Characterization of 2,4-Dianilinopyrimidines Against Five P. falciparum Kinases PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKBClick to copy article linkArticle link copied!
- Han Wee OngHan Wee OngStructural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesMore by Han Wee Ong
- Chandi de SilvaChandi de SilvaLuceome Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United StatesMore by Chandi de Silva
- Krisha AvalaniKrisha AvalaniLuceome Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United StatesMore by Krisha Avalani
- Frank KwarcinskiFrank KwarcinskiLuceome Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United StatesMore by Frank Kwarcinski
- Christopher R. MansfieldChristopher R. MansfieldDepartment of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, North Carolina 27710, United StatesMore by Christopher R. Mansfield
- Michael ChirgwinMichael ChirgwinDepartment of Chemistry, Duke University, 124 Science Drive, Durham, North Carolina 27708, United StatesMore by Michael Chirgwin
- Anna TruongAnna TruongDepartment of Chemistry, Duke University, 124 Science Drive, Durham, North Carolina 27708, United StatesMore by Anna Truong
- Emily R. Derbyshire*Emily R. Derbyshire*E.R.D.: email, [email protected]Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, North Carolina 27710, United StatesDepartment of Chemistry, Duke University, 124 Science Drive, Durham, North Carolina 27708, United StatesMore by Emily R. Derbyshire
- Reena Zutshi*Reena Zutshi*R.Z.: email, [email protected]Luceome Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United StatesMore by Reena Zutshi
- David H. Drewry*David H. Drewry*D.H.D.: email, [email protected]Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesLineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesMore by David H. Drewry
Abstract
Plasmodium kinases are increasingly recognized as potential novel antiplasmodial targets for the treatment of malaria, but only a small subset of these kinases have had structure–activity relationship (SAR) campaigns reported. Herein we report the discovery of CZC-54252 (1) as an inhibitor of five P. falciparum kinases PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB. 39 analogues were evaluated against all five kinases to establish SAR at three regions of the kinase active site. Nanomolar inhibitors of each kinase were discovered. We identified common and divergent SAR trends across all five kinases, highlighting substituents in each region that improve potency and selectivity for each kinase. Potent analogues were evaluated against the P. falciparum blood stage. Eight submicromolar inhibitors were discovered, of which 37 demonstrated potent antiplasmodial activity (EC50 = 0.16 μM). Our results provide an understanding of features needed to inhibit each individual kinase and lay groundwork for future optimization efforts toward novel antimalarials.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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 within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*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.
Malaria is a devastating disease caused by Plasmodium infection, with 247 million cases and 619 thousand deaths globally in 2021. (1) The rising resistance toward existing antimalarials is worrying. (1) New medicines with novel mechanisms of action are needed to address this evolving challenge. Plasmodium protein kinases present an untapped opportunity for drug development. (2−5) Of the 85–99 P. falciparum protein kinases, (6,7) 36–40 have been identified as essential for the asexual blood stage of P. falciparum. (8,9) Yet, most Plasmodium protein kinases have had few medicinal chemistry campaigns initiated against them to facilitate inhibitor development. To address this gap, we aimed to discover hits against lesser-studied Plasmodium kinases and understand critical molecular features to guide the inhibitor development of Plasmodium kinases.
Pyrimidines have been recognized as common hinge-binding groups of kinase inhibitors. (10,11) 2,4-Dianilinopyrimidines are among the hits in the Tres Cantos Antimalarial Set (TCAMS), a set of 13 533 inhibitors of P. falciparum asexual reproduction. (12) Screening TCAMS against five P. falciparum kinases led to the identification of PfCDPK1, PfCDPK4, and PfPK6 as putative targets of 2,4-dianilinopyrimidines. (13) Medicinal chemistry campaigns of 2,4-dianilinopyrimidines have been reported in the literature, optimizing for antiplasmodial activity by phenotypic screening. (14,15) The authors suggested that this series of inhibitors may target the Plasmodium CDPK and NEK families. (15) Recently, six human-PLK1-targeted 2,4-diarylaminopyrimidine inhibitors have also demonstrated antiplasmodial activity, of which two of them demonstrate inhibition of PfCDPK2, PfNEK3, and PfPKB. (16)
The 2,4-dianilinopyrimidines CZC-54252 (1) and CZC-25146 (2) (Figure 1A) were developed as human LRRK2 inhibitors. (17) During our screening efforts to discover novel kinase-targeted antiplasmodials, we found 1 to reduce parasite viability of P. falciparum 3D7 (Pf3D7) blood stage to 21% of the negative control at 1 μM. Follow-up experiments confirmed dose-dependent reduction of parasite viability with a EC50 of 0.34 μM. As far as we are aware, no information about inhibition of Plasmodium kinases have been reported for 1. We screened 1 against 11 P. falciparum kinases using the KinaseSeeker assay (18) at 1 μM. Five kinase targets were identified and demonstrated dose-dependent inhibition: PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB (Table 1). Of these five targets, 1 was the most potent against PfPKB with an IC50 of 70 nM. Interestingly, while 2,4-dianilinopyrimidines were proposed to target PfPK6, (13) we did not observe inhibition of PfPK6 by 1. We also did not observe significant inhibition of CDPK family members PfCDPK2 and PfCDPK5 and NEK family member PfNEK1.
kinase | % activity remaining at 1 μMa | IC50 (μM)b |
---|---|---|
PfARK1 | 53 | 3.4 |
PfARK3 | 50 | 1.8 |
PfCDPK2 | 78 | |
PfCDPK5 | 97 | |
PfGSK3 | 94 | |
PfNEK1 | 85 | |
PfNEK3 | 34 | 1.0 |
PfPK5 | 90 | |
PfPK6 | 90 | |
PfPK9 | 43 | 1.2 |
PfPKB | 11 | 0.070 |
Mean values from duplicate experiments using the KinaseSeeker assay.
Mean values from two experiments using the KinaseSeeker assay performed in duplicate.
