Targeting the JAK/STAT Pathway: A Combined Ligand- and Target-Based Approach
- Maria Galvez-Llompart*Maria Galvez-Llompart*Email: [email protected]Department of Physical Chemistry, University of Valencia, Av. Vicente Estelles s/n, 46100 Burjassot (Valencia), SpainInstituto de Tecnología Química (UPV-CSIC) Universidad Politécnica de Valencia Av. Naranjos s/n, 46022 Valencia, SpainMore by Maria Galvez-Llompart
- ,
- Riccardo OcelloRiccardo OcelloDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Riccardo Ocello
- ,
- Laura RulloLaura RulloDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Laura Rullo
- ,
- Serena StamatakosSerena StamatakosDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Serena Stamatakos
- ,
- Irene AlessandriniIrene AlessandriniDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Irene Alessandrini
- ,
- Riccardo ZanniRiccardo ZanniDepartment of Physical Chemistry, University of Valencia, Av. Vicente Estelles s/n, 46100 Burjassot (Valencia), SpainMore by Riccardo Zanni
- ,
- Iñaki TuñónIñaki TuñónDepartment of Physical Chemistry, University of Valencia, Av. Vicente Estelles s/n, 46100 Burjassot (Valencia), SpainMore by Iñaki Tuñón
- ,
- Andrea CavalliAndrea CavalliDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyItalian Institute of Technology (IIT), Via Morego 30, 16163 Genoa, ItalyMore by Andrea Cavalli
- ,
- Sanzio CandelettiSanzio CandelettiDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Sanzio Candeletti
- ,
- Matteo MasettiMatteo MasettiDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Matteo Masetti
- ,
- Patrizia RomualdiPatrizia RomualdiDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Patrizia Romualdi
- , and
- Maurizio RecanatiniMaurizio RecanatiniDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Belmeloro 6, 40126 Bologna, ItalyMore by Maurizio Recanatini
Abstract

Janus kinases (JAKs) are a family of proinflammatory enzymes able to mediate the immune responses and the inflammatory cascade by modulating multiple cytokine expressions as well as various growth factors. In the present study, the inhibition of the JAK–signal transducer and activator of transcription (STAT) signaling pathway is explored as a potential strategy for treating autoimmune and inflammatory disorders. A computationally driven approach aimed at identifying novel JAK inhibitors based on molecular topology, docking, and molecular dynamics simulations was carried out. For the best candidates selected, the inhibitory activity against JAK2 was evaluated in vitro. Two hit compounds with a novel chemical scaffold, 4 (IC50 = 0.81 μM) and 7 (IC50 = 0.64 μM), showed promising results when compared with the reference drug Tofacitinib (IC50 = 0.031 μM).
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
1. Introduction
Figure 1

Figure 1. Screening and profiling workflow employed in this work.
2. Results and Discussion
2.1. Development and Validation of the QSAR Classification Models
Figure 2

Figure 2. Chemical graph and adjacency matrix of the isopentane.
2.1.1. General Model for Predicting the JAK Inhibitory Activity

Figure 3

Figure 3. Example of SRW05 and CIC2 values for DFpan for two JAK inhibitors (Tofacitinib and NSC33994) and two decoys (102-05-6 and 119-24-4).
Figure 4

Figure 4. Examples of GATS6m values for two JAK inhibitors (Hexabromocyclohexane and AG490) and two decoys (12334-10-1 and 114145-29-8).
compounds classified as active | compounds classified as inactive | correct classification (%) | |
---|---|---|---|
Training set | |||
active group | 33 | 9 | 79 |
inactive group | 14 | 45 | 76 |
total | 47 | 54 | 77 |
Test set | |||
active group | 23 | 3 | 88 |
inactive group | 9 | 24 | 72 |
total | 31 | 28 | 80 |
Figure 5

Figure 5. PDD for the general model. Blue bars represent the distribution of JAK inhibitors, and orange bars represent decoys.
2.1.2. Specific QSAR Models for Predicting Selective Inhibitory Activity toward Each JAK Subtype
percent of correct classification | |||
---|---|---|---|
DF1 | DF2 | DF3 | |
active group compounds | 73 | 90 | 82 |
inactive group compounds | 89 | 90 | 94 |
total | 83 | 90 | 89 |
internal validationa | 77 | 83 | 87 |
Average value.
Figure 6

Figure 6. PDD for the subtype-specific models: DF1, DF2, and DF3 in panels (a), (b), and (c), respectively. Black bars represent the distribution of JAK inhibitors, and white bars represent decoys.
2.2. Virtual Screening of a Commercial Database
JAK model | JAK1 model | JAK2 model | JAK3 model | ||||
---|---|---|---|---|---|---|---|
compound | DFgen | DF1 | class. | DF2 | class. | DF3 | class. |
AA-516/30011028 | 1.370 | 4.408 | JAK1 | 5.096 | JAK2 | –7.554 | |
AB-323/13887443 | 0.996 | 4.940 | JAK1 | –2.528 | –3.924 | ||
AC-907/34131030 | 1.729 | 1.524 | JAK1a | 3.415 | JAK2 | –5.154 | |
AE-848/34779061 | 1.682 | 1.830 | JAK1a | 0.956 | –5.407 | ||
AF-399/13277002 | 2.313 | 1.661 | JAK1a | –9.474 | 34.181 | N.C. | |
AF-399/13426006 | 1.013 | 8.821 | N.C. | 6.795 | N.C. | –6.897 | |
AF-399/15031149 | 1.546 | 0.951 | JAK1a | –0.561 | –5.680 | ||
AF-399/15032375 | 0.981 | 5.213 | JAK1 | –0.973 | –7.423 | ||
AF-399/33696009 | 1.977 | 5.731 | JAK1 | 2.553 | JAK2 | –3.293 | |
AF-399/37297037 | 1.454 | –2.344 | –2.130 | –0.599 | |||
AF-399/41668884 | 1.270 | 6.604 | JAK1 | 2.890 | JAK2 | –7.146 | |
AF-399/41945530 | 1.875 | 0.214 | JAK1a | 2.056 | JAK2 | –2.432 | |
AF-399/42056988 | 0.978 | 2.044 | JAK1a | 5.767 | JAK2 | 4.314 | JAK3 |
AF-399/42100326 | 1.649 | 4.054 | JAK1 | 2.115 | JAK2 | –4.967 | |
AF-399/42762404 | 1.901 | 5.191 | JAK1 | 3.680 | JAK2 | –8.082 | |
AG-205/11444099 | 0.921 | 2.650 | JAK1 | 6.925 | N.C. | 7.388 | JAK3 |
AG-205/11674118 | 0.993 | 1.653 | JAK1a | 2.922 | JAK2 | 7.297 | JAK3 |
AG-205/12010072 | 0.954 | 4.327 | JAK1 | 1.075 | JAK2 | –3.811 | |
AG-205/14250132 | 0.508 | 1.765 | JAK1a | –3.150 | –4.043 | ||
AG-205/14673025 | 1.414 | 1.456 | JAK1a | 2.119 | JAK2 | –0.373 | |
AG-401/02041003 | 1.810 | –3.699 | –5.515 | 3.404 | JAK3 | ||
AG-670/13619018 | -0.978 | 3.426 | JAK1 | 3.325 | JAK2 | –0.314 | |
AG-690/36926024 | 2.249 | –2.545 | –0.516 | JAK2 | 4.618 | JAK3 | |
AH-357/03329001 | 0.530 | 0.563 | JAK1a | 0.175 | JAK2 | –5.608 | |
AK-778/43206447 | 2.346 | 0.171 | JAK1a | 5.559 | JAK2 | –0.863 | |
AK-968/15359231 | 2.103 | 3.603 | JAK1 | 3.093 | JAK2 | –4.128 | |
AM-807/37225018 | 0.646 | 11.083 | N.C. | 1.879 | JAK2 | –16.247 | |
AN-329/11658808 | 2.503 | 2.659 | JAK1 | 4.496 | JAK2 | 0.445 | JAK3 |
AN-329/41717385 | –1.097 | 7.420 | JAK1 | 3.673 | JAK2 | –5.092 | |
AN-584/40652663 | 2.879 | 7.470 | JAK1 | 7.042 | N.C. | 5.262 | JAK3 |
AN-584/43492329 | 1.641 | 3.542 | JAK1 | 3.279 | JAK2 | 6.518 | JAK3 |
AN-988/41531688 | 0.663 | –1.129 | JAK1a | 4.160 | JAK2 | –3.345 | |
AO-365/43473564 | 1.559 | 4.322 | JAK1 | –0.516 | JAK2 | 5.543 | JAK3 |
AO-476/41610187 | 1.340 | 0.351 | JAK1a | –9.178 | 3.540 | JAK3 | |
AO-476/43250148 | 1.253 | 5.967 | JAK1 | –2.347 | 0.294 | JAK3 | |
AO-476/43250150 | 2.215 | 6.816 | JAK1 | –1.103 | –2.306 | ||
AO-476/43250160 | 1.120 | 6.705 | JAK1 | –1.464 | -3.291 | ||
AO-476/43417077 | 1.690 | –1.031 | JAK1a | 3.833 | JAK2 | 1.214 | JAK3 |
AP-064/42049177 | 0.803 | –6.558 | –0.021 | JAK2 | 1.564 | JAK3 | |
AP-501/43286814 | 1.120 | –0.082 | JAK1a | 1.093 | JAK2 | –4.604 | |
AQ-405/42300191 | 0.548 | 0.990 | JAK1a | 5.571 | JAK2 | –2.172 | |
AQ-432/43399984 | 0.528 | 5.710 | JAK1 | 3.595 | JAK2 | –5.423 | |
AQ-432/43400108 | 1.617 | 2.557 | JAK1 | 4.888 | JAK2 | 0.677 | JAK3 |
AQ-432/43400219 | 0.862 | 0.113 | JAK1a | 3.845 | JAK2 | 2.936 | JAK3 |
AQ-432/43400304 | 0.772 | 5.631 | JAK1 | 3.870 | JAK2 | –5.792 | |
AQ-432/43400319 | 1.602 | 2.562 | JAK1 | 4.251 | JAK2 | 3.808 | JAK3 |
AT-417/43503979 | 0.903 | 3.253 | JAK1 | 8.632 | N.C. | 20.232 | N.C. |
Overlapping zone with other JAK inhibitor subtypes, nonabsolutely sure being correctly classified by this model.
N.C., not classifiable by this model, out of range of the applicability domain.
Bold: in vitro tested.
2.3. Molecular Docking
Figure 7

Figure 7. Representation of the binding sites of the JAK1, JAK2, and JAK3 structures in complex with the native ligands that were selected for the docking calculations (PDB-ID: 4IVD, 5CF6, and 6GLA, respectively). (20−22) The conserved residues are represented as sticks with carbon atoms colored in white, while specific amino acids are differently colored (orange, yellow, and green for JAK1, JAK2, and JAK3, respectively). Hydrogen bonds are explicitly reported as black dots.
compound | docking score JAK1 (PDB-ID: 4IVD) | docking score JAK2 (PDB-ID: 5CF6) | docking score JAK3 (PDB-ID: 6GLA) |
---|---|---|---|
AT9283 | –6.14a | –6.95 | |
AZ-960 | –8.23 | ||
AZD1480 | –8.63 | ||
Baricitinib | –8.97 | –9.49 | |
BMS-911543 | –9.68 | ||
CEP33779 | –9.27 | ||
Cerdulatinib | –6.28 | –8.38 | –8.28 |
Decernotinib | –9.62 | ||
Filgotinib | –7.29 | –9.62 | –8.85 |
FLLL32 | –5.35 | ||
Gandotinib | –8.94 | ||
Go6976 | –8.28 | ||
Hexabromocyclohexane | –2.93 | ||
Itacitinib | –8.88 | ||
JANEX-1 | –8.43 | ||
Momelotinib | –8.84 | –7.63 | |
NVP-BSK805 | –10.05 | ||
Oclacitinib | –9.28 | ||
Pacritinib | –6.84 | ||
PF-04965842 | –8.61 | –7.96 | |
PF 06551600 malonate | –8.97 | ||
Ruxolitinib | –8.75 | –9.27 | |
Solcitinib | –8.43 | ||
TG101209 | –6.01 | ||
Tofacitinib | –9.31 | –8.50 | –8.08 |
WHI-P154 | –7.19 | ||
WHI-P97 | –6.60 | ||
WP1066 | –6.15 | ||
XL019 | –9.30 | ||
ZM39923 hydrochloride | –6.92 | –6.30 |
kcal/mol.
Bold: reference drug.
Figure 8

Figure 8. Chemical representation and codification for the eight selected compounds as potential JAK inhibitors.
docking score (kcal/mol) | |||
---|---|---|---|
compound | JAK1 (PDB-ID: 4IVD) | JAK2 (PDB-ID: 5CF6) | JAK3 (PDB-ID: 6GLA) |
Tofacitinib | –9.31a | –8.50 | –8.08 |
1 | –7.06 | –7.61 | –8.00 |
2 | –6.82 | –9.46 | –6.96 |
3 | –8.24 | –7.31 | –7.89 |
4 | –6.81 | –8.17 | –7.26 |
5 | –9.05 | –7.11 | –6.90 |
6 | –8.14 | –9.03 | –8.32 |
7 | –5.34 | –7.34 | –6.88 |
8 | –8.13 | –6.28 | –5.73 |
kcal/mol.
Underlined: docking score from the reference drug.
Bold: top docking score for each JAK subtype under analysis.
Figure 9

Figure 9. Ball and stick representation of the three top-ranked compounds for each JAK subtype: 5 bound to JAK1, 2 bound to JAK2, and 6 bound to JAK3 (panels b, d, and f, respectively). For comparison, the binding mode of the reference compound Tofacitinib is also reported (panels a, c, and e for JAK1, JAK2, and JAK3, respectively).
Figure 10

Figure 10. RMSD values of Cα atoms of the three JAK subtypes (a: JAK1; b: JAK2; and c: JAK3) in the complexes with the best-ranking molecule for each subtype and Tofacitinib along the 50 ns of MD simulations (blue lines). In purple, the RMSD computed using the heavy atoms of the ligands (after least-squares-fit superimposition to the Cα atoms of the protein) is also shown.
Figure 11

