncato R, Fornasier G, Barbiero G, Dal Cin L, Brancati S, Scaglione F. JAK-Inhibitors for the Treatment of Rheumatoid Arthritis: A Focus on the Present and an Outlook on the Future. Biomolecules 2020; 10:E1002. [PMID: 32635659 PMCID: PMC7408575 DOI: 10.3390/biom10071002] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
Janus kinase inhibitors (JAKi) belong to a new class of oral targeted disease-modifying drugs which have recently revolutionized the therapeutic panorama of rheumatoid arthritis (RA) and other immune-mediated diseases, placing alongside or even replacing conventional and biological drugs. JAKi are characterized by a novel mechanism of action, consisting of the intracellular interruption of the JAK-STAT pathway crucially involved in the immune response. The aim of this narrative review is to globally report the most relevant pharmacological features and clinical outcomes of the developed and incoming JAKi for RA, based on the available preclinical and clinical evidence. A total of 219 papers, including narrative and systematic reviews, randomized controlled trials (RCTs), observational studies, case reports, guidelines, and drug factsheets, were selected. The efficacy and safety profile of both the first generation JAKi (baricitinib and tofacitinib) and the second generation JAKi (upadacitinib, filgotinib, peficitinib, decernotinib and itacitinib) were compared and discussed. Results from RCTs and real-life data are encouraging and outline a rapid onset of the pharmacologic effects, which are maintained during the time. Their efficacy and safety profile are comparable or superior to those of biologic agents and JAKi proved to be efficacious when given as monotherapy. Finally, the manufacturing of JAKi is relatively easier and cheaper than that of biologics, thus increasing the number of compounds being formulated and tested for clinical use.
Collapse
Affiliation(s)
- Jacopo Angelini
- Postgraduate School of Clinical Pharmacology and Toxicology, University of Milan, 20133 Milan, Italy; (J.A.); (G.F.); (G.B.); (L.D.C.); (S.B.)
| | - Rossella Talotta
- Department of Clinical and Experimental Medicine, Rheumatology Unit, AOU “Gaetano Martino”, University of Messina, 98100 Messina, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pordenone, 33081 Aviano, Italy;
| | - Giulia Fornasier
- Postgraduate School of Clinical Pharmacology and Toxicology, University of Milan, 20133 Milan, Italy; (J.A.); (G.F.); (G.B.); (L.D.C.); (S.B.)
- Pharmacy Unit, IRCCS-Burlo Garofolo di Trieste, 34137 Trieste, Italy
| | - Giorgia Barbiero
- Postgraduate School of Clinical Pharmacology and Toxicology, University of Milan, 20133 Milan, Italy; (J.A.); (G.F.); (G.B.); (L.D.C.); (S.B.)
| | - Lisa Dal Cin
- Postgraduate School of Clinical Pharmacology and Toxicology, University of Milan, 20133 Milan, Italy; (J.A.); (G.F.); (G.B.); (L.D.C.); (S.B.)
| | - Serena Brancati
- Postgraduate School of Clinical Pharmacology and Toxicology, University of Milan, 20133 Milan, Italy; (J.A.); (G.F.); (G.B.); (L.D.C.); (S.B.)
| | - Francesco Scaglione
- Head of Clinical Pharmacology and Toxicology Unit, Grande Ospedale Metropolitano Niguarda, Department of Oncology and Onco-Hematology, Director of Postgraduate School of Clinical Pharmacology and Toxicology, University of Milan, 20162 Milan, Italy;
| |
Collapse
|
38
|
Singh S, Singh S. JAK-STAT inhibitors: Immersing therapeutic approach for management of rheumatoid arthritis. Int Immunopharmacol 2020; 86:106731. [PMID: 32590315 DOI: 10.1016/j.intimp.2020.106731] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis is a world leading cause of musculoskeletal disease. With the introduction of biological agents as treatment alternatives the clinical possibilities have grown exponentially. Currently most common Disease-modifying anti-rheumatic drugs (DMARDs) treatment option involves intravenous or subcutaneous injection, and some patients struggle to respond to DMARDs or lose their primary reaction. An oral drug formulation with lowered costs of manufacturing and flexibility for healthcare workers to preferably perform treatment will result in decreased healthcare expenditures and increased medication compliance. The JAK-STAT inhibitors, a new class of small molecules drugs, fulfills these criteria and has recently shown efficacy in rheumatoid arthritis. Here we give a summary of how JAK-STAT inhibitors function and a detailed review of current clinical trials. Convincing clinical results suggest that therapeutic inhibition of the JAK proteins can effectively modulate a complex cytokine-driven inflammation.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India.