PfARK1 (PF3D7_0605300) and PfARK3 (PF3D7_1356800) are Aurora kinases suggested to be involved in nuclear division processes. (19,20) Genetic studies have indicated that PfARK1 is essential for asexual blood stage of P. falciparum. (8,9,19) In contrast, PfARK3 is genetically validated to be essential by reverse genetics (8) but not by saturation mutagenesis. (9) No inhibitor of either kinase has been confirmed in the literature, although inhibitors of human Aurora kinases have demonstrated antiplasmodial activity. (21,22) PfARK1 mutations confer resistance to the human Aurora kinase inhibitor hesperadin (Figure 1B), suggesting that PfARK1 may be its target. (22)
PfNEK3 (PF3D7_1201600) belongs to the NEK family. (23) Separate studies have regarded PfNEK3 as either dispensable for the asexual blood stage proliferation (8) or that it could be disrupted, albeit with a fitness penalty. (9) PfNEK3 participates in an atypical MAPK signaling cascade by upregulating the activity of PfMAP2, (23,24) although the implications of this signaling pathway are not currently clear. Human PLK1 inhibitors including BI-2536 (Figure 1B) have been found to inhibit PfNEK3. (16) We have also previously identified PfNEK3 as one of the secondary targets of our potent PfPK6-targeted type II inhibitors. (25)
PfPK9 (PF3D7_1315100) is an orphan kinase that does not cluster with any typical kinase group (26) and is genetically validated to be essential for asexual blood stage proliferation. (8,9) PfPK9 regulates the activity of PfUBC13, (26,27) an essential E2 ubiquitin-conjugating enzyme involved with DNA replication and repair. (28) The only known inhibitors of PfPK9 are the human TAK1 inhibitor Takinib (Figure 1B) and analogues. (27)
PfPKB (PF3D7_1246900) is a kinase from the AGC group. (29) While a reverse genetics study found PfPKB to be essential, (8) it was found to be dispensable by saturation mutagenesis. (9) PfPKB is implicated in regulation of merozoite invasion into erythrocytes by phosphorylating PfGAP45. (30−32) Inhibition of PfPKB by Go 6983 or A-443654 (Figure 1B) decreases parasitemia and formation of new rings in the subsequent round of invasion, consistent with its proposed role in regulating invasion. (29,33) PfPKB has also been determined to be a secondary target of the above-mentioned 2,4-diarylaminopyrimidine inhibitors (16) and our type II inhibitors. (25)
Given the limited medicinal chemistry studies and inhibitor development for PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB, we aim to expand on the body of knowledge to facilitate future inhibitor development for these five diverse kinases (only 19.5–33.7% pairwise sequence identity in kinase domains; see Supporting Information (SI), Figure S1) across the Plasmodium kinome. These efforts will impact chemical probe development to resolve Plasmodium kinase function as well as the design of novel antimalarial agents. Herein, we report the results of our structure–activity relationship (SAR) study for inhibition of these five kinases by inhibitors with the 2,4-dianilinopyrimidine scaffold.
As crystal structures of 1 with any Plasmodium kinase are unavailable, we docked 1 into the structures of PfARK1, PfNEK3, PfPK9, and PfPKB predicted by AlphaFold (34,35) (docking was not performed for PfARK3 because its structure was unavailable) (Figure 2, and SI, Figures S2 and S3). As expected based on cocrystal structures of other 2,4-dianilinopyrimidines with kinases, the model suggests that 1 forms two hydrogen bonds with the peptide backbone of the outer hinge residue (hinge.46 by KLIFS notation (36)) using the N1 of the pyrimidine and exocyclic NH at the 2-position. The 2-position aryl substituent extends toward the solvent, with the morpholine ring exposed to solvent. The model also suggests that the methylsulfonamide group of 1 forms a hydrogen bond with the exocyclic NH at the 4-position of the pyrimidine and with the catalytic lysine.
The 5-position group of the pyrimidines binds close to the gatekeeper residue of the kinase, a residue that is often targeted for potency and specificity. Apart from steric complementarity, 5-position groups on pyrimidines may form specific interactions with gatekeeper residues, such as hydrogen bonding, (37) halogen/chalcogen bonds, (38,39) or lone pair−π interactions. (40) We thus first investigated the effect of substituting the 5-position chloro group of 1 with other groups (Table 2).
Mean values from duplicate experiments using the KinaseSeeker assay.
Mean values from two experiments using the KinaseSeeker assay performed in duplicate.
Both removal of the chlorine atom (3) or introduction of an endocyclic N atom at this position, converting the pyrimidine to a 1,3,5-triazine (20), abolished activity for all five kinases, suggesting that the chlorine makes key interactions with each kinase. Replacement with fluorine (2) recovered some activity only for PfPKB (IC50 = 1.5 μM) and PfNEK3 (68% activity remaining at 1 μM). Replacement with the heavier halogen bromine (4) or iodine (5) maintained the potencies for PfARK1, PfARK3, and PfPKB, while a modest 2.3-fold improvement in PfNEK3 potency was observed for both analogues. Interestingly, PfPK9 saw a 4-fold improvement in potency with 4 and a 17-fold improvement with 5. Generally, an improvement in potency was observed with increasing halogen size, suggesting that there are favorable hydrophobic or halogen-bonding interactions with heavier halogens. Looking closer into this general trend, we observed that this effect plateaued at chlorine for PfARK1, PfARK3, and PfPKB, and at bromine for PfNEK3, while it continued to iodine for PfPK9. This trend is independent of their gatekeeper residues (methionine for PfARK1 and PfPK9, leucine for PfARK3, PfNEK3, and PfPKB), suggesting that the origin of this varied sensitivity to halogens is more complex than could be simply predicted based on these residues.
The methyl (6) and ethyl (7) groups both abolished PfARK1, PfARK3, and PfPK9 potency but were tolerated by PfNEK3 and PfPKB. Further increase in steric size of the alkyl group to a cyclopropyl ring (8) was tolerated by PfPKB but abolished PfNEK3 activity. Among the five kinases, 8 is exquisitely selective for PfPKB. Interestingly, the trifluoromethyl group (9) is potent against all five kinases, improving potency, relative to 1, by PfARK1 3-fold, PfNEK3 2-fold, PfPK9 16-fold, and PfPKB 5-fold. PfARK3 was unaffected. Excitingly, 9 is the most potent PfPKB inhibitor reported (IC50 = 13 nM).
Similar to the alkyl groups, a methoxy group (10) or a dimethylamino group (11) were not tolerated by PfARK1, PfARK3, and PfPK9. Comparing with 7, with a similar-sized ethyl group, <2-fold change in PfNEK3 IC50, but a 4-fold drop in PfPKB potency with 10 was observed. In contrast, we observed a 3-fold drop in PfNEK3 potency with <2-fold change in PfPKB potency with 11. Both hydroxyl (12) and amino (13) groups completely abolished activity on all five kinases, suggesting that there is a lack of productive hydrogen bonding partners to accommodate these hydrogen bond donors here.
Surprisingly, the nitrile group (14) was not tolerated by PfARK1, PfARK3, and PfPK9, slightly disfavored by PfPKB (2.6-fold drop in potency compared to 1), but strongly preferred by PfNEK3 (7-fold improvement in potency over 1). This highlights a possible avenue for improving selectivity for PfNEK3 over the other kinases. The N-methylpyrazol-4-yl substituent (15) also demonstrated selectivity for PfNEK3, maintaining the PfNEK3 potency of 1 but completely abolishing activity on the other kinases. The tolerability of the large N-methylpyrazol-4-yl substituent inspired us to further investigate the steric requirements of this region. Similarly sized ethers (16, 17) or an ester substituent (18) were unfortunately not tolerated by any kinase, suggesting that the aromaticity of the N-methylpyrazol-4-yl substituent was important.
The carboxamide substituent (19) improved in potency for PfARK1 (3-fold) and PfPK9 (6-fold) over 1, but decreased potency for PfNEK3 (2.5-fold) and PfPKB (4-fold) and completely lost PfARK3 activity. This hence offers an avenue to design for selectivity for PfARK1 over related family member PfARK3.
In summary, we demonstrate that different kinases have different sensitivities to the 5-position substituent, and this knowledge could help to design selective inhibitors among these five kinases. We have shown that PfARK1, PfARK3, and PfPK9 are rather sensitive at this position, only tolerating several substituents (PfARK1: Cl, CF3, CONH2; PfARK3: Cl, Br, I, and CF3; PfPK9: Cl, Br, I, CF3, CONH2). In contrast, PfNEK3 and PfPKB have their preferred substituents (CN and CF3, respectively) but are generally more tolerant of changes.