Figure 11. Protein–ligand contact interaction over the MD trajectory. Hydrogen bonds are shown in blue, water-mediated hydrogen bonds in red, hydrophobic interactions in gray, salt bridges in yellow, π–π interactions in green, and cation−π interactions in orange.
docking score (kcal/mol) | ||||||
---|---|---|---|---|---|---|
compound | predicted subtype | JAK1 (PDB-ID: 4IVD) | JAK2 (PDB-ID: 5CF6) | JAK3 (PDB-ID: 6GLA) | ||
1 | JAK1 | JAK2 | –7.06b | –7.61 | –8.00 | |
2 | JAK1 | JAK2 | –6.82 | –9.46 | –6.96 | |
3 | JAK1 | JAK2 | –8.24 | –7.31 | –7.89 | |
4 | JAK1 | JAK2 | –6.81 | –8.17 | –7.26 | |
5 | JAK1a | JAK2 | –9.05 | –7.11 | –6.90 | |
6 | JAK1a | JAK2 | JAK3 | –8.14 | –9.03 | –8.32 |
7 | JAK1a | JAK3 | –5.34 | –7.34 | –6.88 | |
8 | JAK1a | JAK2 | –8.13 | –6.28 | –5.73 |
Overlapping zone with other JAK inhibitor subtypes, nonabsolutely sure being correctly classified by this model.
kcal/mol.
2.4. Chemical Diversity of Potential JAK Inhibitors
2.5. In Vitro Tests
2.5.1. MTT Cell Viability
Figure 12

Figure 12. Cell viability of SH-SY5Y cells exposed to different concentrations of tested drugs for 5 and 24 h, evaluated by the MTT assay. Data are expressed as a percentage of OD values of treated cells compared to vehicle-treated ones and reported as the mean ± standard error of the mean (SEM) (*p < 0.05; **p < 0.01 vs the respective control, one-way analysis of variance (ANOVA) test followed by Dunnett’s test).
2.5.2. JAK2 Activity Assay
compounds | IC50 (nM) |
---|---|
1 | >10 000 |
3 | >10 000 |
4 | 807 (643–1007) |
5 | >10 000 |
6 | >10 000 |
7 | 637 (367–1076) |
8 | >10 000 |
Tofacitinib | 31.4 (16–61) |
95% confidence limits are shown in brackets.
Figure 13

Figure 13. Chemical structure and IC50 (μM) for commercially available JAK2 inhibitors. (26−28)
Figure 14

Figure 14. Amino acid interaction between in vitro-tested compounds 4 (a) and 7 (b) and JAK2 (PDB: 5CF6).
3. Conclusions
4. Experimental: In Silico Modeling
4.1. QSAR Model
4.1.1. Strategy to Identify Novel JAK Inhibitors
4.1.2. Data-Set Compilation
Data set for the general model. Active compounds were JAK inhibitors retrieved from the literature (26,30,31) and from the commercial database, and inactive compounds were taken from the Sigma-Aldrich catalog (therefore they act as putative inactive compounds, or decoys). A chemical similarity analysis was performed between active and inactive groups, by selecting compounds with similar MW, and the number of carbon, nitrogen, oxygen, and halogens atoms.
Data set for the JAK1 subtype-specific model. Active compounds were commercial JAK1 inhibitors, and inactive compounds were commercial JAK2 and JAK3 inhibitors.
Data set for the JAK2 subtype-specific model. Active compounds were commercial JAK2 inhibitors, and inactive compounds were commercial JAK1 and JAK3 inhibitors.
Data set for the JAK3 subtype-specific model. Active compounds were commercial JAK3 inhibitors, and inactive compounds were commercial JAK1 and JAK2 inhibitors.
4.1.3. Calculation of Descriptors
4.1.4. Statistical Analysis to Build the Model