| | - Shantanu Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| |
Collapse
|
39
|
Zhu J, Yu Q, Cai Y, Chen Y, Liu H, Liang W, Jin J. Theoretical Exploring Selective-Binding Mechanisms of JAK3 by 3D-QSAR, Molecular Dynamics Simulation and Free Energy Calculation. Front Mol Biosci 2020; 7:83. [PMID: 32528970 PMCID: PMC7266956 DOI: 10.3389/fmolb.2020.00083] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 04/16/2020] [Indexed: 12/31/2022] Open
Abstract
Janus kinase 3 (JAK3) plays a critical role in the JAK/STAT signaling pathway and has become an attractive selective target for the treatment of immune-mediated disorders. Therefore, great efforts have been made for the development of JAK3 inhibitors, but developing selective JAK3 inhibitors remains a great challenge because of the high sequence homology with other kinases. In order to reveal the selective-binding mechanisms of JAK3 and to find the key structural features that refer to specific JAK3 inhibition, a systematic computational method, including 3D-QSAR, molecular dynamics simulation, and free energy calculations, was carried out on a series of JAK3 isoform-selective inhibitors. Necessary pharmacodynamic structures and key residues involved in efficient JAK3-inhibition were then highlighted. Finally, 10 novel JAK3 inhibitors were designed, the satisfactory predicted binding affinity to JAK3 of these analogous demonstrated that this study may facilitate the rational design of novel and selective JAK3 inhibitors.
Collapse
Affiliation(s)
- Jingyu Zhu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Qianqian Yu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Yanfei Cai
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Yun Chen
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Hui Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | | | - Jian Jin
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| |
Collapse
|
40
|
Affiliation(s)
- Ju Wang
- College of Pharmaceutical Science and Institute of Drug Development & Chemical Biology; Zhejiang University of Technology; 310014 Hangzhou P. R. China
| | - Chen-Fu Liu
- School of Pharmaceutical Sciences; Gannan Medical University; 341000 Ganzhou P. R. China
| | - Quan Zheng
- College of Pharmaceutical Science and Institute of Drug Development & Chemical Biology; Zhejiang University of Technology; 310014 Hangzhou P. R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science and Institute of Drug Development & Chemical Biology; Zhejiang University of Technology; 310014 Hangzhou P. R. China
| |
Collapse
|
41
|
Hosseini A, Gharibi T, Marofi F, Javadian M, Babaloo Z, Baradaran B. Janus kinase inhibitors: A therapeutic strategy for cancer and autoimmune diseases. J Cell Physiol 2020; 235:5903-5924. [DOI: 10.1002/jcp.29593] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Arezoo Hosseini
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
- Student Research CommitteeTabriz University of Medical SciencesTabriz Iran
- Aging Research InstituteTabriz University of Medical SciencesTabriz Iran
| | - Tohid Gharibi
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
- Student Research CommitteeTabriz University of Medical SciencesTabriz Iran
- Aging Research InstituteTabriz University of Medical SciencesTabriz Iran
| | - Faroogh Marofi
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Mahsa Javadian
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Zohreh Babaloo
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| |
Collapse
|
42
|
Mei H, Han J, Klika KD, Izawa K, Sato T, Meanwell NA, Soloshonok VA. Applications of fluorine-containing amino acids for drug design. Eur J Med Chem 2019; 186:111826. [PMID: 31740056 DOI: 10.1016/j.ejmech.2019.111826] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/21/2019] [Accepted: 10/26/2019] [Indexed: 01/26/2023]
Abstract
Fluorine-containing amino acids are becoming increasingly prominent in new drugs due to two general trends in the modern pharmaceutical industry. Firstly, the growing acceptance of peptides and modified peptides as drugs; and secondly, fluorine editing has become a prevalent protocol in drug-candidate optimization. Accordingly, fluorine-containing amino acids represent one of the more promising and rapidly developing areas of research in organic, bio-organic and medicinal chemistry. The goal of this Review article is to highlight the current state-of-the-art in this area by profiling 42 selected compounds that combine fluorine and amino acid structural elements. The compounds under discussion represent pharmaceutical drugs currently on the market, or in clinical trials as well as examples of drug-candidates that although withdrawn from development had a significant impact on the progress of medicinal chemistry and/or provided a deeper understanding of the nature and mechanism of biological action. For each compound, we present features of biological activity, a brief history of the design principles and the development of the synthetic approach, focusing on the source of tailor-made amino acid structures and fluorination methods. General aspects of the medicinal chemistry of fluorine-containing amino acids and synthetic methodology are briefly discussed.