We next explored SAR at the 4-position of the pyrimidine (Table 3). The removal of the sulfonyl group of the sulfonamide (21) was detrimental toward activity for all five kinases, possibly attributed to the loss of hydrogen bonding potential with the catalytic lysine. Cyclizing the 4-position dianiline ring to a benzimidazolone (30) abolished activity against all five kinases, demonstrating that the position of the hydrogen bond acceptor is key in this region. Having established the importance of a hydrogen bond acceptor, we next replaced the sulfonamide with an amide (22) or urea (23). While 22 retained weak activity on PfARK1, PfPK9, and PfPKB (65–70% activity remaining at 1 μM), this change led to a loss of activity against PfARK3 and PfNEK3. In contrast, 23 is 12-fold more potent against PfPK9 than 1, equipotent against PfPKB, while abolishing activity against PfARK1, PfARK3, and PfNEK3.
Mean values from duplicate experiments using the KinaseSeeker assay.
Mean values from two experiments using the KinaseSeeker assay performed in duplicate.
To investigate the steric requirement of the pocket around the sulfonamide, we increased the size of the methyl group on the sulfonamide to an ethyl (24) or cyclopropyl (25) group, both improving PfARK3 and PfPK9 activity 4-fold and 2-fold, respectively. While 24 was weakly active on PfARK1 and equipotent against PfNEK3 as compared to 1, 25 was 3-fold more potent than 1 on both kinases. A 2- to 2.5-fold drop in potency was observed with PfPKB with both compounds, suggesting that these modifications were tolerated but not preferred. A further increase in size to an isobutyl group (26) was unfavorable for all kinases except PfARK3, which maintained the potency of 1. A phenyl group (27) was preferred by PfARK3, affording a 5-fold improvement in potency over 1, while maintaining potency against PfARK1 and PfNEK3 and dramatically decreasing potency against PfPK9 and PfPKB. A phenyl ring may thus offer an opportunity to achieve selectivity for PfARK3 over the related family member PfARK1. With a N-methylimidazole group (28) however, the activity against PfARK3 was lost, while the potencies with the other four kinases were similar to 27, which suggests that PfARK3 does not prefer polar functionalities here.
For the N-isopropyl sulfamoyl amide (29), inhibition results for all five kinases were similar to those of the isosteric isobutyl sulfonamide 26. This shows that despite favoring additional hydrogen bond donors in urea 23, PfPK9 has a strict steric requirement for this region precluding binding of 29.
Our SAR results for these five Plasmodium kinases demonstrate that the 4-position of the pyrimidine offers an avenue to generate selectivity. With different steric requirements in the active site around this region, we emphasize how single-atom changes here could modulate selectivity between kinases. Selective inhibition of PfARK3 may be achieved using larger lipophilic groups at this region, something not as well tolerated by PfARK1, PfNEK3, PfPK9, and PfPKB. The methyl group is the ideal group for PfPKB, while the slightly larger cyclopropyl group is most ideal for PfARK1 and PfNEK3. We have also found that PfPK9 strongly prefers urea at this position.
The next position for SAR exploration is the 2-position of the pyrimidine. Acknowledging that exocyclic NH typically forms hydrogen bonds with the hinge region of kinases, we opted not to disrupt this key pharmacophore. Substituents on the aryl ring here are frequently used to design for selectivity. For example, a substituent at the position ortho- to the exocyclic NH confers selectivity of TAE684 for ALK, (41) or BI-2536 for PLK1. (42) Because the morpholine ring at the 4-position of the aniline ring was expected to be solvent-exposed, we focused on and established the SAR of 2- (ortho-) and 3-position (meta-) substituents (Table 4).
PfARK1 | PfARK3 | PfNEK3 | PfPK9 | PfPKB | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|
compound | R | % activity remaining at 1 μMa | IC50 (μM)b | % activity remaining at 1 μMa | IC50 (μM)b | % activity remaining at 1 μMa | IC50 (μM)b | % activity remaining at 1 μMa | IC50 (μM)b | % activity remaining at 1 μMa | IC50 (μM)b |
1 | 2-OMe | 53 | 3.4 | 50 | 1.8 | 34 | 1.0 | 43 | 1.2 | 11 | 0.070 |
31 | 2-H, 3-H | 21 | 0.38 | 42 | 1.2 | 2 | 0.11 | 6 | 0.044 | 9 | 0.021 |
32 | 2-OEt | 100 | 12 | 73 | 3.0 | 49 | 2.0 | 78 | 11 | 46 | 0.75 |
33 | 2-OH | 69 | 3.1 | 81 | 4.3 | 11 | 0.22 | 49 | 1.2 | 7 | 0.43 |
34 | 2-Me | 43 | 2.0 | 67 | 1.2 | 7 | 0.29 | 20 | 0.46 | 10 | 0.12 |
35 | 2-F | 38 | 1.9 | 46 | 2.5 | 8 | 0.42 | 20 | 0.35 | 8 | 0.051 |
36 | 2-Cl | 84 | 2.7 | 74 | 4.0 | 7 | 0.55 | 32 | 0.59 | 0 | 0.037 |
37 | 3-OMe | 9 | 0.24 | 11 | 0.43 | 6 | 0.060 | 4 | 0.040 | 0 | 0.014 |
38 | 3-Me | 20 | 0.18 | 35 | 0.68 | 7 | 0.057 | 3 | 0.030 | 5 | 0.015 |
39 | 3-Cl | 19 | 0.34 | 44 | 1.0 | 14 | 0.12 | 8 | 0.056 | 6 | 0.017 |
40 | 3-CF3 | 23 | 0.54 | 47 | 0.99 | 8 | 0.049 | 15 | 0.11 | 0 | 0.054 |
Mean values from duplicate experiments using the KinaseSeeker assay.
Mean values from two experiments using the KinaseSeeker assay performed in duplicate.
Removal of the ortho-methoxy group of 1 (31) revealed a dramatic improvement in potency for PfARK1 (9-fold), PfNEK3 (9-fold), and PfPK9 (27-fold), but only a modest improvement in potency against PfPKB (3-fold), and a <2-fold change in potency against PfARK3. Conversely, replacement of the methoxy group with a larger ethoxy group (32) led to a loss of potency against PfARK1 (4-fold), PfNEK3 (2-fold), PfPK9 (9-fold), and PfPKB (11-fold), with a <2-fold change in potency against PfARK3. Interestingly, a smaller hydroxyl group (33) maintained the potency of 1 against PfARK1 and PfPK9, while decreasing the potency against PfARK3 (2.4-fold) and PfPKB (6-fold), but is 5-fold more potent on PfNEK3. The small methyl (34), fluoro (35), or chloro groups (36) maintained potency of 1 against PfARK1, PfARK3, and PfPKB, while improving upon activity of both PfNEK3 and PfPK9 2-to-5-fold. Looking at these trends, we infer that PfARK1 and PfPK9 are particularly sensitive to the steric requirement of the ortho-position group at this region, whereas PfNEK3 and PfPKB are less sensitive and PfARK3 is nondiscriminatory. The steric requirement and preference for a unsubstituted ortho-position could arise from the relief of a steric clash with the middle hinge residue (hinge.47 by KLIFS notation (36)) of the kinase. This steric clash typically occurs for kinases with larger tyrosine or phenylalanine residues while being accommodating of the smaller leucine residue. (41−45) However, because the middle hinge residues of these five kinases are all tyrosine or phenylalanine residues, this does not provide a simple explanation regarding the differential sensitivity toward substituents at this position. We thus emphasize the importance of investigating and understanding SAR to complement sequence-based hypotheses.