4.1.5. Pharmacological Distribution Diagram
4.1.6. Validation of the Models
4.2. Molecular Docking Simulations
4.2.1. Molecular Docking
4.2.2. Cross-Docking Analysis
4.3. Molecular Dynamics Simulations
4.4. Chemical Diversity of Potential JAK Inhibitors
5. Experimental: In Vitro Assays
5.1. Cell Culture
5.2. Cell Viability Assay
5.3. JAK2 Assay
5.4. Statistical Analysis
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.0c01468.
Descriptor values, classification of compounds and probability of activity for all data sets of models 1–4 (Tables S1–S5); leave-some-out validation test for DF2-4 (Tables S6–S8); docking score values from potential JAK inhibitors selected by molecular topology (Table S9); cross-docking analysis for JAK1, JAK2, and JAK3 subtypes (Figure S1); similarity-cluster analysis dendrogram (Figure S2) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 40 other publications.
- 1Harpur, A. G.; Andres, A. C.; Ziemiecki, A.; Aston, R. R.; Wilks, A. F. JAK2, a third member of the JAK family of protein tyrosine kinases. Oncogene 1992, 7, 1347– 1353Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XmtVOrtLc%253D&md5=db385133406115942157615e9c56b47fJAK2, a third member of the JAK family of protein tyrosine kinasesHarpur, Ailsa G.; Andres, Anne Catherine; Ziemiecki, Andrew; Aston, Raja R.; Wilks, Andrew F.Oncogene (1992), 7 (7), 1347-53CODEN: ONCNES; ISSN:0950-9232.CDNA clones encoding a third, widely expressed, member of the JAK family of protein tyrosine kinases (PTKs) were isolated. The anticipated amino acid sequence of JAK2 predicts the presence of two kinase-related domains, a feature characteristic of this family of PTKs. The structural similarity of JAK2 to the other members of this family extends towards their N-termini, beyond the two kinase-related domains, and reveals five further domains of substantial amino acid similarity. The C-terminal portion of one of these domains, the JH4 domain, bears an intriguing, albeit tenuous, similarity to the core element of the SH2 domain, whereas the remaining JAK homol. domains do not appear to be a feature of other known proteins.
- 2Kawamura, M.; McVicar, D. W.; Johnston, J. A.; Blake, T. B.; Chen, Y.; Lal, B. K.; Lloyd, A. R.; Kelvin, D. J.; Staples, J. E.; Ortaldo, J. R. Molecular cloning of L-JAK, a Janus family protein-tyrosine kinase expressed in natural killer cells and activated leukocytes. Proc. Natl. Acad. Sci. U.S.A. 1994, 91, 6374– 6378, DOI: 10.1073/pnas.91.14.6374Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXlvF2jurY%253D&md5=c898c32fb7ff39b6987cca1717d9f229Molecular cloning of L-JAK, a Janus family protein-tyrosine kinase expressed in natural killer cells and activated leukocytesKawamura, Masaru; McVicar, Daniel W.; Johnston, James A.; Blake, Trevor B.; Chen, Yi-Qing; Lal, Brajesh K.; Lloyd, Andrew R.; Kelvin, David J.; Staples, J. Erin; et al.Proceedings of the National Academy of Sciences of the United States of America (1994), 91 (14), 6374-8CODEN: PNASA6; ISSN:0027-8424.Protein-tyrosine kinases (PTKs) are crit. enzymes for receptor-mediated signaling in lymphocytes. Because natural killer (NK) cells are large granular lymphocytes with specialized effector function, PTKs preferentially expressed in these cells were investigated. One such PTK was identified and molecularly cloned. The predicted amino acid sequence shows that this kinase lacks SH2 or SH3 domains typical of src family kinases but has tandem nonidentical catalytic domains, indicating that it is a member of the Janus family of PTKs. Immunopptn. using antiserum generated against a peptide corresponding to the deduced amino acid sequence of this gene revealed a kinase with a mol. wt. of ∼125,000. The pattern of expression of this kinase contrasted sharply with that of other Janus kinases, which are ubiquitously expressed. The kinase described in the present study was more limited in its expression; expression was found in NK cells and an NK-like cell line but not in resting T cells or in other tissues. In contrast, stimulated and transformed T cells expressed the gene, suggesting a role in lymphoid activation. Because of its homol. and tissue expression, this PTK gene was tentatively termed L-JAK for leukocyte Janus kinase.
- 3Wilks, A. F. The JAK kinases: not just another kinase drug discovery target. In Seminars in cell & developmental biology; Elsevier, 2008; Vol. 19, pp 319– 328.Google ScholarThere is no corresponding record for this reference.
- 4Williams, N. K.; Bamert, R. S.; Patel, O.; Wang, C.; Walden, P. M.; Wilks, A. F.; Fantino, E.; Rossjohn, J.; Lucet, I. S. Dissecting specificity in the Janus kinases: the structures of JAK-specific inhibitors complexed to the JAK1 and JAK2 protein tyrosine kinase domains. J. Mol. Biol. 2009, 387, 219– 232, DOI: 10.1016/j.jmb.2009.01.041Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXivVegtbo%253D&md5=79957755d04323057b5acf7e9d2c56caDissecting Specificity in the Janus Kinases: The Structures of JAK-Specific Inhibitors Complexed to the JAK1 and JAK2 Protein Tyrosine Kinase DomainsWilliams, Neal K.; Bamert, Rebecca S.; Patel, Onisha; Wang, Christina; Walden, Patricia M.; Wilks, Andrew F.; Fantino, Emmanuelle; Rossjohn, Jamie; Lucet, Isabelle S.Journal of Molecular Biology (2009), 387 (1), 219-232CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)The Janus kinases (JAKs) are a pivotal family of protein tyrosine kinases (PTKs) that play prominent roles in numerous cytokine signaling pathways, with aberrant JAK activity assocd. with a variety of hematopoietic malignancies, cardiovascular diseases and immune-related disorders. Whereas the structures of the JAK2 and JAK3 PTK domains have been detd., the structure of the JAK1 PTK domain is unknown. Here, we report the high-resoln. crystal structures of the "active form" of the JAK1 PTK domain in complex with two JAK inhibitors, a tetracyclic pyridone 2-t-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-one (CMP6) and (3R,4R)-3-[4-methyl-3-[N-methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]piperidin-1-yl]-3-oxopropionitrile (CP-690,550), and compare them with the corresponding JAK2 PTK inhibitor complexes. Both inhibitors bound in a similar manner to JAK1, namely buried deep within a constricted ATP-binding site, thereby providing a basis for the potent inhibition of JAK1. As expected, the mode of inhibitor binding in JAK1 was very similar to that obsd. in JAK2, highlighting the challenges in developing JAK-specific inhibitors that target the ATP-binding site. Nevertheless, differences surrounding the JAK1 and JAK2 ATP-binding sites were apparent, thereby providing a platform for the rational design of JAK2- and JAK1-specific inhibitors.
- 5Saharinen, P.; Takaluoma, K.; Silvennoinen, O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain. Mol. Cell. Biol. 2000, 20, 3387– 3395, DOI: 10.1128/MCB.20.10.3387-3395.2000Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXivFyhtrw%253D&md5=17ac4581ee9c36e03e95f0f88405339cRegulation of the Jak2 tyrosine kinase by its pseudokinase domainSaharinen, Pipsa; Takaluoma, Kati; Silvennoinen, OlliMolecular and Cellular Biology (2000), 20 (10), 3387-3395CODEN: MCEBD4; ISSN:0270-7306. (American Society for Microbiology)Activation of Jak tyrosine kinases through hematopoietic cytokine receptors occurs as a consequence of ligand-induced aggregation of receptor-assocd. Jaks and their subsequent autophosphorylation. Jak kinases consist of a C-terminal tyrosine kinase domain, a pseudokinase domain of unknown function, and Jak homol. (JH) domains 3 to 7, implicated in receptor-Jak interaction. We analyzed the functional roles of the different protein domains in activation of Jak2. Deletion anal. of Jak2 showed that the pseudokinase domain but not JH domains 3 to 7 neg. regulated the catalytic activity of Jak2 as well Jak2-mediated activation of Stat5. Phosphorylation of Stat5 by wild-type Jak2 was dependent on the SH2 domain of Stat5; however, this requirement was lost upon deletion of the pseudokinase domain of Jak2. Investigation of the mechanisms of the pseudokinase domain-mediated inhibition of Jak2 suggested that this regulation did not involve protein tyrosine phosphatases. Instead, anal. of interactions between the tyrosine kinase domain and Jak2 suggested that the pseudokinase domain interacted with the kinase domain. Furthermore, co-expression of the pseudokinase domain inhibited the activity of the single tyrosine kinase domain. Finally, deletion of the pseudokinase domain of Jak2 deregulated signal transduction through the gamma interferon receptor by significantly increasing ligand-independent activation of Stat transcription factors. These results indicate that the pseudokinase domain neg. regulates the activity of Jak2, probably through an interaction with the kinase domain, and this regulation is required to keep Jak2 inactive in the absence of ligand stimulation. Furthermore, the pseudokinase domain may have a role in regulation of Jak2-substrate interactions.
- 6Damsky, W.; Peterson, D.; Ramseier, J.; Al-Bawardy, B.; Chun, H.; Proctor, D.; Strand, V.; Flavell, R. A.; King, B. The emerging role of Janus kinase inhibitors in the treatment of autoimmune and inflammatory diseases. J. Allergy Clin. Immunol. 2021, 147, 814– 826, DOI: 10.1016/j.jaci.2020.10.022Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1agurY%253D&md5=74561a7a05af19978f183215bdf4f5f6The emerging role of Janus kinase inhibitors in the treatment of autoimmune and inflammatory diseasesDamsky, William; Peterson, Danielle; Ramseier, Julie; Al-Bawardy, Badr; Chun, Hyung; Proctor, Deborah; Strand, Vibeke; Flavell, Richard A.; King, BrettJournal of Allergy and Clinical Immunology (2021), 147 (3), 814-826CODEN: JACIBY; ISSN:0091-6749. (Elsevier Inc.)A review. Autoimmune and inflammatory diseases are common and diverse, and they can affect nearly any organ system. Much of the pathogenesis of these diseases is related to dysregulated cytokine activity. Historically, autoimmune and inflammatory diseases have been treated with medications that nonspecifically suppress the immune system. mAbs that block the action of pathogenic cytokines emerged 2 decades ago and have become widely useful. More recently, agents that simultaneously block multiple pathogenic cytokines via inhibition of the downstream Janus kinase (JAK)-signal transducer and activator of transcription pathway have emerged and are becoming increasingly important. These small-mol. inhibitors, collectively termed JAK inhibitors, are US Food and Drug Administration-approved in a few autoimmune/inflammatory disorders and are being evaluated in many others. Here, we review the biol. of the JAK-signal transducer and activator of transcription pathway and the use of JAK inhibitors to treat autoimmune and inflammatory diseases across medical subspecialties.
- 7Jamilloux, Y.; El Jammal, T.; Vuitton, L.; Gerfaud-Valentin, M.; Kerever, S.; Sève, P. JAK inhibitors for the treatment of autoimmune and inflammatory diseases. Autoimmun. Rev. 2019, 18, 102390 DOI: 10.1016/j.autrev.2019.102390Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvFKhtb3P&md5=d31a5113949fdf1391c3d7ceeac3682eJAK inhibitors for the treatment of autoimmune and inflammatory diseasesJamilloux, Yvan; El Jammal, Thomas; Vuitton, Lucine; Gerfaud-Valentin, Mathieu; Kerever, Sebastien; Seve, PascalAutoimmunity Reviews (2019), 18 (11), 102390CODEN: ARUEBU; ISSN:1568-9972. (Elsevier B.V.)A review. Cytokines play a central role in the pathophysiol. of autoimmune and inflammatory diseases. Several cytokines signal through the JAK-STAT pathway, which is now recognized as a major target to inhibit the effect of a wide array of cytokines. JAK inhibitors are increasingly used in the setting of inflammatory and autoimmune diseases. While the currently approved drugs are panJAK inhibitors, more selective small mols. are being developed and tested in various rheumatic disorders. In this extensive review, we present evidence- or hypothesis-based perspectives for these drugs in various rheumatol. conditions, such as rheumatoid arthritis, systemic lupus erythematosus, giant cell arteritis, and autoinflammatory diseases.
- 8Schett, G.; Sticherling, M.; Neurath, M. F. COVID-19: risk for cytokine targeting in chronic inflammatory diseases?. Nat. Rev. Immunol. 2020, 20, 271– 272, DOI: 10.1038/s41577-020-0312-7Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXntFOrs7Y%253D&md5=f3f55b99938ede6632815c92168a61b9COVID-19: risk for cytokine targeting in chronic inflammatory diseases?Schett, Georg; Sticherling, Michael; Neurath, Markus F.Nature Reviews Immunology (2020), 20 (5), 271-272CODEN: NRIABX; ISSN:1474-1733. (Nature Research)COVID-19, caused by the SARS-CoV-2 virus, has become pandemic. With sharply rising infection rates, patient groups characterized by an enhanced infection risk will be challenged by the virus. In this context, patients with chronic immune-mediated inflammatory diseases are of particular interest, as these diseases are characterized by an intrinsic immune dysfunction leading to inflammation that may enhance risk for severe infection.
- 9Wu, D.; Yang, X. O. TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor Fedratinib. J. Microbiol. Immunol. Infect. 2020, 53, 368– 370, DOI: 10.1016/j.jmii.2020.03.005Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlsFalurc%253D&md5=bab4a2496a1381dd062fbaf5349baf57TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor FedratinibWu, Dandan; Yang, Xuexian O.Journal of Microbiology, Immunology and Infection (2020), 53 (3), 368-370CODEN: JMIIFG; ISSN:1995-9133. (Elsevier Taiwan LLC)COVID-19 emerges as a pandemic disease with high mortality. Development of effective prevention and treatment is an urgent need. We reviewed TH17 responses in patients with SARS-CoV-2 and proposed an FDA approved JAK2 inhibitor Fedratinib for reducing mortality of patients with TH17 type immune profiles.
- 10Spinelli, F. R.; Conti, F.; Gadina, M. HiJAKing SARS-CoV-2? The potential role of JAK inhibitors in the management of COVID-19. Sci. Immunol. 2020, 5, eabc5367 DOI: 10.1126/sciimmunol.abc5367Google ScholarThere is no corresponding record for this reference.
- 11Vázquez, J.; López, M.; Gibert, E.; Herrero, E.; Luque, F. J. Merging Ligand-Based and Structure-Based Methods in Drug Discovery: An Overview of Combined Virtual Screening Approaches. Molecules 2020, 25, 4723, DOI: 10.3390/molecules25204723Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1aitrnN&md5=50e00b66bc0191e4f1f076357ebe210fMerging ligand-based and structure-based methods in drug discovery: an overview of combined virtual screening approachesVazquez, Javier; Lopez, Manel; Gibert, Enric; Herrero, Enric; Luque, F. JavierMolecules (2020), 25 (20), 4723CODEN: MOLEFW; ISSN:1420-3049. (MDPI AG)A review. Virtual screening (VS) is an outstanding cornerstone in the drug discovery pipeline. A variety of computational approaches, which are generally classified as ligand-based (LB) and structure-based (SB) techniques, exploit key structural and physicochem. properties of ligands and targets to enable the screening of virtual libraries in the search of active compds. Though LB and SB methods have found widespread application in the discovery of novel drug-like candidates, their complementary natures have stimulated continued efforts toward the development of hybrid strategies that combine LB and SB techniques, integrating them in a holistic computational framework that exploits the available information of both ligand and target to enhance the success of drug discovery projects. In this review, we analyze the main strategies and concepts that have emerged in the last years for defining hybrid LB + SB computational schemes in VS studies. Particularly, attention is focused on the combination of mol. similarity and docking, illustrating them with selected applications taken from the literature.