Collapse
Affiliation(s)
- Haibo Mei
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Jianlin Han
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Karel D Klika
- Molecular Structure Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Kunisuke Izawa
- Hamari Chemicals Ltd., 1-4-29 Kunijima, Higashi-Yodogawa-ku, Osaka, 533-0024, Japan.
| | - Tatsunori Sato
- Hamari Chemicals Ltd., 1-4-29 Kunijima, Higashi-Yodogawa-ku, Osaka, 533-0024, Japan
| | - Nicholas A Meanwell
- Department of Discovery Chemistry, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ, 08543-4000, United States.
| | - Vadim A Soloshonok
- Department of Organic Chemistry I, University of the Basque Country UPV/EHU, Paseo Manuel Lardizábal 3, 20018, San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, María Díaz de Haro 3, Plaza Bizkaia, 48013, Bilbao, Spain.
| |
Collapse
|
43
|
Musumeci F, Greco C, Giacchello I, Fallacara AL, Ibrahim MM, Grossi G, Brullo C, Schenone S. An Update on JAK Inhibitors. Curr Med Chem 2019; 26:1806-1832. [PMID: 29589523 DOI: 10.2174/0929867325666180327093502] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 02/03/2023]
Abstract
Janus kinases (JAKs) are a family of non-receptor tyrosine kinases, composed by four members, JAK1, JAK2, JAK3 and TYK2. JAKs are involved in different inflammatory and autoimmune diseases, as well as in malignancies, through the activation of the JAK/STAT signalling pathway. Furthermore, the V617F mutation in JAK2 was identified in patients affected by myeloproliferative neoplasms. This knowledge prompted researchers from academia and pharmaceutical companies to investigate this field in order to discover small molecule JAK inhibitors. These efforts recently afforded to the market approval of four JAK inhibitors. Despite the fact that all these drugs are pyrrolo[2,3-d]pyrimidine derivatives, many compounds endowed with different heterocyclic scaffolds have been reported in the literature as selective or multi-JAK inhibitors, and a number of them is currently being evaluated in clinical trials. In this review we will report many representative compounds that have been published in articles or patents in the last five years (period 2013-2017). The inhibitors will be classified on the basis of their chemical structure, focusing, when possible, on their structure activity relationships, selectivity and biological activity. For every class of derivatives, compounds disclosed before 2013 that have entered clinical trials will also be briefly reported, to underline the importance of a particular chemical scaffold in the search for new inhibitors.