We next installed substituents meta- on the NH of the aniline ring. Electron-donating methoxy (37) and methyl (38) substituents improved PfARK1, PfARK3, and PfNEK3 potencies modestly (1.5- to 3-fold) when compared to 31, while PfPK9 and PfPKB potencies were unaffected (≤1.5-fold difference). Addition of a chloro substituent (39) did not change the potency of 31 across all five kinases. Addition of the trifluoromethyl group (40) did not change potency of 31 against PfARK1 and PfARK3, while improving PfNEK3 potency 2.2-fold, yet decreasing PfPK9 and PfPKB potency 2.5-fold. Unlike when substituents are varied at the ortho-position, variation of the meta-position substituents mostly induces modest changes in potency. This striking contrast further demonstrates that the potency changes observed with ortho-position substituents were primarily due to steric effects rather than electronic contributions to the aniline ring. Despite the possibilities of meta-position substituents changing the electron density of the adjacent morpholine ring and its dihedral angle with the phenyl ring, the modest differences in potency suggest that such changes were well-tolerated by these five kinases. Their general tolerability toward substituents at the meta-position also offers opportunities for using substituents at this position to modulate ADME and PK properties of inhibitors in future lead optimization efforts.
Having established SAR on the five Plasmodium kinases, we next investigated how the modulation of their inhibitory activities would translate to antiplasmodial activity. We selected compounds with an IC50 ≤ 0.3 μM against any kinase, and screened them at 1 μM against Pf3D7 using a SYBR Green I-based fluorescence assay to evaluate asexual blood stage viability. (46) We found nine new analogues with <50% viability at 1 μM and further profiled them in dose–response experiments (Table 5). In parallel, we screened them for cytotoxicity (CC50) in HepG2 cells, a human cell line previously used to screen for cytotoxicity of antimalarials. (12)
Pf3D7 blood stage | ||||
---|---|---|---|---|
compound | % viability at 1 μMa | EC50 (μM)b | HepG2 CC50 (μM)c | CC50/EC50 |
1 | 21 | 0.34 | ||
2 | 85 | |||
4 | 15 | 0.69 | 9.8 | 14 |
5 | 14 | 0.54 | >15d | >28 |
6 | 110 | |||
7 | 83 | |||
8 | 39 | 1.2 | 23 | 19 |
9 | 14 | 0.38 | >15d | >39 |
14 | 21 | 0.43 | >50e | >116 |
23 | 95 | |||
24 | 82 | |||
25 | 78 | |||
27 | 65 | |||
31 | 28 | 0.43 | 1.5 | 3.5 |
33 | 82 | |||
34 | 85 | |||
35 | 69 | |||
36 | 90 | |||
37 | 15 | 0.16 | 1.6 | 10 |
38 | 10 | 0.35 | 2.1 | 6.1 |
39 | 7 | 0.43 | 2.8 | 6.4 |
40 | 57 |
Mean values of triplicate experiments using the SYBR Green I-based assay.
Mean values from two experiments performed in duplicate using the SYBR Green I-based assay.
Mean values from two experiments performed in triplicate using the CellTiter-Fluor assay.
No significant cytotoxicity observed up to 15 μM, the observed solubility limit.
Decreases cell viability around 1 μM to ∼60%, but never decreases cell viability below 50%.
We found compounds 4, 5, 9, and 14 to possess submicromolar potencies against Pf3D7 in the asexual blood stage, a comparable potency to 1. These compounds were also only weakly cytotoxic against HepG2 cells, with CC50s of ≥9.8 μM. For 5 and 9, no significant cytotoxicity was observed up to 15 μM, their observed solubility limit in the assay conditions. These four compounds offer at least a 14-fold window between antiplasmodial activity and cytotoxicity. 8 was also weakly active against Pf3D7 with an EC50 of 1.2 μM, while the CC50 was 23 μM, offering a 19-fold window of selectivity. Compounds 31, 37, 38, and 39 were also found to be submicromolar inhibitors of Pf3D7 but exhibit slightly greater cytotoxicity, with CC50s in the low micromolar ranges. One possible reason for the greater cytotoxicity could be increased promiscuity from the removal of the ortho-position methoxy group, possibly enabling inhibition of off-target kinases. Nevertheless, we have found 37 to be a potent antimalarial compound in this series (EC50 = 0.16 μM), a 2-fold improvement over 1, which still possesses a 10-fold window with cytotoxicity (CC50 = 1.6 μM).
Throughout these experiments, we observed that the antiplasmodial activity does not correlate directly with the potency against any kinase, suggesting that there could be other targets for these 2,4-dianilinopyrimidines yet to be identified or the antiplasmodial activity may be attributed to polypharmacology.
To further characterize these compounds, we have additionally determined the kinetic solubility, permeability, human plasma protein binding, and plasma stability of 1 and the nine novel analogues with dose-dependent antiplasmodial activity (Table 6). 1 possesses a good solubility of 71.1 μM. Replacement of the 5-position Cl atom with other substituents (4, 5, 8, 9, 14) decreased solubility. This was especially evident with the CF3 (9) and CN (14) groups, possibly attributed to the decreased basicity of the pyrimidine ring. The methoxy group on the ortho-position on the 2-position aniline is important for solubility, as its removal (31) decreases solubility 8-fold compared to 1. Reinstating the methoxy group at the meta-position (37) restored the solubility to 116 μM. However, a meta-position methyl (38) or chloro (39) group is unfavorable for solubility. Given the modest changes in potency against the five kinase targets with changes at the meta-position, we thus reiterate that this position offers a potential avenue to modulate the physiochemical and pharmacokinetic properties of this chemotype.
compd | kinetic solubility in PBS pH 7.4 (μM)a | PAMPA Pe (×10–6 cm/s)b | human PPB (% bound)a | human plasma stability (% remaining at 6 h)a |
---|---|---|---|---|
1 | 71.1 | 21.4 | 99.0 | 97 |
4 | 46.8 | 18.2 | 99.3 | 101 |
5 | 21.0 | 15.5 | 99.5 | 99 |
8 | 36.0 | 14.8 | 97.6 | 101 |
9 | 1.1 | 17.8 | 97.6 | 103 |
14 | 0.9 | 15.1 | 94.6 | 92 |
31 | 8.5 | 19.1 | 95.9 | 105 |
37 | 116.0 | 20.9 | 98.6 | 103 |
38 | 12.8 | 20.9 | 99.1 | 101 |
39 | 0.4 | 12.0 | 99.4 | 98 |
Mean values of duplicate experiments.
Mean values of triplicate experiments.