- 12Lin, T. E.; HuangFu, W.; Chao, M.; Sung, T.; Chang, C.; Chen, Y.; Hsieh, J.; Tu, H.; Huang, H.; Pan, S.; Hsu, K.-C. A novel selective JAK2 inhibitor identified using pharmacological interactions. Front. Pharmacol. 2018, 9, 1379, DOI: 10.3389/fphar.2018.01379Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVGmtLzL&md5=deaab846ce59da3f6aebe14390be1a92A novel selective JAK2 inhibitor identified using pharmacological interactionsLin, Tony Eight; Huangfu, Wei-Chun; Chao, Min-Wu; Sung, Tzu-Ying; Chang, Chao-Di; Chen, Yi-Ying; Hsieh, Jui-Hua; Tu, Huang-Ju; Huang, Han-Li; Pan, Shiow-Lin; Hsu, Kai-ChengFrontiers in Pharmacology (2018), 9 (), 1379CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)The JAK2/STAT signaling pathway mediates cytokine receptor signals that are involved in cell growth, survival and homeostasis. JAK2 is a member of the Janus kinase (JAK) family and aberrant JAK2/STAT is involved with various diseases, making the pathway a therapeutic target. The similarity between the ATP binding site of protein kinases has made development of specific inhibitors difficult. Current JAK2 inhibitors are not selective and produce unwanted side effects. It is thought that increasing selectivity of kinase inhibitors may reduce the side effects seen with current treatment options. Thus, there is a great need for a selective JAK inhibitor. In this study, we identified a JAK2 specific inhibitor. We first identified key pharmacol. interactions in the JAK2 binding site by analyzing known JAK2 inhibitors. Then, we performed structure-based virtual screening and filtered compds. based on their pharmacol. interactions and identified compd. NSC13626 as a potential JAK2 inhibitor. Results of enzymic assays revealed that against a panel of kinases, compd. NSC13626 is a JAK2 inhibitor and has high selectivity toward the JAK2 and JAK3 isoenzymes. Our cellular assays revealed that compd. NSC13626 inhibits colorectal cancer cell (CRC) growth by downregulating phosphorylation of STAT3 and arresting the cell cycle in the S phase. Thus, we believe that compd. NSC13626 has potential to be further optimized as a selective JAK2 drug.
- 13Itteboina, R.; Ballu, S.; Sivan, S. K.; Manga, V. Molecular modeling-driven approach for identification of Janus kinase 1 inhibitors through 3D-QSAR, docking and molecular dynamics simulations. J. Recept. Signal Transduction 2017, 37, 453– 469, DOI: 10.1080/10799893.2017.1328442Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXosVansL0%253D&md5=1cb8a96420c1a6820326e5361adbc389Molecular modeling-driven approach for identification of Janus kinase 1 inhibitors through 3D-QSAR, docking and molecular dynamics simulationsItteboina, Ramesh; Ballu, Srilata; Sivan, Sree Kanth; Manga, VijjulathaJournal of Receptors and Signal Transduction (2017), 37 (5), 453-469CODEN: JRSTCT ISSN:. (Taylor & Francis Ltd.)Janus kinase 1 (JAK 1) belongs to the JAK family of intracellular nonreceptor tyrosine kinase. JAK-signal transducer and activator of transcription (JAK-STAT) pathway mediate signaling by cytokines, which control survival, proliferation and differentiation of a variety of cells. Three-dimensional quant. structure activity relationship (3 D-QSAR), mol. docking and mol. dynamics (MD) methods was carried out on a dataset of Janus kinase 1(JAK 1) inhibitors. Ligands were constructed and docked into the active site of protein using GLIDE 5.6. Best docked poses were selected after anal. for further 3 D-QSAR anal. using comparative mol. field anal. (CoMFA) and comparative mol. similarity indexes anal. (CoMSIA) methodol. Employing 60 mols. in the training set, 3 D-QSAR models were generate that showed good statistical reliability, which is clearly obsd. in terms of r2ncv and q2loo values. The predictive ability of these models was detd. using a test set of 25 mols. that gave acceptable predictive correlation (r2Pred) values. The key amino acid residues were identified by means of mol. docking, and the stability and rationality of the derived mol. conformations were also validated by MD simulation. The good consonance between the docking results and CoMFA/CoMSIA contour maps provides helpful clues about the reasonable modification of mols. in order to design more efficient JAK 1 inhibitors. The developed models are expected to provide some directives for further synthesis of highly effective JAK 1 inhibitors.
- 14Itteboina, R.; Ballu, S.; Sivan, S. K.; Manga, V. Molecular docking, 3D QSAR and dynamics simulation studies of imidazo-pyrrolopyridines as janus kinase 1 (JAK 1) inhibitors. Comput. Biol. Chem. 2016, 64, 33– 46, DOI: 10.1016/j.compbiolchem.2016.04.009Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xotlegu7Y%253D&md5=c90e4120727b9b4df112de83d5cabb93Molecular docking, 3D QSAR and dynamics simulation studies of imidazo-pyrrolopyridines as janus kinase 1 (JAK 1) inhibitorsItteboina, Ramesh; Ballu, Srilata; Sivan, Sree Kanth; Manga, VijjulathaComputational Biology and Chemistry (2016), 64 (), 33-46CODEN: CBCOCH; ISSN:1476-9271. (Elsevier B.V.)Janus kinase 1 (JAK 1) plays a crit. role in initiating responses to cytokines by the JAK-signal transducer and activator of transcription (JAK-STAT). This controls survival, proliferation and differentiation of a variety of cells. Docking, 3D quant. structure activity relationship (3D-QSAR) and mol. dynamics (MD) studies were performed on a series of Imidazo-pyrrolopyridine derivs. reported as JAK 1 inhibitors. QSAR model was generated using 30 mols. in the training set; developed model showed good statistical reliability, which is evident from r2ncv and r2loo values. The predictive ability of this model was detd. using a test set of 13 mols. that gave acceptable predictive correlation (r2Pred) values. Finally, mol. dynamics simulation was performed to validate docking results and MM/GBSA calcns. This facilitated us to compare binding free energies of cocrystal ligand and newly designed mol. R1. The good concordance between the docking results and CoMFA/CoMSIA contour maps afforded obliging clues for the rational modification of mols. to design more potent JAK 1 inhibitors.
- 15Sanachai, K.; Mahalapbutr, P.; Choowongkomon, K.; Poo-Arporn, R. P.; Wolschann, P.; Rungrotmongkol, T. Insights into the binding recognition and susceptibility of tofacitinib toward janus kinases. ACS Omega 2020, 5, 369– 377, DOI: 10.1021/acsomega.9b02800Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVCjtg%253D%253D&md5=de8290f477cc88ae016f67815bb9ee68Insights into the Binding Recognition and Susceptibility of Tofacitinib toward Janus KinasesSanachai, Kamonpan; Mahalapbutr, Panupong; Choowongkomon, Kiattawee; Poo-arporn, Rungtiva P.; Wolschann, Peter; Rungrotmongkol, ThanyadaACS Omega (2020), 5 (1), 369-377CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)Janus kinases (JAKs) are enzymes involved in signaling pathways that affect hematopoiesis and immune cell functions. JAK1, JAK2, and JAK3 play different roles in numerous diseases of the immune system and have also been considered as potential targets for cancer therapy. In the present study, the susceptibility of the oral JAK inhibitor tofacitinib against these three JAKs was elucidated using the 500-ns mol. dynamics (MD) simulations and free energy calcns. based on MM-PB(GB)SA, QM/MM-GBSA (PM3 and SCC-DFTB), and SIE methods. The obtained results revealed that tofacitinib could interact with all JAKs at the ATP-binding site via electrostatic attraction, hydrogen bond formation, and in particular van der Waals interaction. The conserved glutamate and leucine residues (E957 and L959 of JAK1, E930 and L932 of JAK2, and E903 and L905 of JAK3) located in the hinge region stabilized tofacitinib binding through strongly formed hydrogen bonds. Complexation with the incoming tofacitinib led to a closed conformation of the ATP-binding site and a decreased protein fluctuation at the glycine loop of the JAK protein. The binding affinities of tofacitinib/JAKs were ranked in the order of JAK3 > JAK2 ∼ JAK1, which are in line with the reported exptl. data.
- 16Clark, J. D.; Flanagan, M. E.; Telliez, J. Discovery and development of Janus Kinase (JAK) inhibitors for inflammatory diseases: Miniperspective. J. Med. Chem. 2014, 57, 5023– 5038, DOI: 10.1021/jm401490pGoogle Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVCjsA%253D%253D&md5=6763ae2d46f52ddcda23e235a60634aeDiscovery and Development of Janus Kinase (JAK) Inhibitors for Inflammatory DiseasesClark, James D.; Flanagan, Mark E.; Telliez, Jean-BaptisteJournal of Medicinal Chemistry (2014), 57 (12), 5023-5038CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The Janus kinases (JAKs) are a family of intracellular tyrosine kinases that play an essential role in the signaling of numerous cytokines that have been implicated in the pathogenesis of inflammatory diseases. As a consequence, the JAKs have received significant attention in recent years from the pharmaceutical and biotechnol. industries as therapeutic targets. Here, we provide a review of the JAK pathways, the structure, function, and activation of the JAK enzymes followed by a detailed look at the JAK inhibitors currently in the clinic or approved for these indications. Finally, a perspective is provided on what the past decade of research with JAK inhibitors for inflammatory indications has taught along with thoughts on what the future may hold in terms of addressing the opportunities and challenges that remain.
- 17Yao, T.; Xie, J.; Liu, X.; Cheng, J.; Zhu, C.; Zhao, J.; Dong, X. Integration of pharmacophore mapping and molecular docking in sequential virtual screening: towards the discovery of novel JAK2 inhibitors. RSC Adv. 2017, 7, 10353– 10360, DOI: 10.1039/C6RA24959KGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXit1Wgu7o%253D&md5=be2ddd7109bfb150b99d9d762b7e35b1Integration of pharmacophore mapping and molecular docking in sequential virtual screening: towards the discovery of novel JAK2 inhibitorsYao, Ting-Ting; Xie, Jiang-Feng; Liu, Xing-Guo; Cheng, Jing-Li; Zhu, Cheng-Yuan; Zhao, Jin-Hao; Dong, Xiao-WuRSC Advances (2017), 7 (17), 10353-10360CODEN: RSCACL; ISSN:2046-2069. (Royal Society of Chemistry)An integrated virtual screening protocol by combining mol. docking and pharmacophore mapping was established to identify novel inhibitors of JAK2 from a com. compd. database. Twelve novel and structurally diverse hits were selected and subjected to in vitro biol. tests, and three compds. (A5, A6 and A9) with remarkable JAK2 inhibitory activity were identified. Then, the obtained structures were further used as the template for a subsequent similarity search, leading to the identification of another two promising compds. (B2 and B4). Selectivity profiles of JAK subtype and in vitro anti-cancer activity of the promising compds. were studied, revealing the promising compd. B2 was of interest for further study because of its JAK2 selective profile, novelty of skeleton and significantly anti-proliferative effect against cancer cells. Finally, binding patterns of the compds. A5 and B2 were explored to provide a deeper insight for further structural optimization.
- 18Galvez, J.; Galvez-Llompart, M.; Garcia-Domenech, R. Introduction to molecular topology: basic concepts and application to drug design. Curr. Comput.-Aided Drug Des. 2012, 8, 196– 223, DOI: 10.2174/157340912801619094Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlKqu7nP&md5=6a6834004023ce4245631c184e9c34a6Introduction to molecular topology: basic concepts and application to drug designGalvez, Jorge; Galvez-Llompart, Maria; Garcia-Domenech, RamonCurrent Computer-Aided Drug Design (2012), 8 (3), 196-223CODEN: CCDDAS; ISSN:1573-4099. (Bentham Science Publishers Ltd.)In this review it is dealt the use of mol. topol. (MT) in the selection and design of new drugs. After an introduction of the actual methods used for drug design, the basic concepts of MT are defined, including examples of calcn. of topol. indexes, which are numerical descriptors of mol. structures. The goal is making this calcn. familiar to the potential students and allowing a straightforward comprehension of the topic. Finally, the achievements obtained in this field are detailed, so that the reader can figure out the great interest of this approach.
- 19Zanni, R.; Galvez-Llompart, M.; Garcia-Domenech, R.; Galvez, J. What place does molecular topology have in today’s drug discovery?. Expert Opin. Drug Discovery 2020, 15, 1133– 1144, DOI: 10.1080/17460441.2020.1770223Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVWgtLnN&md5=4644090178dc2b3028f4298869c8fb3dWhat place does molecular topology have in today's drug discoveryZanni, Riccardo; Galvez-Llompart, Maria; Garcia-Domenech, Ramon; Galvez, JorgeExpert Opinion on Drug Discovery (2020), 15 (10), 1133-1144CODEN: EODDBX; ISSN:1746-0441. (Taylor & Francis Ltd.)Introduction Most methods in mol. and drug design are currently based on physicochem. descriptors. However, mol. topol., which relies on topol. descriptors, has also shown value for mol. design even if it does not take into account the phys. or chem. properties of ligands and receptors, including the ligand-receptor interaction itself. Areas covered Herein, the authors provide new insights into the importance of mol. topol. according to some of the latest discoveries in physics and chem. Furthermore, the authors report on the most significant achievements in drug design using mol. topol. over the last 5 years and give their expert perspectives on the subject as a whole. Expert opinionMol. topol. is a new paradigm which is independent of physicochem. mol. descriptors. This fact explains the viability of both the discovery of new lead compds. with a min. of information derived from math.-topol. patterns and the interpretation results in structural and physicochem. terms.
- 20Zak, M.; Hurley, C. A.; Ward, S. I.; Bergeron, P.; Barrett, K.; Balazs, M.; Blair, W. S.; Bull, R.; Chakravarty, P.; Chang, C.; Crackett, P.; Deshmukh, G.; DeVoss, J.; Dragovich, P. S.; Eigenbrot, C.; Ellwood, C.; Gaines, S.; Ghilardi, N.; Gibbons, P.; Gradl, S.; Gribling, P.; Hamman, C.; Harstad, E.; Hewitt, P.; Johnson, A.; Johnson, T.; Kenny, J. R.; Koehler, M. F.; Bir Kohli, P.; Labadie, S.; Lee, W. P.; Liao, J.; Liimatta, M.; Mendonca, R.; Narukulla, R.; Pulk, R.; Reeve, A.; Savage, S.; Shia, S.; Steffek, M.; Ubhayakar, S.; van Abbema, A.; Aliagas, I.; Avitabile-Woo, B.; Xiao, Y.; Yang, J.; Kulagowski, J. J. Identification of C-2 hydroxyethyl imidazopyrrolopyridines as potent JAK1 inhibitors with favorable physicochemical properties and high selectivity over JAK2. J. Med. Chem. 2013, 56, 4764– 4785, DOI: 10.1021/jm4004895Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnsVWit74%253D&md5=0b0d1950adebf325cc434344f57ca139Identification of C-2 Hydroxyethyl Imidazopyrrolopyridines as Potent JAK1 Inhibitors with Favorable Physicochemical Properties and High Selectivity over JAK2Zak, Mark; Hurley, Christopher A.; Ward, Stuart I.; Bergeron, Philippe; Barrett, Kathy; Balazs, Mercedesz; Blair, Wade S.; Bull, Richard; Chakravarty, Paroma; Chang, Christine; Crackett, Peter; Deshmukh, Gauri; DeVoss, Jason; Dragovich, Peter S.; Eigenbrot, Charles; Ellwood, Charles; Gaines, Simon; Ghilardi, Nico; Gibbons, Paul; Gradl, Stefan; Gribling, Peter; Hamman, Chris; Harstad, Eric; Hewitt, Peter; Johnson, Adam; Johnson, Tony; Kenny, Jane R.; Koehler, Michael F. T.; Bir Kohli, Pawan; Labadie, Sharada; Lee, Wyne P.; Liao, Jiangpeng; Liimatta, Marya; Mendonca, Rohan; Narukulla, Raman; Pulk, Rebecca; Reeve, Austin; Savage, Scott; Shia, Steven; Steffek, Micah; Ubhayakar, Savita; van Abbema, Anne; Aliagas, Ignacio; Avitabile-Woo, Barbara; Xiao, Yisong; Yang, Jing; Kulagowski, Janusz J.Journal of Medicinal Chemistry (2013), 56 (11), 4764-4785CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Herein we report on the structure-based discovery of a C-2 hydroxyethyl moiety which provided consistently high levels of selectivity for JAK1 over JAK2 to the imidazopyrrolopyridine series of JAK1 inhibitors. X-ray structures of a C-2 hydroxyethyl analog in complex with both JAK1 and JAK2 revealed differential ligand/protein interactions between the two isoforms and offered an explanation for the obsd. selectivity. Anal. of historical data from related mols. was used to develop a set of physicochem. compd. design parameters to impart desirable properties such as acceptable membrane permeability, potent whole blood activity, and a high degree of metabolic stability. This work culminated in the identification of a highly JAK1 selective compd. (31) exhibiting favorable oral bioavailability across a range of preclin. species and robust efficacy in a rat CIA model.
- 21Hart, A. C.; Schroeder, G. M.; Wan, H.; Grebinski, J.; Inghrim, J.; Kempson, J.; Guo, J.; Pitts, W. J.; Tokarski, J. S.; Sack, J. S.; Khan, J. A.; Lippy, J.; Lorenzi, M. V.; You, D.; McDevitt, T.; Vuppugalla, R.; Zhang, Y.; Lombardo, L. J.; Trainor, G. L.; Purandare, A. V. Structure-Based Design of Selective Janus Kinase 2 Imidazo[4,5-d]pyrrolo[2,3-b]pyridine Inhibitors. ACS Med. Chem. Lett. 2015, 6, 845– 849, DOI: 10.1021/acsmedchemlett.5b00225Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFCrurvP&md5=c9b4e51332496a1b29cc8f4c187d488aStructure-Based Design of Selective Janus Kinase 2 Imidazo[4,5-d]pyrrolo[2,3-b]pyridine InhibitorsHart, Amy C.; Schroeder, Gretchen M.; Wan, Honghe; Grebinski, James; Inghrim, Jennifer; Kempson, James; Guo, Junqing; Pitts, William J.; Tokarski, John S.; Sack, John S.; Khan, Javed A.; Lippy, Jonathan; Lorenzi, Matthew V.; You, Dan; McDevitt, Theresa; Vuppugalla, Ragini; Zhang, Yueping; Lombardo, Louis J.; Trainor, George L.; Purandare, Ashok V.ACS Medicinal Chemistry Letters (2015), 6 (8), 845-849CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Early hit to lead work on a pyrrolopyridine chemotype provided access to compds. with biochem. and cellular potency against Janus kinase 2 (JAK2). Structure-based drug design along the extended hinge region of JAK2 led to the identification of an important H-bond interaction with the side chain of Tyr 931, which improved JAK family selectivity. The 4,5-di-Me thiazole analog I demonstrated high levels of JAK family selectivity and was identified as a promising lead for the program.