Collapse
Affiliation(s)
- Francesca Musumeci
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Chiara Greco
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Ilaria Giacchello
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Anna Lucia Fallacara
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Munjed M Ibrahim
- College of Pharmacy, Department of Pharmaceutical Chemistry, Umm Al-Qura University, 21955-Makkah Al- Mukarramah, Saudi Arabia
| | - Giancarlo Grossi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Chiara Brullo
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy
| |
Collapse
|
44
|
Wei Y, Zhang Z, She N, Chen X, Zhao Y, Zhang J. Atomistic insight into the inhibition mechanisms of suppressors of cytokine signaling on Janus kinase. Phys Chem Chem Phys 2019; 21:12905-12915. [PMID: 31157353 DOI: 10.1039/c9cp02257k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Suppressors of cytokine signaling (SOCS) act as negative feedback regulators of the Janus kinase/signal transducer (JAK-STAT) signaling pathway by inhibiting the activity of JAK kinase. The kinase inhibitory region (KIR) of SOCS1 targets the substrate binding groove of JAK with high specificity, as demonstrated by significantly higher IC50 following the mutation of any of residue. To gain a greater understanding of the mechanisms of the inhibition of SOCS1 for JAK1, the binding mode, binding free energy decomposition, and desorption mechanism of JAK-SOCS1 complexes as well as a number of mutant systems were identified by extensive molecular dynamics (MD) simulations and the constant pulling velocity (PCV) method. Electrostatic interactions were identified for their contribution to protein-protein binding, which drove interactions between JAK1 and SOCS1. The polar residues Arg56, Arg59, and Asp105 of SOCS1 and Asp1042 and Asp1040 of JAK1 were key components in the binding, and electrostatic interactions of the side chains were prominent. The binding free energies of the six mutant proteins were lower when compared with those of the control proteins, and the side chain interactions were weakened. The residue Asp1040 played a crucial role in KIR close to the binding groove of JAK1. Moreover, salt bridges contributed significantly to JAK1 and SOCS1 binding and cleavage processes. The study presented herein provides a comprehensive understanding of the thermodynamic and dynamic processes of SOCS1 and JAK1 binding that will contribute meaningfully to the design of future studies related to peptide inhibitors based on SOCS1.
Collapse
Affiliation(s)
- Yaru Wei
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloy, College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
45
|
Essential Kinases and Transcriptional Regulators and Their Roles in Autoimmunity. Biomolecules 2019; 9:biom9040145. [PMID: 30974919 PMCID: PMC6523499 DOI: 10.3390/biom9040145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022] Open
Abstract
Kinases and transcriptional regulators are fundamental components of cell signaling that are expressed on many types of immune cells which are involved in secretion of cytokines, cell proliferation, differentiation, and apoptosis. Both play important roles in biological responses in health as well as in illnesses such as the autoimmune diseases which comprise at least 80 disorders. These diseases are caused by complex genetic and environmental interactions that lead to a breakage of immunologic tolerance and a disruption of the balance between self-reactive cells and regulatory cells. Kinases or transcriptional regulatory factors often have an abnormal expression in the autoimmune cells that participate in the pathogenesis of autoimmune disease. These abnormally expressed kinases or transcriptional regulators can over-activate the function of self-reactive cells to produce inflammatory cytokines or down-regulate the activity of regulatory cells, thus causing autoimmune diseases. In this review we introduce five kinds of kinase and transcriptional regulator related to autoimmune diseases, namely, members of the Janus kinase (JAK) family (JAK3 and/or tyrosine kinase 2 (TYK2)), fork head box protein 3 (Foxp3), the retinoic acid-related orphan receptor gamma t (RORγt), and T-box expressed in T cells (T-bet) factors. We also provide a mechanistic insight into how these kinases and transcriptional regulators affect the function of the immune cells related to autoimmune diseases, as well as a description of a current drug design targeting these kinases and transcriptional regulators. Understanding their exact role helps offer new therapies for control of the inflammatory responses that could lead to clinical improvement of the autoimmune diseases.