We measured the permeability of these compounds by the PAMPA assay and found all analogues to possess moderate-to-excellent permeability. The structural modifications performed did not change the permeability, with <2-fold differences observed among all 10 compounds. It is of interest to measure plasma protein binding because strong binding to plasma proteins may lead to reduced penetrance into the parasite and demonstrate lower antiplasmodial activity. (47) From our human plasma protein binding assay results, we observed that most compounds are highly bound to human plasma proteins, with all compounds at least 94.6% bound. The compounds with the greatest affinity to plasma proteins are 1, 4, 5, 38, and 39, with ≥99% bound. All compounds tested were highly stable in human plasma. The high degree of binding is a potential factor for the disconnect between the kinase inhibitory potency and antiplasmodial potency of these compounds. We note that there is no apparent correlation between the permeability or the degree of plasma protein binding and antiplasmodial potency of these compounds, suggesting that multiple factors are likely involved. Nevertheless, the high degree of binding to plasma proteins is a characteristic of this chemotype which should be taken into account during further lead optimization of these compounds.
In conclusion, we have identified 1 as an inhibitor of five P. falciparum kinases PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB. We have developed SAR against all five kinases (Figure 3) and found subtle changes that improved potency against each kinase. The most potent PfARK1 inhibitor we discovered was 38, achieved by removal of the highly unfavorable ortho-position methoxy group and addition of a meta-position methyl group on the aniline ring at the 2-position of the pyrimidine of 1. In contrast, the most potent PfARK3 inhibitor in our set of compounds was 27, attained by substituting the methylsulfonamide on the aniline ring at the 4-position of the pyrimidine to a phenylsulfonamide. Many potent PfPKB inhibitors were discovered. One of which was 9, where the 5-position chloro group was replaced with a trifluoromethyl group. Of the PfNEK3 inhibitors that we discovered, we highlight 14, which demonstrated a unique improvement in potency by a simple substitution of the 5-position chloro group with a nitrile. Similarly, we highlight 23 as a compound that demonstrated a unique improvement in potency against PfPK9, through the incorporation of a urea group instead of a sulfonamide. Of these novel analogues, 37 also demonstrated an improvement in potency against the P. falciparum asexual blood stage.
We also emphasize the importance of investigating and developing the SAR understanding using two examples, looking at the SAR around the 5-position of the pyrimidine and the ortho-position substituent on the aniline at the 2-position. In both cases, hypotheses for kinase inhibition and selectivity based on key amino acid residues may be too simplistic and are not able to completely explain or predict potency and selectivity. Currently, there is a dearth of medicinal chemistry studies of these five diverse P. falciparum kinases. We hope that this work provides starting points for lead optimization efforts and that the SAR described allows for the development of strategies to identify potent and selective inhibitors for each kinase.
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.3c00354.
Percent identity and similarity for PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB; Use of additional molecular docking constraints; comparison of 1 docked to the structures of PfARK1, PfNEK3, PfPK9, and PfPKB; summary of 1H-13C HMBC results, methods; experimental spectra for CZC-54252 analogues; supplemental references (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.
Acknowledgments
This work is supported by the National Institutes of Health under grant no. (1R44AI150237-01) and the National Science Foundation under grant no. (1644868 and CHE-1726291). We thank the University of North Carolina’s Department of Chemistry Mass Spectrometry Core Laboratory, especially Diane Weatherspoon, for assistance with mass spectrometry analysis. We are grateful for support by the Structural Genomics Consortium (SGC), a registered charity (no. 1097737) that receives funds from Bayer AG, Boehringer Ingelheim, Bristol Myers Squibb, Genentech, Genome Canada through Ontario Genomics Institute [OGI-196], EU/EFPIA/OICR/McGill/KTH/Diamond Innovative Medicines Initiative 2 Joint Undertaking [EUbOPEN grant 875510], Janssen, Merck KGaA (aka EMD in Canada and US), Pfizer and Takeda.
CDPK | calcium-dependent protein kinase |
LRRK2 | leucine rich repeat kinase 2 |
MAPK | mitogen-activated protein kinase |
NEK | never-in-mitosis-A related kinases |
Pf3D7 | P. falciparum 3D7 |
PfARK1 | P. falciparum serine/threonine protein kinase ARK1 |
PfARK3 | P. falciparum serine/threonine protein kinase ARK3, putative |
PfCDPK1 | P. falciparum calcium-dependent protein kinase 1 |
PfCDPK2 | P. falciparum calcium-dependent protein kinase 2 |
PfCDPK4 | P. falciparum calcium-dependent protein kinase 4 |
PfCDPK5 | P. falciparum calcium-dependent protein kinase 5 |
PfCLK3 | P. falciparum cyclin-dependent-like kinase CLK3 |
PfGAP45 | P. falciparum glideosome-associated protein 45 |
PfMAP2 | P. falciparum mitogen-activated protein kinase 2 |
PfNEK1 | P. falciparum NIMA related kinase 1 |
PfNEK3 | P. falciparum NIMA related kinase 3 |
PfPK6 | P. falciparum protein kinase 6 |
PfPK9 | P. falciparum serine/threonine protein kinase PK9 |
PfPKB | P. falciparum RAC-beta serine/threonine protein kinase |
PfPKG | P. falciparum cGMP-dependent protein kinase |
PfUBC13 | P. falciparum ubiquitin-conjugating enzyme E2 13 |
PLK1 | polo like kinase 1. |
References
This article references 47 other publications.
- 1World Malaria Report 2022; World Health Organization: Geneva, 2022.Google ScholarThere is no corresponding record for this reference.
- 2Brown, J. R.; Drewry, D.; Gamo, F.-J.; Garcia-Bustos, J. F. Kinase Inhibitors Among Hits from Malaria Cellular Screens. In Protein Phosphorylation in Parasites; Wiley-VCH: Weinheim, Germany, 2013; pp 261– 291.Google ScholarThere is no corresponding record for this reference.
- 3Lucet, I. S.; Tobin, A.; Drewry, D.; Wilks, A. F.; Doerig, C. Plasmodium Kinases as Targets for New-Generation Antimalarials. Future Med. Chem. 2012, 4, 2295– 2310, DOI: 10.4155/fmc.12.183Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVamtrvN&md5=86dfa73b9b939679b7f80af3eb8f0b43Plasmodium kinases as targets for new-generation antimalarialsLucet, Isabelle S.; Tobin, Andrew; Drewry, David; Wilks, Andrew F.; Doerig, ChristianFuture Medicinal Chemistry (2012), 4 (18), 2295-2310CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. There is an urgent need for the development of new antimalarial drugs with novel modes of actions. The malarial parasite, Plasmodium falciparum, has a relatively small kinome of <100 kinases, with many members exhibiting a high degree of structural divergence from their host counterparts. A no. of Plasmodium kinases have recently been shown by reverse genetics to be essential for various parts of the complex parasitic life cycle, and are thus genetically validated as potential targets. Implementation of mass spectrometry-based phosphoproteomics approaches has informed on key phospho-signalling pathways in the parasite. In addn., global phenotypic screens have revealed a large no. of putative protein kinase inhibitors with antimalarial potency. Taken together, these investigations point to the Plasmodium kinome as a rich source of potential new targets. In this review, we highlight recent progress made towards this goal.