- 22Forster, M.; Chaikuad, A.; Dimitrov, T.; Döring, E.; Holstein, J.; Berger, B.-T.; Gehringer, M.; Ghoreschi, K.; Müller, S.; Knapp, S.; Laufer, S. A. Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4- d]pyrrolo[2,3- b]pyridine Scaffold. J. Med. Chem. 2018, 61, 5350– 5366, DOI: 10.1021/acs.jmedchem.8b00571Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVGjsrbN&md5=8acdec137df746f854bbee75a10d5f70Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4-d]pyrrolo[2,3-b]pyridine ScaffoldForster, Michael; Chaikuad, Apirat; Dimitrov, Teodor; Doering, Eva; Holstein, Julia; Berger, Benedict-Tilman; Gehringer, Matthias; Ghoreschi, Kamran; Mueller, Susanne; Knapp, Stefan; Laufer, Stefan A.Journal of Medicinal Chemistry (2018), 61 (12), 5350-5366CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Janus kinases are major drivers of immune signaling and have been the focus of anti-inflammatory drug discovery for more than a decade. Because of the invariable colocalization of JAK1 and JAK3 at cytokine receptors, the question if selective JAK3 inhibition is sufficient to effectively block downstream signaling has been highly controversial. Recently, we discovered the covalent-reversible JAK3 inhibitor FM-381 (23) featuring high isoform and kinome selectivity. Crystallog. revealed that this inhibitor induces an unprecedented binding pocket by interactions of a nitrile substituent with arginine residues in JAK3. Herein, we describe detailed structure-activity relationships necessary for induction of the arginine pocket and the impact of this structural change on potency, isoform selectivity, and efficacy in cellular models. Furthermore, we evaluated the stability of this novel inhibitor class in in vitro metabolic assays and were able to demonstrate an adequate stability of key compd. 23 for in vivo use.
- 23De Vivo, M.; Masetti, M.; Bottegoni, G.; Cavalli, A. Role of Molecular Dynamics and Related Methods in Drug Discovery. J. Med. Chem. 2016, 59, 4035– 4061, DOI: 10.1021/acs.jmedchem.5b01684Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlWksLs%253D&md5=b3d8a4fb5705a3555c2bb2a962420396Role of Molecular Dynamics and Related Methods in Drug DiscoveryDe Vivo, Marco; Masetti, Matteo; Bottegoni, Giovanni; Cavalli, AndreaJournal of Medicinal Chemistry (2016), 59 (9), 4035-4061CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mol. dynamics (MD) and related methods are close to becoming routine computational tools for drug discovery. Their main advantage is in explicitly treating structural flexibility and entropic effects. This allows a more accurate est. of the thermodn. and kinetics assocd. with drug-target recognition and binding, as better algorithms and hardware architectures increase their use. Here, we review the theor. background of MD and enhanced sampling methods, focusing on free-energy perturbation, metadynamics, steered MD, and other methods most consistently used to study drug-target binding. We discuss unbiased MD simulations that nowadays allow the observation of unsupervised ligand-target binding, assessing how these approaches help optimizing target affinity and drug residence time toward improved drug efficacy. Further issues discussed include allosteric modulation and the role of water mols. in ligand binding and optimization. We conclude by calling for more prospective studies to attest to these methods' utility in discovering novel drug candidates.
- 24Guterres, H.; Im, W. Improving Protein-Ligand Docking Results with High-Throughput Molecular Dynamics Simulations. J. Chem. Inf. Model. 2020, 60, 2189– 2198, DOI: 10.1021/acs.jcim.0c00057Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFCju7g%253D&md5=3c5d10e0584d0218071dc8d0a37179e7Improving Protein-Ligand Docking Results with High-Throughput Molecular Dynamics SimulationsGuterres, Hugo; Im, WonpilJournal of Chemical Information and Modeling (2020), 60 (4), 2189-2198CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Structure-based virtual screening relies on classical scoring functions that often fail to reliably discriminate binders from nonbinders. In this work, the authors present a high-throughput protein-ligand complex mol. dynamics (MD) simulation that uses the output from AutoDock Vina to improve docking results in distinguishing active from decoy ligands in a directory of useful decoy-enhanced (DUD-E) dataset. MD trajectories are processed by evaluating ligand-binding stability using root-mean-square deviations. The authors select 56 protein targets (of 7 different protein classes) and 560 ligands (280 actives, 280 decoys) and show 22% improvement in ROC AUC (area under the curve, receiver operating characteristics curve), from an initial value of 0.68 (AutoDock Vina) to a final value of 0.83. The MD simulation demonstrates a robust performance across all seven different protein classes. In addn., some predicted ligand-binding modes are moderately refined during MD simulations. These results systematically validate the reliability of a physics-based approach to evaluate protein-ligand binding interactions.
- 25Gioia, D.; Bertazzo, M.; Recanatini, M.; Masetti, M.; Cavalli, A. Dynamic docking: A paradigm shift in computational drug discovery. Molecules 2017, 22, 2029, DOI: 10.3390/molecules22112029Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitl2jtLk%253D&md5=b835af2568bb7f5b0500d6739523ad4eDynamic docking: a paradigm shift in computational drug discoveryGioia, Dario; Bertazzo, Martina; Recanatini, Maurizio; Masetti, Matteo; Cavalli, AndreaMolecules (2017), 22 (11), 2029/1-2029/21CODEN: MOLEFW; ISSN:1420-3049. (MDPI AG)Mol. docking is the methodol. of choice for studying in silico protein-ligand binding and for prioritizing compds. to discover new lead candidates. Traditional docking simulations suffer from major limitations, mostly related to the static or semi-flexible treatment of ligands and targets. They also neglect solvation and entropic effects, which strongly limits their predictive power. During the last decade, methods based on full atomistic mol. dynamics (MD) have emerged as a valid alternative for simulating macromol. complexes. In principle, compared to traditional docking, MD allows the full exploration of drug-target recognition and binding from both the mechanistic and energetic points of view (dynamic docking). Binding and unbinding kinetic consts. can also be detd. While dynamic docking is still too computationally expensive to be routinely used in fast-paced drug discovery programs, the advent of faster computing architectures and advanced simulation methodologies are changing this scenario. It is feasible that dynamic docking will replace static docking approaches in the near future, leading to a major paradigm shift in in silico drug discovery. Against this background, we review the key achievements that have paved the way for this progress.
- 26Selleck. https://www.selleckchem.com/ Last access: April 2021.Google ScholarThere is no corresponding record for this reference.
- 27BioCrick. https://www.biocrick.com/LY2784544-BCC2200.html Last access: April 2021.Google ScholarThere is no corresponding record for this reference.
- 28Boc-Science. https://www.bocsci.com Last access: April 2021.Google ScholarThere is no corresponding record for this reference.
- 29Alvascience Srl, alvaDesc (software for molecular descriptors calculation), 2019, https://www.alvascience.com.Google ScholarThere is no corresponding record for this reference.
- 30O’Shea, J. J.; Kontzias, A.; Yamaoka, K.; Tanaka, Y.; Laurence, A. Janus kinase inhibitors in autoimmune diseases. Ann. Rheum. Dis. 2013, 72, ii111– ii115, DOI: 10.1136/annrheumdis-2012-202576Google ScholarThere is no corresponding record for this reference.
- 31Kubler, P. Janus kinase inhibitors: Mechanisms of action. Aust. Prescr. 2014, 37, 154– 157, DOI: 10.18773/austprescr.2014.061Google ScholarThere is no corresponding record for this reference.
- 32Roskoski, R., Jr. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol. Res. 2016, 111, 784– 803, DOI: 10.1016/j.phrs.2016.07.038Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht12jurzJ&md5=a45fba1bb110eff4d5c1b31b78c1b3acJanus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseasesRoskoski, Robert, Jr.Pharmacological Research (2016), 111 (), 784-803CODEN: PHMREP; ISSN:1043-6618. (Elsevier Ltd.)The Janus kinase (JAK) family of non-receptor protein-tyrosine kinases consists of JAK1, JAK2, JAK3, and TYK2 (tyrosine kinase-2). Each of these proteins contains a JAK homol. pseudokinase (JH2) domain that regulates the adjacent protein kinase domain (JH1). JAK1/2 and TYK2 are ubiquitously expressed whereas JAK3 is found predominantly in hematopoietic cells. The Janus kinase family is regulated by numerous cytokines including interleukins, interferons, and hormones such as erythropoietin, thrombopoietin, and growth hormone. Ligand binding to cytokine and hormone receptors leads to the activation of assocd. Janus kinases, which then mediate the phosphorylation of the receptors. The SH2 domain of STATs (signal transducers and activators of transcription) binds to the receptor phosphotyrosines thereby promoting STAT phosphorylation by the Janus kinases and consequent activation. STAT dimers are translocated to the nucleus where they participate in the regulation of the expression of thousands of proteins. JAK-STAT dysregulation results in autoimmune disorders such as rheumatoid arthritis, ulcerative colitis, and Crohn disease. JAK-STAT dysregulation also plays a role in the pathogenesis of myelofibrosis, polycythemia vera, and other myeloproliferative illnesses. An activating JAK2 V617F mutation occurs in 95% of people with polycythemia vera and in a lower percentage of people with other neoplasms. JAK1/3 signaling participates in the pathogenesis of inflammatory afflictions while JAK1/2 signaling participates in the development of several malignancies including leukemias and lymphomas as well as myeloproliferative neoplasms. Tofacitinib is a pan-JAK inhibitor that is approved by the FDA for the treatment of rheumatoid arthritis and ruxolitinib is a JAK1/2 inhibitor that is approved for the treatment of polycythemia vera and myelofibrosis.
- 33Tocris DB. https://www.tocris.com/. Last access: January 2019.Google ScholarThere is no corresponding record for this reference.
- 34ABCAM. www.abcam.com. Last access: January 2019.Google ScholarThere is no corresponding record for this reference.
- 35Hastie, T.; Tibshirani, R.; Friedman, J. The elements of statistical learning: data mining, inference, and prediction; Springer Science & Business Media, 2009.Google ScholarThere is no corresponding record for this reference.
- 36StatSoft IS, version 9.0; Tulsa, OK, USA, 2009.Google ScholarThere is no corresponding record for this reference.
- 37Gálvez, J.; García-Domenech, R.; de Gregorio Alapont, C.; de Julián-Ortiz, J. V.; Popa, L. Pharmacological distribution diagrams: a tool for de novo drug design. J. Mol. Graph. 1996, 14, 272– 276, DOI: 10.1016/S0263-7855(96)00081-1Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXitlCrur8%253D&md5=84cce553319aa39f1d2c349b0338c54ePharmacological distribution diagrams: a tool for de novo drug designGalvez, J.; Garcia-Domenech, R.; de Gregorio Alapont, C.; de Julian-Ortiz, J. V.; Popa, L.Journal of Molecular Graphics (1996), 14 (5), 272-276CODEN: JMGRDV; ISSN:0263-7855. (Elsevier)Discriminant anal. applied to SAR studies using topol. descriptors allows us to plot frequency distribution diagrams: a function of the no. of drugs within an interval of values of discriminant function vs. these values. We make use of these representations, pharmacol. distribution diagrams (PDDs), in structurally heterogeneous groups where generally they adopt skewed Gaussian shapes or present several maxima. The maxima afford intervals of discriminant function in which exists a good expectancy to find new active drugs. A set of β-blockers with contrasted activity has been selected to test the ability of PPDs as a visualizing technique, for the identification of new β-blocker active compds.
- 38Gramatica, P. Principles of QSAR models validation: internal and external. QSAR Comb. Sci. 2007, 26, 694– 701, DOI: 10.1002/qsar.200610151Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtFKjtrs%253D&md5=05af99ce1a70f5f14ab8a645fbaf00d2Principles of QSAR models validation: internal and externalGramatica, PaolaQSAR & Combinatorial Science (2007), 26 (5), 694-701CODEN: QCSSAU; ISSN:1611-020X. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The recent REACH Policy of the European Union has led to scientists and regulators to focus their attention on establishing general validation principles for QSAR models in the context of chem. regulation (previously known as the Setubal, nowadays, the OECD principles). This paper gives a brief anal. of some principles: unambiguous algorithm, Applicability Domain (AD), and statistical validation. Some concerns related to QSAR algorithm reproducibility and an example of a fast check of the applicability domain for MLR models are presented. Common myths and misconceptions related to popular techniques for verifying internal predictivity, particularly for MLR models (for instance cross-validation, bootstrap), are commented on and compared with commonly used statistical techniques for external validation. The differences in the two validating approaches are highlighted, and evidence is presented that only models that were validated externally, after their internal validation, can be considered reliable and applicable for both external prediction and regulatory purposes.
- 39Schrodinger, L. Schrodinger software suite; Schrödinger, LLC: New York, 2019, p 670.Google ScholarThere is no corresponding record for this reference.
- 40Mosmann, T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55– 63, DOI: 10.1016/0022-1759(83)90303-4Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL2c%252FovFSmtw%253D%253D&md5=c1116f4ca7a7d2b79fab2a0bd2c2d439Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assaysMosmann TJournal of immunological methods (1983), 65 (1-2), 55-63 ISSN:0022-1759.A tetrazolium salt has been used to develop a quantitative colorimetric assay for mammalian cell survival and proliferation. The assay detects living, but not dead cells and the signal generated is dependent on the degree of activation of the cells. This method can therefore be used to measure cytotoxicity, proliferation or activation. The results can be read on a multiwell scanning spectrophotometer (ELISA reader) and show a high degree of precision. No washing steps are used in the assay. The main advantages of the colorimetric assay are its rapidity and precision, and the lack of any radioisotope. We have used the assay to measure proliferative lymphokines, mitogen stimulations and complement-mediated lysis.
Cited By
This article is cited by 3 publications.
- Riccardo Zanni, Jesús Martínez-Cruz, María Gálvez-Llompart, Dolores Fernández-Ortuño, Diego Romero, Ramón García-Domènech, Alejandro Pérez-García, Jorge Gálvez. Rational Design of Chitin Deacetylase Inhibitors for Sustainable Agricultural Use Based on Molecular Topology. Journal of Agricultural and Food Chemistry 2022, 70
(41)
, 13118-13131. https://doi.org/10.1021/acs.jafc.2c02377
- María Gálvez‐Llompart, German Sastre. 1Machine Learning Search for Suitable Structure Directing Agents for the Synthesis of Beta (
BEA
) Zeolite Using Molecular Topology and Monte Carlo Techniques. 2023, 61-80. https://doi.org/10.1002/9781119819783.ch3
- Rita Maria Concetta Di Martino, Andrea Cavalli, Giovanni Bottegoni. Dopamine D3 receptor ligands: a patent review (2014–2020). Expert Opinion on Therapeutic Patents 2022, 32
(6)
, 605-627. https://doi.org/10.1080/13543776.2022.2049240
Abstract
Figure 1
Figure 1. Screening and profiling workflow employed in this work.
Figure 2
Figure 2. Chemical graph and adjacency matrix of the isopentane.
Figure 3
Figure 3. Example of SRW05 and CIC2 values for DFpan for two JAK inhibitors (Tofacitinib and NSC33994) and two decoys (102-05-6 and 119-24-4).
Figure 4
Figure 4. Examples of GATS6m values for two JAK inhibitors (Hexabromocyclohexane and AG490) and two decoys (12334-10-1 and 114145-29-8).
Figure 5
Figure 5. PDD for the general model. Blue bars represent the distribution of JAK inhibitors, and orange bars represent decoys.
Figure 6
Figure 6. PDD for the subtype-specific models: DF1, DF2, and DF3 in panels (a), (b), and (c), respectively. Black bars represent the distribution of JAK inhibitors, and white bars represent decoys.
Figure 7