Collapse
|
46
|
Narancic T, Almahboub SA, O’Connor KE. Unnatural amino acids: production and biotechnological potential. World J Microbiol Biotechnol 2019; 35:67. [DOI: 10.1007/s11274-019-2642-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 03/28/2019] [Indexed: 01/01/2023]
|
47
|
Are peptides a solution for the treatment of hyperactivated JAK3 pathways? Inflammopharmacology 2019; 27:433-452. [PMID: 30929155 DOI: 10.1007/s10787-019-00589-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/18/2019] [Indexed: 01/10/2023]
Abstract
While the inactivation mutations that eliminate JAK3 function lead to the immunological disorders such as severe combined immunodeficiency, activation mutations, causing constitutive JAK3 signaling, are known to trigger various types of cancer or are responsible for autoimmune diseases, such as rheumatoid arthritis, psoriasis, or inflammatory bowel diseases. Treatment of hyperactivated JAK3 is still an obstacle, due to different sensibility of mutation types to conventional drugs and unwanted side effects, because these drugs are not absolutely specific for JAK3, thus inhibiting other members of the JAK family, too. Lack of information, in which way sole inhibition of JAK3 is necessary for elimination of the disease, calls for the development of isoform-specific JAK3 inhibitors. Beside this strategy, up to date peptides are a rising alternative as chemo- or immunotherapeutics, but still sparsely represented in drug development and clinical trials. Beyond a possible direct inhibition function, crossing the cancer cell membrane and interfering in disease-causing pathways or triggering apoptosis, peptides could be used in future as adjunct remedies to potentialize traditional therapy and preserve non-affected cells. To discuss such feasible topics, this review deals with the knowledge about the structure-function of JAK3 and the actual state-of-the-art of isoform-specific inhibitor development, as well as the function of currently approved drugs or those currently being tested in clinical trials. Furthermore, several strategies for the application of peptide-based drugs for cancer therapy and the physicochemical and structural relations to peptide efficacy are discussed, and an overview of peptide sequences, which were qualified for clinical trials, is given.
Collapse
|
48
|
T Virtanen A, Haikarainen T, Raivola J, Silvennoinen O. Selective JAKinibs: Prospects in Inflammatory and Autoimmune Diseases. BioDrugs 2019; 33:15-32. [PMID: 30701418 PMCID: PMC6373396 DOI: 10.1007/s40259-019-00333-w] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines, many of which signal through the JAK-STAT (Janus kinase-Signal Transducers and Activators of Transcription) pathway, play a central role in the pathogenesis of inflammatory and autoimmune diseases. Currently three JAK inhibitors have been approved for clinical use in USA and/or Europe: tofacitinib for rheumatoid arthritis, psoriatic arthritis and ulcerative colitis, baricitinib for rheumatoid arthritis, and ruxolitinib for myeloproliferative neoplasms. The clinical JAK inhibitors target multiple JAKs at high potency and current research has focused on more selective JAK inhibitors, almost a dozen of which currently are being evaluated in clinical trials. In this narrative review, we summarize the status of the pan-JAK and selective JAK inhibitors approved or in clinical trials, and discuss the rationale for selective targeting of JAKs in inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Anniina T Virtanen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
| | - Teemu Haikarainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Juuli Raivola
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Olli Silvennoinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland.
- Fimlab Laboratories, 33520, Tampere, Finland.
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, (Viikinkaari 5), 00014, Helsinki, Finland.
| |
Collapse
|
49
|
Štarha P, Trávníček Z, Crlíková H, Vančo J, Kašpárková J, Dvořák Z. Half-Sandwich Ir(III) Complex of N1-Pyridyl-7-azaindole Exceeds Cytotoxicity of Cisplatin at Various Human Cancer Cells and 3D Multicellular Tumor Spheroids. Organometallics 2018. [DOI: 10.1021/acs.organomet.8b00415] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pavel Štarha
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Zdeněk Trávníček
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Hana Crlíková
- Department of Biophysics, Faculty of Science, Palacký University, 17. listopadu 12, 771 46 Olomouc, Czech Republic
| | - Ján Vančo
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Jana Kašpárková
- Department of Biophysics, Faculty of Science, Palacký University, 17. listopadu 12, 771 46 Olomouc, Czech Republic
| | - Zdeněk Dvořák
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| |
Collapse
|
50
|
Bryan MC, Rajapaksa NS. Kinase Inhibitors for the Treatment of Immunological Disorders: Recent Advances. J Med Chem 2018; 61:9030-9058. [DOI: 10.1021/acs.jmedchem.8b00667] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Marian C. Bryan
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Naomi S. Rajapaksa
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|