- 4Arendse, L. B.; Wyllie, S.; Chibale, K.; Gilbert, I. H. Plasmodium Kinases as Potential Drug Targets for Malaria: Challenges and Opportunities. ACS Infect. Dis. 2021, 7, 518– 534, DOI: 10.1021/acsinfecdis.0c00724Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFOqurs%253D&md5=d58c6825f248e5acb8351f78e68de65fPlasmodium Kinases as Potential Drug Targets for Malaria: Challenges and OpportunitiesArendse, Lauren B.; Wyllie, Susan; Chibale, Kelly; Gilbert, Ian H.ACS Infectious Diseases (2021), 7 (3), 518-534CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A review. Protein and phosphoinositide kinases have been successfully exploited as drug targets in various disease areas, principally in oncol. In malaria, several protein kinases are under investigation as potential drug targets, and an inhibitor of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4KIIIβ) is currently in phase 2 clin. studies. In this Perspective, we review the potential of kinases as drug targets for the treatment of malaria. Kinases are known to be readily druggable, and many are essential for parasite survival. A key challenge in the design of Plasmodium kinase inhibitors is obtaining selectivity over the corresponding human ortholog(s) and other human kinases due to the highly conserved nature of the shared ATP binding site. Notwithstanding this, there are some notable differences between the Plasmodium and human kinome that may be exploitable. There is also the potential for designed polypharmacol., where several Plasmodium kinases are inhibited by the same drug. Prior to starting the drug discovery process, it is important to carefully assess potential kinase targets to ensure that the inhibition of the desired kinase will kill the parasites in the required life-cycle stages with a sufficiently fast rate of kill. Here, we highlight key target attributes and exptl. approaches to consider and summarize the progress that has been made targeting Plasmodium PI4KIIIβ, cGMP-dependent protein kinase, and cyclin-dependent-like kinase 3.
- 5Ong, H. W.; Adderley, J.; Tobin, A. B.; Drewry, D. H.; Doerig, C. Parasite and Host Kinases as Targets for Antimalarials. Expert Opin. Ther. Targets 2023, 27, 151– 169, DOI: 10.1080/14728222.2023.2185511Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXmt1ynurg%253D&md5=f226d5ba4e0e8836e62fa62fb0058e95Parasite and host kinases as targets for antimalarialsOng, Han Wee; Adderley, Jack; Tobin, Andrew B.; Drewry, David H.; Doerig, ChristianExpert Opinion on Therapeutic Targets (2023), 27 (2), 151-169CODEN: EOTTAO; ISSN:1472-8222. (Taylor & Francis Ltd.)A review. The deployment of Artemisinin-based combination therapies and transmission control measures led to a decrease in the global malaria burden over the recent decades. Unfortunately, this trend is now reversing, in part due to resistance against available treatments, calling for the development of new drugs against untapped targets to prevent cross-resistance. In view of their demonstrated druggability in noninfectious diseases, protein kinases represent attractive targets. Kinase-focussed antimalarial drug discovery is facilitated by the availability of kinase-targeting scaffolds and large libraries of inhibitors, as well as high-throughput phenotypic and biochem. assays. We present an overview of validated Plasmodium kinase targets and their inhibitors, and briefly discuss the potential of host cell kinases as targets for host-directed therapy. We propose priority research areas, including (i) diversification of Plasmodium kinase targets (at present most efforts focus on a very small no. of targets); (ii) polypharmacol. as an avenue to limit resistance (kinase inhibitors are highly suitable in this respect); and (iii) preemptive limitation of resistance through host-directed therapy (targeting host cell kinases that are required for parasite survival) and transmission-blocking through targeting sexual stage-specific kinases as a strategy to protect curative drugs from the spread of resistance.
- 6Ward, P.; Equinet, L.; Packer, J.; Doerig, C. Protein Kinases of the Human Malaria Parasite Plasmodium Falciparum: The Kinome of a Divergent Eukaryote. BMC Genomics 2004, 5, 79, DOI: 10.1186/1471-2164-5-79Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2crlvValtQ%253D%253D&md5=1d5ff6155dbb978b6b2b42c44de18b90Protein kinases of the human malaria parasite Plasmodium falciparum: the kinome of a divergent eukaryoteWard Pauline; Equinet Leila; Packer Jeremy; Doerig ChristianBMC genomics (2004), 5 (), 79 ISSN:.BACKGROUND: Malaria, caused by the parasitic protist Plasmodium falciparum, represents a major public health problem in the developing world. The P. falciparum genome has been sequenced, which provides new opportunities for the identification of novel drug targets. Eukaryotic protein kinases (ePKs) form a large family of enzymes with crucial roles in most cellular processes; hence malarial ePKS represent potential drug targets. We report an exhaustive analysis of the P. falciparum genomic database (PlasmoDB) aimed at identifying and classifying all ePKs in this organism. RESULTS: Using a variety of bioinformatics tools, we identified 65 malarial ePK sequences and constructed a phylogenetic tree to position these sequences relative to the seven established ePK groups. Predominant features of the tree were: (i) that several malarial sequences did not cluster within any of the known ePK groups; (ii) that the CMGC group, whose members are usually involved in the control of cell proliferation, had the highest number of malarial ePKs; and (iii) that no malarial ePK clustered with the tyrosine kinase (TyrK) or STE groups, pointing to the absence of three-component MAPK modules in the parasite. A novel family of 20 ePK-related sequences was identified and called FIKK, on the basis of a conserved amino acid motif. The FIKK family seems restricted to Apicomplexa, with 20 members in P. falciparum and just one member in some other Apicomplexan species. CONCLUSION: The considerable phylogenetic distance between Apicomplexa and other Eukaryotes is reflected by profound divergences between the kinome of malaria parasites and that of yeast or mammalian cells.
- 7Anamika; Srinivasan, N.; Krupa, A. A Genomic Perspective of Protein Kinases in Plasmodium Falciparum. Proteins Struct. Funct. Bioinforma. 2005, 58, 180– 189, DOI: 10.1002/prot.20278Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2cnhvFWgsg%253D%253D&md5=24c832b005212c072ca8dac6143336e6A genomic perspective of protein kinases in Plasmodium falciparumAnamika; Srinivasan N; Krupa AProteins (2005), 58 (1), 180-9 ISSN:.Protein kinases are central to regulation of cellular signaling in the eukaryotes. Well-conserved and lineage-specific protein kinases have previously been identified from various completely sequenced genomes of eukaryotes. The current work describes a genome-wide analysis for protein kinases encoded in the Plasmodium falciparum genome. Using a few different profile matching methods, we have identified 99 protein kinases or related proteins in the parasite genome. We have classified these kinases into subfamilies and analyzed them in the context of noncatalytic domains that occur in these catalytic kinase domain-containing proteins. Compared to most eukaryotic protein kinases, these sequences vary significantly in terms of their lengths, inserts in catalytic domains, and co-occurring domains. Catalytic and noncatalytic domains contain long stretches of repeats of positively charged and other polar amino acids. Various components of the cell cycle, including 4 cyclin-dependent kinase (CDK) homologues, 2 cyclins, 1 CDK regulatory subunit, and 1 kinase-associated phosphatase, are identified. Identification of putative mitogen-activated protein (MAP) Kinase and MAP Kinase Kinase of P. falciparum suggests a new paradigm in the highly conserved signaling pathway of eukaryotes. The calcium-dependent kinase family, well represented in P. falciparum, shows varying domain combinations with EF-hands and pleckstrin homology domains. The analysis reveals a new subfamily of protein kinases having limited sequence similarity with previously known subfamilies. A new transmembrane kinase with 6 membrane-spanning regions is identified. Putative apicoplast targeting sequences have been detected in some of these protein kinases, suggesting their export to the apicoplast.