Figure 7. Representation of the binding sites of the JAK1, JAK2, and JAK3 structures in complex with the native ligands that were selected for the docking calculations (PDB-ID: 4IVD, 5CF6, and 6GLA, respectively). (20−22) The conserved residues are represented as sticks with carbon atoms colored in white, while specific amino acids are differently colored (orange, yellow, and green for JAK1, JAK2, and JAK3, respectively). Hydrogen bonds are explicitly reported as black dots.
Figure 8
Figure 8. Chemical representation and codification for the eight selected compounds as potential JAK inhibitors.
Figure 9
Figure 9. Ball and stick representation of the three top-ranked compounds for each JAK subtype: 5 bound to JAK1, 2 bound to JAK2, and 6 bound to JAK3 (panels b, d, and f, respectively). For comparison, the binding mode of the reference compound Tofacitinib is also reported (panels a, c, and e for JAK1, JAK2, and JAK3, respectively).
Figure 10
Figure 10. RMSD values of Cα atoms of the three JAK subtypes (a: JAK1; b: JAK2; and c: JAK3) in the complexes with the best-ranking molecule for each subtype and Tofacitinib along the 50 ns of MD simulations (blue lines). In purple, the RMSD computed using the heavy atoms of the ligands (after least-squares-fit superimposition to the Cα atoms of the protein) is also shown.
Figure 11
Figure 11. Protein–ligand contact interaction over the MD trajectory. Hydrogen bonds are shown in blue, water-mediated hydrogen bonds in red, hydrophobic interactions in gray, salt bridges in yellow, π–π interactions in green, and cation−π interactions in orange.
Figure 12
Figure 12. Cell viability of SH-SY5Y cells exposed to different concentrations of tested drugs for 5 and 24 h, evaluated by the MTT assay. Data are expressed as a percentage of OD values of treated cells compared to vehicle-treated ones and reported as the mean ± standard error of the mean (SEM) (*p < 0.05; **p < 0.01 vs the respective control, one-way analysis of variance (ANOVA) test followed by Dunnett’s test).
Figure 13
Figure 13. Chemical structure and IC50 (μM) for commercially available JAK2 inhibitors. (26−28)
Figure 14
Figure 14. Amino acid interaction between in vitro-tested compounds 4 (a) and 7 (b) and JAK2 (PDB: 5CF6).
References
ARTICLE SECTIONSThis article references 40 other publications.
- 1Harpur, A. G.; Andres, A. C.; Ziemiecki, A.; Aston, R. R.; Wilks, A. F. JAK2, a third member of the JAK family of protein tyrosine kinases. Oncogene 1992, 7, 1347– 1353Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XmtVOrtLc%253D&md5=db385133406115942157615e9c56b47fJAK2, a third member of the JAK family of protein tyrosine kinasesHarpur, Ailsa G.; Andres, Anne Catherine; Ziemiecki, Andrew; Aston, Raja R.; Wilks, Andrew F.Oncogene (1992), 7 (7), 1347-53CODEN: ONCNES; ISSN:0950-9232.CDNA clones encoding a third, widely expressed, member of the JAK family of protein tyrosine kinases (PTKs) were isolated. The anticipated amino acid sequence of JAK2 predicts the presence of two kinase-related domains, a feature characteristic of this family of PTKs. The structural similarity of JAK2 to the other members of this family extends towards their N-termini, beyond the two kinase-related domains, and reveals five further domains of substantial amino acid similarity. The C-terminal portion of one of these domains, the JH4 domain, bears an intriguing, albeit tenuous, similarity to the core element of the SH2 domain, whereas the remaining JAK homol. domains do not appear to be a feature of other known proteins.
- 2Kawamura, M.; McVicar, D. W.; Johnston, J. A.; Blake, T. B.; Chen, Y.; Lal, B. K.; Lloyd, A. R.; Kelvin, D. J.; Staples, J. E.; Ortaldo, J. R. Molecular cloning of L-JAK, a Janus family protein-tyrosine kinase expressed in natural killer cells and activated leukocytes. Proc. Natl. Acad. Sci. U.S.A. 1994, 91, 6374– 6378, DOI: 10.1073/pnas.91.14.6374Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXlvF2jurY%253D&md5=c898c32fb7ff39b6987cca1717d9f229Molecular cloning of L-JAK, a Janus family protein-tyrosine kinase expressed in natural killer cells and activated leukocytesKawamura, Masaru; McVicar, Daniel W.; Johnston, James A.; Blake, Trevor B.; Chen, Yi-Qing; Lal, Brajesh K.; Lloyd, Andrew R.; Kelvin, David J.; Staples, J. Erin; et al.Proceedings of the National Academy of Sciences of the United States of America (1994), 91 (14), 6374-8CODEN: PNASA6; ISSN:0027-8424.Protein-tyrosine kinases (PTKs) are crit. enzymes for receptor-mediated signaling in lymphocytes. Because natural killer (NK) cells are large granular lymphocytes with specialized effector function, PTKs preferentially expressed in these cells were investigated. One such PTK was identified and molecularly cloned. The predicted amino acid sequence shows that this kinase lacks SH2 or SH3 domains typical of src family kinases but has tandem nonidentical catalytic domains, indicating that it is a member of the Janus family of PTKs. Immunopptn. using antiserum generated against a peptide corresponding to the deduced amino acid sequence of this gene revealed a kinase with a mol. wt. of ∼125,000. The pattern of expression of this kinase contrasted sharply with that of other Janus kinases, which are ubiquitously expressed. The kinase described in the present study was more limited in its expression; expression was found in NK cells and an NK-like cell line but not in resting T cells or in other tissues. In contrast, stimulated and transformed T cells expressed the gene, suggesting a role in lymphoid activation. Because of its homol. and tissue expression, this PTK gene was tentatively termed L-JAK for leukocyte Janus kinase.
- 3Wilks, A. F. The JAK kinases: not just another kinase drug discovery target. In Seminars in cell & developmental biology; Elsevier, 2008; Vol. 19, pp 319– 328.Google ScholarThere is no corresponding record for this reference.
- 4Williams, N. K.; Bamert, R. S.; Patel, O.; Wang, C.; Walden, P. M.; Wilks, A. F.; Fantino, E.; Rossjohn, J.; Lucet, I. S. Dissecting specificity in the Janus kinases: the structures of JAK-specific inhibitors complexed to the JAK1 and JAK2 protein tyrosine kinase domains. J. Mol. Biol. 2009, 387, 219– 232, DOI: 10.1016/j.jmb.2009.01.041Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXivVegtbo%253D&md5=79957755d04323057b5acf7e9d2c56caDissecting Specificity in the Janus Kinases: The Structures of JAK-Specific Inhibitors Complexed to the JAK1 and JAK2 Protein Tyrosine Kinase DomainsWilliams, Neal K.; Bamert, Rebecca S.; Patel, Onisha; Wang, Christina; Walden, Patricia M.; Wilks, Andrew F.; Fantino, Emmanuelle; Rossjohn, Jamie; Lucet, Isabelle S.Journal of Molecular Biology (2009), 387 (1), 219-232CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)The Janus kinases (JAKs) are a pivotal family of protein tyrosine kinases (PTKs) that play prominent roles in numerous cytokine signaling pathways, with aberrant JAK activity assocd. with a variety of hematopoietic malignancies, cardiovascular diseases and immune-related disorders. Whereas the structures of the JAK2 and JAK3 PTK domains have been detd., the structure of the JAK1 PTK domain is unknown. Here, we report the high-resoln. crystal structures of the "active form" of the JAK1 PTK domain in complex with two JAK inhibitors, a tetracyclic pyridone 2-t-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-one (CMP6) and (3R,4R)-3-[4-methyl-3-[N-methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]piperidin-1-yl]-3-oxopropionitrile (CP-690,550), and compare them with the corresponding JAK2 PTK inhibitor complexes. Both inhibitors bound in a similar manner to JAK1, namely buried deep within a constricted ATP-binding site, thereby providing a basis for the potent inhibition of JAK1. As expected, the mode of inhibitor binding in JAK1 was very similar to that obsd. in JAK2, highlighting the challenges in developing JAK-specific inhibitors that target the ATP-binding site. Nevertheless, differences surrounding the JAK1 and JAK2 ATP-binding sites were apparent, thereby providing a platform for the rational design of JAK2- and JAK1-specific inhibitors.
- 5Saharinen, P.; Takaluoma, K.; Silvennoinen, O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain. Mol. Cell. Biol. 2000, 20, 3387– 3395, DOI: 10.1128/MCB.20.10.3387-3395.2000Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXivFyhtrw%253D&md5=17ac4581ee9c36e03e95f0f88405339cRegulation of the Jak2 tyrosine kinase by its pseudokinase domainSaharinen, Pipsa; Takaluoma, Kati; Silvennoinen, OlliMolecular and Cellular Biology (2000), 20 (10), 3387-3395CODEN: MCEBD4; ISSN:0270-7306. (American Society for Microbiology)Activation of Jak tyrosine kinases through hematopoietic cytokine receptors occurs as a consequence of ligand-induced aggregation of receptor-assocd. Jaks and their subsequent autophosphorylation. Jak kinases consist of a C-terminal tyrosine kinase domain, a pseudokinase domain of unknown function, and Jak homol. (JH) domains 3 to 7, implicated in receptor-Jak interaction. We analyzed the functional roles of the different protein domains in activation of Jak2. Deletion anal. of Jak2 showed that the pseudokinase domain but not JH domains 3 to 7 neg. regulated the catalytic activity of Jak2 as well Jak2-mediated activation of Stat5. Phosphorylation of Stat5 by wild-type Jak2 was dependent on the SH2 domain of Stat5; however, this requirement was lost upon deletion of the pseudokinase domain of Jak2. Investigation of the mechanisms of the pseudokinase domain-mediated inhibition of Jak2 suggested that this regulation did not involve protein tyrosine phosphatases. Instead, anal. of interactions between the tyrosine kinase domain and Jak2 suggested that the pseudokinase domain interacted with the kinase domain. Furthermore, co-expression of the pseudokinase domain inhibited the activity of the single tyrosine kinase domain. Finally, deletion of the pseudokinase domain of Jak2 deregulated signal transduction through the gamma interferon receptor by significantly increasing ligand-independent activation of Stat transcription factors. These results indicate that the pseudokinase domain neg. regulates the activity of Jak2, probably through an interaction with the kinase domain, and this regulation is required to keep Jak2 inactive in the absence of ligand stimulation. Furthermore, the pseudokinase domain may have a role in regulation of Jak2-substrate interactions.
- 6Damsky, W.; Peterson, D.; Ramseier, J.; Al-Bawardy, B.; Chun, H.; Proctor, D.; Strand, V.; Flavell, R. A.; King, B. The emerging role of Janus kinase inhibitors in the treatment of autoimmune and inflammatory diseases. J. Allergy Clin. Immunol. 2021, 147, 814– 826, DOI: 10.1016/j.jaci.2020.10.022Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1agurY%253D&md5=74561a7a05af19978f183215bdf4f5f6The emerging role of Janus kinase inhibitors in the treatment of autoimmune and inflammatory diseasesDamsky, William; Peterson, Danielle; Ramseier, Julie; Al-Bawardy, Badr; Chun, Hyung; Proctor, Deborah; Strand, Vibeke; Flavell, Richard A.; King, BrettJournal of Allergy and Clinical Immunology (2021), 147 (3), 814-826CODEN: JACIBY; ISSN:0091-6749. (Elsevier Inc.)A review. Autoimmune and inflammatory diseases are common and diverse, and they can affect nearly any organ system. Much of the pathogenesis of these diseases is related to dysregulated cytokine activity. Historically, autoimmune and inflammatory diseases have been treated with medications that nonspecifically suppress the immune system. mAbs that block the action of pathogenic cytokines emerged 2 decades ago and have become widely useful. More recently, agents that simultaneously block multiple pathogenic cytokines via inhibition of the downstream Janus kinase (JAK)-signal transducer and activator of transcription pathway have emerged and are becoming increasingly important. These small-mol. inhibitors, collectively termed JAK inhibitors, are US Food and Drug Administration-approved in a few autoimmune/inflammatory disorders and are being evaluated in many others. Here, we review the biol. of the JAK-signal transducer and activator of transcription pathway and the use of JAK inhibitors to treat autoimmune and inflammatory diseases across medical subspecialties.
- 7Jamilloux, Y.; El Jammal, T.; Vuitton, L.; Gerfaud-Valentin, M.; Kerever, S.; Sève, P. JAK inhibitors for the treatment of autoimmune and inflammatory diseases. Autoimmun. Rev. 2019, 18, 102390 DOI: 10.1016/j.autrev.2019.102390Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvFKhtb3P&md5=d31a5113949fdf1391c3d7ceeac3682eJAK inhibitors for the treatment of autoimmune and inflammatory diseasesJamilloux, Yvan; El Jammal, Thomas; Vuitton, Lucine; Gerfaud-Valentin, Mathieu; Kerever, Sebastien; Seve, PascalAutoimmunity Reviews (2019), 18 (11), 102390CODEN: ARUEBU; ISSN:1568-9972. (Elsevier B.V.)A review. Cytokines play a central role in the pathophysiol. of autoimmune and inflammatory diseases. Several cytokines signal through the JAK-STAT pathway, which is now recognized as a major target to inhibit the effect of a wide array of cytokines. JAK inhibitors are increasingly used in the setting of inflammatory and autoimmune diseases. While the currently approved drugs are panJAK inhibitors, more selective small mols. are being developed and tested in various rheumatic disorders. In this extensive review, we present evidence- or hypothesis-based perspectives for these drugs in various rheumatol. conditions, such as rheumatoid arthritis, systemic lupus erythematosus, giant cell arteritis, and autoinflammatory diseases.
- 8Schett, G.; Sticherling, M.; Neurath, M. F. COVID-19: risk for cytokine targeting in chronic inflammatory diseases?. Nat. Rev. Immunol. 2020, 20, 271– 272, DOI: 10.1038/s41577-020-0312-7Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXntFOrs7Y%253D&md5=f3f55b99938ede6632815c92168a61b9COVID-19: risk for cytokine targeting in chronic inflammatory diseases?Schett, Georg; Sticherling, Michael; Neurath, Markus F.Nature Reviews Immunology (2020), 20 (5), 271-272CODEN: NRIABX; ISSN:1474-1733. (Nature Research)COVID-19, caused by the SARS-CoV-2 virus, has become pandemic. With sharply rising infection rates, patient groups characterized by an enhanced infection risk will be challenged by the virus. In this context, patients with chronic immune-mediated inflammatory diseases are of particular interest, as these diseases are characterized by an intrinsic immune dysfunction leading to inflammation that may enhance risk for severe infection.
- 9Wu, D.; Yang, X. O. TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor Fedratinib. J. Microbiol. Immunol. Infect. 2020, 53, 368– 370, DOI: 10.1016/j.jmii.2020.03.005Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlsFalurc%253D&md5=bab4a2496a1381dd062fbaf5349baf57TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor FedratinibWu, Dandan; Yang, Xuexian O.Journal of Microbiology, Immunology and Infection (2020), 53 (3), 368-370CODEN: JMIIFG; ISSN:1995-9133. (Elsevier Taiwan LLC)COVID-19 emerges as a pandemic disease with high mortality. Development of effective prevention and treatment is an urgent need. We reviewed TH17 responses in patients with SARS-CoV-2 and proposed an FDA approved JAK2 inhibitor Fedratinib for reducing mortality of patients with TH17 type immune profiles.
- 10Spinelli, F. R.; Conti, F.; Gadina, M. HiJAKing SARS-CoV-2? The potential role of JAK inhibitors in the management of COVID-19. Sci. Immunol. 2020, 5, eabc5367 DOI: 10.1126/sciimmunol.abc5367Google ScholarThere is no corresponding record for this reference.
- 11Vázquez, J.; López, M.; Gibert, E.; Herrero, E.; Luque, F. J. Merging Ligand-Based and Structure-Based Methods in Drug Discovery: An Overview of Combined Virtual Screening Approaches. Molecules 2020, 25, 4723, DOI: 10.3390/molecules25204723Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1aitrnN&md5=50e00b66bc0191e4f1f076357ebe210fMerging ligand-based and structure-based methods in drug discovery: an overview of combined virtual screening approachesVazquez, Javier; Lopez, Manel; Gibert, Enric; Herrero, Enric; Luque, F. JavierMolecules (2020), 25 (20), 4723CODEN: MOLEFW; ISSN:1420-3049. (MDPI AG)A review. Virtual screening (VS) is an outstanding cornerstone in the drug discovery pipeline. A variety of computational approaches, which are generally classified as ligand-based (LB) and structure-based (SB) techniques, exploit key structural and physicochem. properties of ligands and targets to enable the screening of virtual libraries in the search of active compds. Though LB and SB methods have found widespread application in the discovery of novel drug-like candidates, their complementary natures have stimulated continued efforts toward the development of hybrid strategies that combine LB and SB techniques, integrating them in a holistic computational framework that exploits the available information of both ligand and target to enhance the success of drug discovery projects. In this review, we analyze the main strategies and concepts that have emerged in the last years for defining hybrid LB + SB computational schemes in VS studies. Particularly, attention is focused on the combination of mol. similarity and docking, illustrating them with selected applications taken from the literature.
- 12Lin, T. E.; HuangFu, W.; Chao, M.; Sung, T.; Chang, C.; Chen, Y.; Hsieh, J.; Tu, H.; Huang, H.; Pan, S.; Hsu, K.-C. A novel selective JAK2 inhibitor identified using pharmacological interactions. Front. Pharmacol. 2018, 9, 1379, DOI: 10.3389/fphar.2018.01379Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVGmtLzL&md5=deaab846ce59da3f6aebe14390be1a92A novel selective JAK2 inhibitor identified using pharmacological interactionsLin, Tony Eight; Huangfu, Wei-Chun; Chao, Min-Wu; Sung, Tzu-Ying; Chang, Chao-Di; Chen, Yi-Ying; Hsieh, Jui-Hua; Tu, Huang-Ju; Huang, Han-Li; Pan, Shiow-Lin; Hsu, Kai-ChengFrontiers in Pharmacology (2018), 9 (), 1379CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)The JAK2/STAT signaling pathway mediates cytokine receptor signals that are involved in cell growth, survival and homeostasis. JAK2 is a member of the Janus kinase (JAK) family and aberrant JAK2/STAT is involved with various diseases, making the pathway a therapeutic target. The similarity between the ATP binding site of protein kinases has made development of specific inhibitors difficult. Current JAK2 inhibitors are not selective and produce unwanted side effects. It is thought that increasing selectivity of kinase inhibitors may reduce the side effects seen with current treatment options. Thus, there is a great need for a selective JAK inhibitor. In this study, we identified a JAK2 specific inhibitor. We first identified key pharmacol. interactions in the JAK2 binding site by analyzing known JAK2 inhibitors. Then, we performed structure-based virtual screening and filtered compds. based on their pharmacol. interactions and identified compd. NSC13626 as a potential JAK2 inhibitor. Results of enzymic assays revealed that against a panel of kinases, compd. NSC13626 is a JAK2 inhibitor and has high selectivity toward the JAK2 and JAK3 isoenzymes. Our cellular assays revealed that compd. NSC13626 inhibits colorectal cancer cell (CRC) growth by downregulating phosphorylation of STAT3 and arresting the cell cycle in the S phase. Thus, we believe that compd. NSC13626 has potential to be further optimized as a selective JAK2 drug.
- 13Itteboina, R.; Ballu, S.; Sivan, S. K.; Manga, V. Molecular modeling-driven approach for identification of Janus kinase 1 inhibitors through 3D-QSAR, docking and molecular dynamics simulations. J. Recept. Signal Transduction 2017, 37, 453– 469, DOI: 10.1080/10799893.2017.1328442Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXosVansL0%253D&md5=1cb8a96420c1a6820326e5361adbc389Molecular modeling-driven approach for identification of Janus kinase 1 inhibitors through 3D-QSAR, docking and molecular dynamics simulationsItteboina, Ramesh; Ballu, Srilata; Sivan, Sree Kanth; Manga, VijjulathaJournal of Receptors and Signal Transduction (2017), 37 (5), 453-469CODEN: JRSTCT ISSN:. (Taylor & Francis Ltd.)Janus kinase 1 (JAK 1) belongs to the JAK family of intracellular nonreceptor tyrosine kinase. JAK-signal transducer and activator of transcription (JAK-STAT) pathway mediate signaling by cytokines, which control survival, proliferation and differentiation of a variety of cells. Three-dimensional quant. structure activity relationship (3 D-QSAR), mol. docking and mol. dynamics (MD) methods was carried out on a dataset of Janus kinase 1(JAK 1) inhibitors. Ligands were constructed and docked into the active site of protein using GLIDE 5.6. Best docked poses were selected after anal. for further 3 D-QSAR anal. using comparative mol. field anal. (CoMFA) and comparative mol. similarity indexes anal. (CoMSIA) methodol. Employing 60 mols. in the training set, 3 D-QSAR models were generate that showed good statistical reliability, which is clearly obsd. in terms of r2ncv and q2loo values. The predictive ability of these models was detd. using a test set of 25 mols. that gave acceptable predictive correlation (r2Pred) values. The key amino acid residues were identified by means of mol. docking, and the stability and rationality of the derived mol. conformations were also validated by MD simulation. The good consonance between the docking results and CoMFA/CoMSIA contour maps provides helpful clues about the reasonable modification of mols. in order to design more efficient JAK 1 inhibitors. The developed models are expected to provide some directives for further synthesis of highly effective JAK 1 inhibitors.
- 14Itteboina, R.; Ballu, S.; Sivan, S. K.; Manga, V. Molecular docking, 3D QSAR and dynamics simulation studies of imidazo-pyrrolopyridines as janus kinase 1 (JAK 1) inhibitors. Comput. Biol. Chem. 2016, 64, 33– 46, DOI: 10.1016/j.compbiolchem.2016.04.009Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xotlegu7Y%253D&md5=c90e4120727b9b4df112de83d5cabb93Molecular docking, 3D QSAR and dynamics simulation studies of imidazo-pyrrolopyridines as janus kinase 1 (JAK 1) inhibitorsItteboina, Ramesh; Ballu, Srilata; Sivan, Sree Kanth; Manga, VijjulathaComputational Biology and Chemistry (2016), 64 (), 33-46CODEN: CBCOCH; ISSN:1476-9271. (Elsevier B.V.)Janus kinase 1 (JAK 1) plays a crit. role in initiating responses to cytokines by the JAK-signal transducer and activator of transcription (JAK-STAT). This controls survival, proliferation and differentiation of a variety of cells. Docking, 3D quant. structure activity relationship (3D-QSAR) and mol. dynamics (MD) studies were performed on a series of Imidazo-pyrrolopyridine derivs. reported as JAK 1 inhibitors. QSAR model was generated using 30 mols. in the training set; developed model showed good statistical reliability, which is evident from r2ncv and r2loo values. The predictive ability of this model was detd. using a test set of 13 mols. that gave acceptable predictive correlation (r2Pred) values. Finally, mol. dynamics simulation was performed to validate docking results and MM/GBSA calcns. This facilitated us to compare binding free energies of cocrystal ligand and newly designed mol. R1. The good concordance between the docking results and CoMFA/CoMSIA contour maps afforded obliging clues for the rational modification of mols. to design more potent JAK 1 inhibitors.
- 15Sanachai, K.; Mahalapbutr, P.; Choowongkomon, K.; Poo-Arporn, R. P.; Wolschann, P.; Rungrotmongkol, T. Insights into the binding recognition and susceptibility of tofacitinib toward janus kinases. ACS Omega 2020, 5, 369– 377, DOI: 10.1021/acsomega.9b02800Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVCjtg%253D%253D&md5=de8290f477cc88ae016f67815bb9ee68Insights into the Binding Recognition and Susceptibility of Tofacitinib toward Janus KinasesSanachai, Kamonpan; Mahalapbutr, Panupong; Choowongkomon, Kiattawee; Poo-arporn, Rungtiva P.; Wolschann, Peter; Rungrotmongkol, ThanyadaACS Omega (2020), 5 (1), 369-377CODEN: ACSODF; ISSN:2470-1343. (American Chemical Society)Janus kinases (JAKs) are enzymes involved in signaling pathways that affect hematopoiesis and immune cell functions. JAK1, JAK2, and JAK3 play different roles in numerous diseases of the immune system and have also been considered as potential targets for cancer therapy. In the present study, the susceptibility of the oral JAK inhibitor tofacitinib against these three JAKs was elucidated using the 500-ns mol. dynamics (MD) simulations and free energy calcns. based on MM-PB(GB)SA, QM/MM-GBSA (PM3 and SCC-DFTB), and SIE methods. The obtained results revealed that tofacitinib could interact with all JAKs at the ATP-binding site via electrostatic attraction, hydrogen bond formation, and in particular van der Waals interaction. The conserved glutamate and leucine residues (E957 and L959 of JAK1, E930 and L932 of JAK2, and E903 and L905 of JAK3) located in the hinge region stabilized tofacitinib binding through strongly formed hydrogen bonds. Complexation with the incoming tofacitinib led to a closed conformation of the ATP-binding site and a decreased protein fluctuation at the glycine loop of the JAK protein. The binding affinities of tofacitinib/JAKs were ranked in the order of JAK3 > JAK2 ∼ JAK1, which are in line with the reported exptl. data.
- 16Clark, J. D.; Flanagan, M. E.; Telliez, J. Discovery and development of Janus Kinase (JAK) inhibitors for inflammatory diseases: Miniperspective. J. Med. Chem. 2014, 57, 5023– 5038, DOI: 10.1021/jm401490pGoogle Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVCjsA%253D%253D&md5=6763ae2d46f52ddcda23e235a60634aeDiscovery and Development of Janus Kinase (JAK) Inhibitors for Inflammatory DiseasesClark, James D.; Flanagan, Mark E.; Telliez, Jean-BaptisteJournal of Medicinal Chemistry (2014), 57 (12), 5023-5038CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The Janus kinases (JAKs) are a family of intracellular tyrosine kinases that play an essential role in the signaling of numerous cytokines that have been implicated in the pathogenesis of inflammatory diseases. As a consequence, the JAKs have received significant attention in recent years from the pharmaceutical and biotechnol. industries as therapeutic targets. Here, we provide a review of the JAK pathways, the structure, function, and activation of the JAK enzymes followed by a detailed look at the JAK inhibitors currently in the clinic or approved for these indications. Finally, a perspective is provided on what the past decade of research with JAK inhibitors for inflammatory indications has taught along with thoughts on what the future may hold in terms of addressing the opportunities and challenges that remain.
- 17Yao, T.; Xie, J.; Liu, X.; Cheng, J.; Zhu, C.; Zhao, J.; Dong, X. Integration of pharmacophore mapping and molecular docking in sequential virtual screening: towards the discovery of novel JAK2 inhibitors. RSC Adv. 2017, 7, 10353– 10360, DOI: 10.1039/C6RA24959KGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXit1Wgu7o%253D&md5=be2ddd7109bfb150b99d9d762b7e35b1Integration of pharmacophore mapping and molecular docking in sequential virtual screening: towards the discovery of novel JAK2 inhibitorsYao, Ting-Ting; Xie, Jiang-Feng; Liu, Xing-Guo; Cheng, Jing-Li; Zhu, Cheng-Yuan; Zhao, Jin-Hao; Dong, Xiao-WuRSC Advances (2017), 7 (17), 10353-10360CODEN: RSCACL; ISSN:2046-2069. (Royal Society of Chemistry)An integrated virtual screening protocol by combining mol. docking and pharmacophore mapping was established to identify novel inhibitors of JAK2 from a com. compd. database. Twelve novel and structurally diverse hits were selected and subjected to in vitro biol. tests, and three compds. (A5, A6 and A9) with remarkable JAK2 inhibitory activity were identified. Then, the obtained structures were further used as the template for a subsequent similarity search, leading to the identification of another two promising compds. (B2 and B4). Selectivity profiles of JAK subtype and in vitro anti-cancer activity of the promising compds. were studied, revealing the promising compd. B2 was of interest for further study because of its JAK2 selective profile, novelty of skeleton and significantly anti-proliferative effect against cancer cells. Finally, binding patterns of the compds. A5 and B2 were explored to provide a deeper insight for further structural optimization.
- 18Galvez, J.; Galvez-Llompart, M.; Garcia-Domenech, R. Introduction to molecular topology: basic concepts and application to drug design. Curr. Comput.-Aided Drug Des. 2012, 8, 196– 223, DOI: 10.2174/157340912801619094Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlKqu7nP&md5=6a6834004023ce4245631c184e9c34a6Introduction to molecular topology: basic concepts and application to drug designGalvez, Jorge; Galvez-Llompart, Maria; Garcia-Domenech, RamonCurrent Computer-Aided Drug Design (2012), 8 (3), 196-223CODEN: CCDDAS; ISSN:1573-4099. (Bentham Science Publishers Ltd.)In this review it is dealt the use of mol. topol. (MT) in the selection and design of new drugs. After an introduction of the actual methods used for drug design, the basic concepts of MT are defined, including examples of calcn. of topol. indexes, which are numerical descriptors of mol. structures. The goal is making this calcn. familiar to the potential students and allowing a straightforward comprehension of the topic. Finally, the achievements obtained in this field are detailed, so that the reader can figure out the great interest of this approach.
- 19Zanni, R.; Galvez-Llompart, M.; Garcia-Domenech, R.; Galvez, J. What place does molecular topology have in today’s drug discovery?. Expert Opin. Drug Discovery 2020, 15, 1133– 1144, DOI: 10.1080/17460441.2020.1770223Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVWgtLnN&md5=4644090178dc2b3028f4298869c8fb3dWhat place does molecular topology have in today's drug discoveryZanni, Riccardo; Galvez-Llompart, Maria; Garcia-Domenech, Ramon; Galvez, JorgeExpert Opinion on Drug Discovery (2020), 15 (10), 1133-1144CODEN: EODDBX; ISSN:1746-0441. (Taylor & Francis Ltd.)Introduction Most methods in mol. and drug design are currently based on physicochem. descriptors. However, mol. topol., which relies on topol. descriptors, has also shown value for mol. design even if it does not take into account the phys. or chem. properties of ligands and receptors, including the ligand-receptor interaction itself. Areas covered Herein, the authors provide new insights into the importance of mol. topol. according to some of the latest discoveries in physics and chem. Furthermore, the authors report on the most significant achievements in drug design using mol. topol. over the last 5 years and give their expert perspectives on the subject as a whole. Expert opinionMol. topol. is a new paradigm which is independent of physicochem. mol. descriptors. This fact explains the viability of both the discovery of new lead compds. with a min. of information derived from math.-topol. patterns and the interpretation results in structural and physicochem. terms.
- 20Zak, M.; Hurley, C. A.; Ward, S. I.; Bergeron, P.; Barrett, K.; Balazs, M.; Blair, W. S.; Bull, R.; Chakravarty, P.; Chang, C.; Crackett, P.; Deshmukh, G.; DeVoss, J.; Dragovich, P. S.; Eigenbrot, C.; Ellwood, C.; Gaines, S.; Ghilardi, N.; Gibbons, P.; Gradl, S.; Gribling, P.; Hamman, C.; Harstad, E.; Hewitt, P.; Johnson, A.; Johnson, T.; Kenny, J. R.; Koehler, M. F.; Bir Kohli, P.; Labadie, S.; Lee, W. P.; Liao, J.; Liimatta, M.; Mendonca, R.; Narukulla, R.; Pulk, R.; Reeve, A.; Savage, S.; Shia, S.; Steffek, M.; Ubhayakar, S.; van Abbema, A.; Aliagas, I.; Avitabile-Woo, B.; Xiao, Y.; Yang, J.; Kulagowski, J. J. Identification of C-2 hydroxyethyl imidazopyrrolopyridines as potent JAK1 inhibitors with favorable physicochemical properties and high selectivity over JAK2. J. Med. Chem. 2013, 56, 4764– 4785, DOI: 10.1021/jm4004895Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXnsVWit74%253D&md5=0b0d1950adebf325cc434344f57ca139Identification of C-2 Hydroxyethyl Imidazopyrrolopyridines as Potent JAK1 Inhibitors with Favorable Physicochemical Properties and High Selectivity over JAK2Zak, Mark; Hurley, Christopher A.; Ward, Stuart I.; Bergeron, Philippe; Barrett, Kathy; Balazs, Mercedesz; Blair, Wade S.; Bull, Richard; Chakravarty, Paroma; Chang, Christine; Crackett, Peter; Deshmukh, Gauri; DeVoss, Jason; Dragovich, Peter S.; Eigenbrot, Charles; Ellwood, Charles; Gaines, Simon; Ghilardi, Nico; Gibbons, Paul; Gradl, Stefan; Gribling, Peter; Hamman, Chris; Harstad, Eric; Hewitt, Peter; Johnson, Adam; Johnson, Tony; Kenny, Jane R.; Koehler, Michael F. T.; Bir Kohli, Pawan; Labadie, Sharada; Lee, Wyne P.; Liao, Jiangpeng; Liimatta, Marya; Mendonca, Rohan; Narukulla, Raman; Pulk, Rebecca; Reeve, Austin; Savage, Scott; Shia, Steven; Steffek, Micah; Ubhayakar, Savita; van Abbema, Anne; Aliagas, Ignacio; Avitabile-Woo, Barbara; Xiao, Yisong; Yang, Jing; Kulagowski, Janusz J.Journal of Medicinal Chemistry (2013), 56 (11), 4764-4785CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Herein we report on the structure-based discovery of a C-2 hydroxyethyl moiety which provided consistently high levels of selectivity for JAK1 over JAK2 to the imidazopyrrolopyridine series of JAK1 inhibitors. X-ray structures of a C-2 hydroxyethyl analog in complex with both JAK1 and JAK2 revealed differential ligand/protein interactions between the two isoforms and offered an explanation for the obsd. selectivity. Anal. of historical data from related mols. was used to develop a set of physicochem. compd. design parameters to impart desirable properties such as acceptable membrane permeability, potent whole blood activity, and a high degree of metabolic stability. This work culminated in the identification of a highly JAK1 selective compd. (31) exhibiting favorable oral bioavailability across a range of preclin. species and robust efficacy in a rat CIA model.