- 8Solyakov, L.; Halbert, J.; Alam, M. M.; Semblat, J.-P.; Dorin-Semblat, D.; Reininger, L.; Bottrill, A. R.; Mistry, S.; Abdi, A.; Fennell, C.; Holland, Z.; Demarta, C.; Bouza, Y.; Sicard, A.; Nivez, M.-P.; Eschenlauer, S.; Lama, T.; Thomas, D. C.; Sharma, P.; Agarwal, S.; Kern, S.; Pradel, G.; Graciotti, M.; Tobin, A. B.; Doerig, C. Global Kinomic and Phospho-Proteomic Analyses of the Human Malaria Parasite Plasmodium Falciparum. Nat. Commun. 2011, 2, 565, DOI: 10.1038/ncomms1558Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38%252Fjs1SrtQ%253D%253D&md5=8887de8eb952447f55afe5b773875981Global kinomic and phospho-proteomic analyses of the human malaria parasite Plasmodium falciparumSolyakov Lev; Halbert Jean; Alam Mahmood M; Semblat Jean-Philippe; Dorin-Semblat Dominique; Reininger Luc; Bottrill Andrew R; Mistry Sharad; Abdi Abdirhaman; Fennell Clare; Holland Zoe; Demarta Claudia; Bouza Yvan; Sicard Audrey; Nivez Marie-Paule; Eschenlauer Sylvain; Lama Tenzing; Thomas Divya Catherine; Sharma Pushkar; Agarwal Shruti; Kern Selina; Pradel Gabriele; Graciotti Michele; Tobin Andrew B; Doerig ChristianNature communications (2011), 2 (), 565 ISSN:.The role of protein phosphorylation in the life cycle of malaria parasites is slowly emerging. Here we combine global phospho-proteomic analysis with kinome-wide reverse genetics to assess the importance of protein phosphorylation in Plasmodium falciparum asexual proliferation. We identify 1177 phosphorylation sites on 650 parasite proteins that are involved in a wide range of general cellular activities such as DNA synthesis, transcription and metabolism as well as key parasite processes such as invasion and cyto-adherence. Several parasite protein kinases are themselves phosphorylated on putative regulatory residues, including tyrosines in the activation loop of PfGSK3 and PfCLK3; we show that phosphorylation of PfCLK3 Y526 is essential for full kinase activity. A kinome-wide reverse genetics strategy identified 36 parasite kinases as likely essential for erythrocytic schizogony. These studies not only reveal processes that are regulated by protein phosphorylation, but also define potential anti-malarial drug targets within the parasite kinome.
- 9Zhang, M.; Wang, C.; Otto, T. D.; Oberstaller, J.; Liao, X.; Adapa, S. R.; Udenze, K.; Bronner, I. F.; Casandra, D.; Mayho, M.; Brown, J.; Li, S.; Swanson, J.; Rayner, J. C.; Jiang, R. H. Y.; Adams, J. H. Uncovering the Essential Genes of the Human Malaria Parasite Plasmodium Falciparum by Saturation Mutagenesis. Science 2018, 360, eaap7847 DOI: 10.1126/science.aap7847Google ScholarThere is no corresponding record for this reference.
- 10Xing, L.; Rai, B.; Lunney, E. A. Scaffold Mining of Kinase Hinge Binders in Crystal Structure Database. J. Comput. Aided. Mol. Des. 2014, 28, 13– 23, DOI: 10.1007/s10822-013-9700-4Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXitVWhsLfO&md5=0dfcd290ca89041e699b610020e7109bScaffold mining of kinase hinge binders in crystal structure databaseXing, Li; Rai, Brajesh; Lunney, Elizabeth A.Journal of Computer-Aided Molecular Design (2014), 28 (1), 13-23CODEN: JCADEQ; ISSN:0920-654X. (Springer)Protein kinases are the second most prominent group of drug targets, after G-protein-coupled receptors. Despite their distinct inhibition mechanisms, the majority of kinase inhibitors engage the conserved hydrogen bond interactions with the backbone of hinge residues. The authors mined Pfizer internal crystal structure database (CSDb) comprising of several thousand of public as well as internal x-ray binary complexes to compile an inclusive list of hinge binding scaffolds. The min. ring scaffolds with directly attached hetero-atoms and functional groups were extd. from the full compds. by applying a rule-based filtering procedure employing a comprehensive annotation of ATP-binding site of the human kinase complements. The results indicated large no. of kinase inhibitors of diverse chem. structures are derived from a relatively small no. of common scaffolds, which serve as the crit. recognition elements for protein kinase interaction. Out of the nearly 4,000 kinase-inhibitor complexes in the CSDb the authors identified approx. 600 unique scaffolds. Hinge scaffolds are overwhelmingly flat with very little sp3 characteristics, and are less lipophilic than their corresponding parent compds. Examples of the most common as well as the uncommon hinge scaffolds are presented. Although the most common scaffolds are found in complex with multiple kinase targets, a large no. of them are uniquely bound to a specific kinase, suggesting certain scaffolds could be more promiscuous than the others. The compiled collection of hinge scaffolds along with their three-dimensional binding coordinates could serve as basis set for hinge hopping, a practice frequently employed to generate novel invention as well as to optimize existing leads in medicinal chem.
- 11Xing, L.; Klug-Mcleod, J.; Rai, B.; Lunney, E. A. Kinase Hinge Binding Scaffolds and Their Hydrogen Bond Patterns. Bioorg. Med. Chem. 2015, 23, 6520– 6527, DOI: 10.1016/j.bmc.2015.08.006Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVentr3L&md5=048b4b99628c6e1ae273b00a47ab2377Kinase hinge binding scaffolds and their hydrogen bond patternsXing, Li; Klug-Mcleod, Jacquelyn; Rai, Brajesh; Lunney, Elizabeth A.Bioorganic & Medicinal Chemistry (2015), 23 (19), 6520-6527CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Protein kinases constitute a major class of intracellular signaling mols., and describe some of the most prominent drug targets. Kinase inhibitors commonly employ small chem. scaffolds that form hydrogen bonds with the kinase hinge residues connecting the N- and C-terminal lobes of the catalytic domain. In general the satisfied hydrogen bonds are required for potent inhibition, therefore constituting a conserved feature in the majority of inhibitor-kinase interactions. From systematically analyzing the kinase scaffolds extd. from Pfizer crystal structure database (CSDb) the authors recognize that large no. of kinase inhibitors of diverse chem. structures are derived from a relatively small no. of common scaffolds. Depending on specific substitution patterns, scaffolds may demonstrate versatile binding capacities to interact with kinase hinge. Afforded by thousands of ligand-protein binary complexes, the hinge hydrogen bond patterns were analyzed with a focus on their three-dimensional configurations. Most of the compds. engage H6 NH for hinge recognition. Dual hydrogen bonds are commonly obsd. with addnl. recruitment of H4 CO upstream and/or H6 CO downstream. Triple hydrogen bonds accounts for small no. of binary complexes. An unusual hydrogen bond with a non-canonical H5 conformation is obsd., requiring a peptide bond flip by a glycine residue at the H6 position. Addnl. hydrogen bonds to kinase hinge do not necessarily correlate with an increase in potency; conversely they appear to compromise kinase selectivity. Such learnings could enhance the prospect of successful therapy design.