- 21Hart, A. C.; Schroeder, G. M.; Wan, H.; Grebinski, J.; Inghrim, J.; Kempson, J.; Guo, J.; Pitts, W. J.; Tokarski, J. S.; Sack, J. S.; Khan, J. A.; Lippy, J.; Lorenzi, M. V.; You, D.; McDevitt, T.; Vuppugalla, R.; Zhang, Y.; Lombardo, L. J.; Trainor, G. L.; Purandare, A. V. Structure-Based Design of Selective Janus Kinase 2 Imidazo[4,5-d]pyrrolo[2,3-b]pyridine Inhibitors. ACS Med. Chem. Lett. 2015, 6, 845– 849, DOI: 10.1021/acsmedchemlett.5b00225Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFCrurvP&md5=c9b4e51332496a1b29cc8f4c187d488aStructure-Based Design of Selective Janus Kinase 2 Imidazo[4,5-d]pyrrolo[2,3-b]pyridine InhibitorsHart, Amy C.; Schroeder, Gretchen M.; Wan, Honghe; Grebinski, James; Inghrim, Jennifer; Kempson, James; Guo, Junqing; Pitts, William J.; Tokarski, John S.; Sack, John S.; Khan, Javed A.; Lippy, Jonathan; Lorenzi, Matthew V.; You, Dan; McDevitt, Theresa; Vuppugalla, Ragini; Zhang, Yueping; Lombardo, Louis J.; Trainor, George L.; Purandare, Ashok V.ACS Medicinal Chemistry Letters (2015), 6 (8), 845-849CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Early hit to lead work on a pyrrolopyridine chemotype provided access to compds. with biochem. and cellular potency against Janus kinase 2 (JAK2). Structure-based drug design along the extended hinge region of JAK2 led to the identification of an important H-bond interaction with the side chain of Tyr 931, which improved JAK family selectivity. The 4,5-di-Me thiazole analog I demonstrated high levels of JAK family selectivity and was identified as a promising lead for the program.
- 22Forster, M.; Chaikuad, A.; Dimitrov, T.; Döring, E.; Holstein, J.; Berger, B.-T.; Gehringer, M.; Ghoreschi, K.; Müller, S.; Knapp, S.; Laufer, S. A. Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4- d]pyrrolo[2,3- b]pyridine Scaffold. J. Med. Chem. 2018, 61, 5350– 5366, DOI: 10.1021/acs.jmedchem.8b00571Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVGjsrbN&md5=8acdec137df746f854bbee75a10d5f70Development, Optimization, and Structure-Activity Relationships of Covalent-Reversible JAK3 Inhibitors Based on a Tricyclic Imidazo[5,4-d]pyrrolo[2,3-b]pyridine ScaffoldForster, Michael; Chaikuad, Apirat; Dimitrov, Teodor; Doering, Eva; Holstein, Julia; Berger, Benedict-Tilman; Gehringer, Matthias; Ghoreschi, Kamran; Mueller, Susanne; Knapp, Stefan; Laufer, Stefan A.Journal of Medicinal Chemistry (2018), 61 (12), 5350-5366CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Janus kinases are major drivers of immune signaling and have been the focus of anti-inflammatory drug discovery for more than a decade. Because of the invariable colocalization of JAK1 and JAK3 at cytokine receptors, the question if selective JAK3 inhibition is sufficient to effectively block downstream signaling has been highly controversial. Recently, we discovered the covalent-reversible JAK3 inhibitor FM-381 (23) featuring high isoform and kinome selectivity. Crystallog. revealed that this inhibitor induces an unprecedented binding pocket by interactions of a nitrile substituent with arginine residues in JAK3. Herein, we describe detailed structure-activity relationships necessary for induction of the arginine pocket and the impact of this structural change on potency, isoform selectivity, and efficacy in cellular models. Furthermore, we evaluated the stability of this novel inhibitor class in in vitro metabolic assays and were able to demonstrate an adequate stability of key compd. 23 for in vivo use.
- 23De Vivo, M.; Masetti, M.; Bottegoni, G.; Cavalli, A. Role of Molecular Dynamics and Related Methods in Drug Discovery. J. Med. Chem. 2016, 59, 4035– 4061, DOI: 10.1021/acs.jmedchem.5b01684Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlWksLs%253D&md5=b3d8a4fb5705a3555c2bb2a962420396Role of Molecular Dynamics and Related Methods in Drug DiscoveryDe Vivo, Marco; Masetti, Matteo; Bottegoni, Giovanni; Cavalli, AndreaJournal of Medicinal Chemistry (2016), 59 (9), 4035-4061CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mol. dynamics (MD) and related methods are close to becoming routine computational tools for drug discovery. Their main advantage is in explicitly treating structural flexibility and entropic effects. This allows a more accurate est. of the thermodn. and kinetics assocd. with drug-target recognition and binding, as better algorithms and hardware architectures increase their use. Here, we review the theor. background of MD and enhanced sampling methods, focusing on free-energy perturbation, metadynamics, steered MD, and other methods most consistently used to study drug-target binding. We discuss unbiased MD simulations that nowadays allow the observation of unsupervised ligand-target binding, assessing how these approaches help optimizing target affinity and drug residence time toward improved drug efficacy. Further issues discussed include allosteric modulation and the role of water mols. in ligand binding and optimization. We conclude by calling for more prospective studies to attest to these methods' utility in discovering novel drug candidates.
- 24Guterres, H.; Im, W. Improving Protein-Ligand Docking Results with High-Throughput Molecular Dynamics Simulations. J. Chem. Inf. Model. 2020, 60, 2189– 2198, DOI: 10.1021/acs.jcim.0c00057Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFCju7g%253D&md5=3c5d10e0584d0218071dc8d0a37179e7Improving Protein-Ligand Docking Results with High-Throughput Molecular Dynamics SimulationsGuterres, Hugo; Im, WonpilJournal of Chemical Information and Modeling (2020), 60 (4), 2189-2198CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Structure-based virtual screening relies on classical scoring functions that often fail to reliably discriminate binders from nonbinders. In this work, the authors present a high-throughput protein-ligand complex mol. dynamics (MD) simulation that uses the output from AutoDock Vina to improve docking results in distinguishing active from decoy ligands in a directory of useful decoy-enhanced (DUD-E) dataset. MD trajectories are processed by evaluating ligand-binding stability using root-mean-square deviations. The authors select 56 protein targets (of 7 different protein classes) and 560 ligands (280 actives, 280 decoys) and show 22% improvement in ROC AUC (area under the curve, receiver operating characteristics curve), from an initial value of 0.68 (AutoDock Vina) to a final value of 0.83. The MD simulation demonstrates a robust performance across all seven different protein classes. In addn., some predicted ligand-binding modes are moderately refined during MD simulations. These results systematically validate the reliability of a physics-based approach to evaluate protein-ligand binding interactions.
- 25Gioia, D.; Bertazzo, M.; Recanatini, M.; Masetti, M.; Cavalli, A. Dynamic docking: A paradigm shift in computational drug discovery. Molecules 2017, 22, 2029, DOI: 10.3390/molecules22112029Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitl2jtLk%253D&md5=b835af2568bb7f5b0500d6739523ad4eDynamic docking: a paradigm shift in computational drug discoveryGioia, Dario; Bertazzo, Martina; Recanatini, Maurizio; Masetti, Matteo; Cavalli, AndreaMolecules (2017), 22 (11), 2029/1-2029/21CODEN: MOLEFW; ISSN:1420-3049. (MDPI AG)Mol. docking is the methodol. of choice for studying in silico protein-ligand binding and for prioritizing compds. to discover new lead candidates. Traditional docking simulations suffer from major limitations, mostly related to the static or semi-flexible treatment of ligands and targets. They also neglect solvation and entropic effects, which strongly limits their predictive power. During the last decade, methods based on full atomistic mol. dynamics (MD) have emerged as a valid alternative for simulating macromol. complexes. In principle, compared to traditional docking, MD allows the full exploration of drug-target recognition and binding from both the mechanistic and energetic points of view (dynamic docking). Binding and unbinding kinetic consts. can also be detd. While dynamic docking is still too computationally expensive to be routinely used in fast-paced drug discovery programs, the advent of faster computing architectures and advanced simulation methodologies are changing this scenario. It is feasible that dynamic docking will replace static docking approaches in the near future, leading to a major paradigm shift in in silico drug discovery. Against this background, we review the key achievements that have paved the way for this progress.
- 26Selleck. https://www.selleckchem.com/ Last access: April 2021.Google ScholarThere is no corresponding record for this reference.
- 27BioCrick. https://www.biocrick.com/LY2784544-BCC2200.html Last access: April 2021.Google ScholarThere is no corresponding record for this reference.
- 28Boc-Science. https://www.bocsci.com Last access: April 2021.Google ScholarThere is no corresponding record for this reference.
- 29Alvascience Srl, alvaDesc (software for molecular descriptors calculation), 2019, https://www.alvascience.com.Google ScholarThere is no corresponding record for this reference.
- 30O’Shea, J. J.; Kontzias, A.; Yamaoka, K.; Tanaka, Y.; Laurence, A. Janus kinase inhibitors in autoimmune diseases. Ann. Rheum. Dis. 2013, 72, ii111– ii115, DOI: 10.1136/annrheumdis-2012-202576Google ScholarThere is no corresponding record for this reference.
- 31Kubler, P. Janus kinase inhibitors: Mechanisms of action. Aust. Prescr. 2014, 37, 154– 157, DOI: 10.18773/austprescr.2014.061Google ScholarThere is no corresponding record for this reference.
- 32Roskoski, R., Jr. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol. Res. 2016, 111, 784– 803, DOI: 10.1016/j.phrs.2016.07.038Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht12jurzJ&md5=a45fba1bb110eff4d5c1b31b78c1b3acJanus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseasesRoskoski, Robert, Jr.Pharmacological Research (2016), 111 (), 784-803CODEN: PHMREP; ISSN:1043-6618. (Elsevier Ltd.)The Janus kinase (JAK) family of non-receptor protein-tyrosine kinases consists of JAK1, JAK2, JAK3, and TYK2 (tyrosine kinase-2). Each of these proteins contains a JAK homol. pseudokinase (JH2) domain that regulates the adjacent protein kinase domain (JH1). JAK1/2 and TYK2 are ubiquitously expressed whereas JAK3 is found predominantly in hematopoietic cells. The Janus kinase family is regulated by numerous cytokines including interleukins, interferons, and hormones such as erythropoietin, thrombopoietin, and growth hormone. Ligand binding to cytokine and hormone receptors leads to the activation of assocd. Janus kinases, which then mediate the phosphorylation of the receptors. The SH2 domain of STATs (signal transducers and activators of transcription) binds to the receptor phosphotyrosines thereby promoting STAT phosphorylation by the Janus kinases and consequent activation. STAT dimers are translocated to the nucleus where they participate in the regulation of the expression of thousands of proteins. JAK-STAT dysregulation results in autoimmune disorders such as rheumatoid arthritis, ulcerative colitis, and Crohn disease. JAK-STAT dysregulation also plays a role in the pathogenesis of myelofibrosis, polycythemia vera, and other myeloproliferative illnesses. An activating JAK2 V617F mutation occurs in 95% of people with polycythemia vera and in a lower percentage of people with other neoplasms. JAK1/3 signaling participates in the pathogenesis of inflammatory afflictions while JAK1/2 signaling participates in the development of several malignancies including leukemias and lymphomas as well as myeloproliferative neoplasms. Tofacitinib is a pan-JAK inhibitor that is approved by the FDA for the treatment of rheumatoid arthritis and ruxolitinib is a JAK1/2 inhibitor that is approved for the treatment of polycythemia vera and myelofibrosis.
- 33Tocris DB. https://www.tocris.com/. Last access: January 2019.Google ScholarThere is no corresponding record for this reference.
- 34ABCAM. www.abcam.com. Last access: January 2019.Google ScholarThere is no corresponding record for this reference.
- 35Hastie, T.; Tibshirani, R.; Friedman, J. The elements of statistical learning: data mining, inference, and prediction; Springer Science & Business Media, 2009.Google ScholarThere is no corresponding record for this reference.
- 36StatSoft IS, version 9.0; Tulsa, OK, USA, 2009.Google ScholarThere is no corresponding record for this reference.
- 37Gálvez, J.; García-Domenech, R.; de Gregorio Alapont, C.; de Julián-Ortiz, J. V.; Popa, L. Pharmacological distribution diagrams: a tool for de novo drug design. J. Mol. Graph. 1996, 14, 272– 276, DOI: 10.1016/S0263-7855(96)00081-1Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXitlCrur8%253D&md5=84cce553319aa39f1d2c349b0338c54ePharmacological distribution diagrams: a tool for de novo drug designGalvez, J.; Garcia-Domenech, R.; de Gregorio Alapont, C.; de Julian-Ortiz, J. V.; Popa, L.Journal of Molecular Graphics (1996), 14 (5), 272-276CODEN: JMGRDV; ISSN:0263-7855. (Elsevier)Discriminant anal. applied to SAR studies using topol. descriptors allows us to plot frequency distribution diagrams: a function of the no. of drugs within an interval of values of discriminant function vs. these values. We make use of these representations, pharmacol. distribution diagrams (PDDs), in structurally heterogeneous groups where generally they adopt skewed Gaussian shapes or present several maxima. The maxima afford intervals of discriminant function in which exists a good expectancy to find new active drugs. A set of β-blockers with contrasted activity has been selected to test the ability of PPDs as a visualizing technique, for the identification of new β-blocker active compds.
- 38Gramatica, P. Principles of QSAR models validation: internal and external. QSAR Comb. Sci. 2007, 26, 694– 701, DOI: 10.1002/qsar.200610151Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtFKjtrs%253D&md5=05af99ce1a70f5f14ab8a645fbaf00d2Principles of QSAR models validation: internal and externalGramatica, PaolaQSAR & Combinatorial Science (2007), 26 (5), 694-701CODEN: QCSSAU; ISSN:1611-020X. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The recent REACH Policy of the European Union has led to scientists and regulators to focus their attention on establishing general validation principles for QSAR models in the context of chem. regulation (previously known as the Setubal, nowadays, the OECD principles). This paper gives a brief anal. of some principles: unambiguous algorithm, Applicability Domain (AD), and statistical validation. Some concerns related to QSAR algorithm reproducibility and an example of a fast check of the applicability domain for MLR models are presented. Common myths and misconceptions related to popular techniques for verifying internal predictivity, particularly for MLR models (for instance cross-validation, bootstrap), are commented on and compared with commonly used statistical techniques for external validation. The differences in the two validating approaches are highlighted, and evidence is presented that only models that were validated externally, after their internal validation, can be considered reliable and applicable for both external prediction and regulatory purposes.
- 39Schrodinger, L. Schrodinger software suite; Schrödinger, LLC: New York, 2019, p 670.Google ScholarThere is no corresponding record for this reference.
- 40Mosmann, T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55– 63, DOI: 10.1016/0022-1759(83)90303-4Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL2c%252FovFSmtw%253D%253D&md5=c1116f4ca7a7d2b79fab2a0bd2c2d439Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assaysMosmann TJournal of immunological methods (1983), 65 (1-2), 55-63 ISSN:0022-1759.A tetrazolium salt has been used to develop a quantitative colorimetric assay for mammalian cell survival and proliferation. The assay detects living, but not dead cells and the signal generated is dependent on the degree of activation of the cells. This method can therefore be used to measure cytotoxicity, proliferation or activation. The results can be read on a multiwell scanning spectrophotometer (ELISA reader) and show a high degree of precision. No washing steps are used in the assay. The main advantages of the colorimetric assay are its rapidity and precision, and the lack of any radioisotope. We have used the assay to measure proliferative lymphokines, mitogen stimulations and complement-mediated lysis.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.0c01468.
Descriptor values, classification of compounds and probability of activity for all data sets of models 1–4 (Tables S1–S5); leave-some-out validation test for DF2-4 (Tables S6–S8); docking score values from potential JAK inhibitors selected by molecular topology (Table S9); cross-docking analysis for JAK1, JAK2, and JAK3 subtypes (Figure S1); similarity-cluster analysis dendrogram (Figure S2) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.