- 12Gamo, F.-J.; Sanz, L. M.; Vidal, J.; de Cozar, C.; Alvarez, E.; Lavandera, J.-L.; Vanderwall, D. E.; Green, D. V. S.; Kumar, V.; Hasan, S.; Brown, J. R.; Peishoff, C. E.; Cardon, L. R.; Garcia-Bustos, J. F. Thousands of Chemical Starting Points for Antimalarial Lead Identification. Nature 2010, 465, 305– 310, DOI: 10.1038/nature09107Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmt1Ois7k%253D&md5=a1fd8c0b89a5650ecba31f5456928b42Thousands of chemical starting points for antimalarial lead identificationGamo, Francisco-Javier; Sanz, Laura M.; Vidal, Jaume; de Cozar, Cristina; Alvarez, Emilio; Lavandera, Jose-Luis; Vanderwall, Dana E.; Green, Darren V. S.; Kumar, Vinod; Hasan, Samiul; Brown, James R.; Peishoff, Catherine E.; Cardon, Lon R.; Garcia-Bustos, Jose F.Nature (London, United Kingdom) (2010), 465 (7296), 305-310CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Malaria is a devastating infection caused by protozoa of the genus Plasmodium. Drug resistance is widespread, no new chem. class of antimalarials has been introduced into clin. practice since 1996 and there is a recent rise of parasite strains with reduced sensitivity to the newest drugs. We screened nearly 2 million compds. in GlaxoSmithKline's chem. library for inhibitors of P. falciparum, of which 13,533 were confirmed to inhibit parasite growth by at least 80% at 2 μM concn. More than 8,000 also showed potent activity against the multidrug resistant strain Dd2. Most (82%) compds. originate from internal company projects and are new to the malaria community. Analyses using historic assay data suggest several novel mechanisms of antimalarial action, such as inhibition of protein kinases and host-pathogen interaction related targets. Chem. structures and assocd. data are hereby made public to encourage addnl. drug lead identification efforts and further research into this disease.
- 13Crowther, G. J.; Hillesland, H. K.; Keyloun, K. R.; Reid, M. C.; Lafuente-Monasterio, M. J.; Ghidelli-Disse, S.; Leonard, S. E.; He, P.; Jones, J. C.; Krahn, M. M.; Mo, J. S.; Dasari, K. S.; Fox, A. M. W.; Boesche, M.; El Bakkouri, M.; Rivas, K. L.; Leroy, D.; Hui, R.; Drewes, G.; Maly, D. J.; Van Voorhis, W. C.; Ojo, K. K. Biochemical Screening of Five Protein Kinases from Plasmodium Falciparum against 14,000 Cell-Active Compounds. PLoS One 2016, 11, e0149996 DOI: 10.1371/journal.pone.0149996Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsFSjurk%253D&md5=a55e4cf30328bbe04218100c69705ebcBiochemical screening of five protein kinases from Plasmodium falciparum against 14,000 cell-active compoundsCrowther, Gregory J.; Hillesland, Heidi K.; Keyloun, Katelyn R.; Reid, Molly C.; Lafuente-Monasterio, Maria Jose; Ghidelli-Disse, Sonja; Leonard, Stephen E.; He, Panqing; Jones, Jackson C.; Krahn, Mallory M.; Mo, Jack S.; Dasari, Kartheek S.; Fox, Anna M. W.; Boesche, Markus; El Bakkouri, Majida; Rivas, Kasey L.; Leroy, Didier; Hui, Raymond; Drewes, Gerard; Maly, Dustin J.; Van Voorhis, Wesley C.; Ojo, Kayode K.PLoS One (2016), 11 (3), e0149996/1-e0149996/16CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)In 2010 the identities of thousands of anti-Plasmodium compds. were released publicly to facilitate malaria drug development. Understanding these compds. mechanisms of action, i.e., the specific mol. targets by which they kill the parasite, would further facilitate the drug development process. Given that kinases are promising anti-malaria targets, the authors screened ∼14,000 cell-active compds. for activity against five different protein kinases. Collections of cell-active compds. from GlaxoSmithKline (the ∼13,000-compd. Tres Cantos Antimalarial Set, or TCAMS), St. Jude Children's Research Hospital (260 compds.), and the Medicines for Malaria Venture (the 400-compd. Malaria Box) were screened in biochem. assays of Plasmodium falciparum calcium-dependent protein kinases 1 and 4 (CDPK1 and CDPK4), mitogen-assocd. protein kinase 2 (MAPK2/ MAP2), protein kinase 6 (PK6), and protein kinase 7 (PK7). Novel potent inhibitors (IC50 < 1 μM) were discovered for three of the kinases: CDPK1, CDPK4, and PK6. The PK6 inhibitors are the most potent yet discovered for this enzyme and deserve further scrutiny. Addnl., kinome-wide competition assays revealed a compd. that inhibits CDPK4 with few effects on ∼150 human kinases, and several related compds. that inhibit CDPK1 and CDPK4 yet have limited cytotoxicity to human (HepG2) cells. The authors' data suggest that inhibiting multiple Plasmodium kinase targets without harming human cells is challenging but feasible.
- 14Phuangsawai, O.; Beswick, P.; Ratanabunyong, S.; Tabtimmai, L.; Suphakun, P.; Obounchoey, P.; Srisook, P.; Horata, N.; Chuckowree, I.; Hannongbua, S.; Ward, S. E.; Choowongkomon, K.; Gleeson, M. P. Evaluation of the Anti-Malarial Activity and Cytotoxicity of 2,4-Diamino-Pyrimidine-Based Kinase Inhibitors. Eur. J. Med. Chem. 2016, 124, 896– 905, DOI: 10.1016/j.ejmech.2016.08.055Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFyms7rM&md5=3c8e425740ebb6387ef42a66ae9bb75eEvaluation of the anti-malarial activity and cytotoxicity of 2,4-diamino-pyrimidine-based kinase inhibitorsPhuangsawai, Oraphan; Beswick, Paul; Ratanabunyong, Siriluk; Tabtimmai, Lueacha; Suphakun, Praphasri; Obounchoey, Phongphat; Srisook, Pimonwan; Horata, Natharinee; Chuckowree, Irina; Hannongbua, Supa; Ward, Simon E.; Choowongkomon, Kiattawee; Gleeson, M. PaulEuropean Journal of Medicinal Chemistry (2016), 124 (), 896-905CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A series of 2,4 diamino-pyrimidines have been identified from an anal. of open access high throughput antimalarial screening data reported by GlaxoSmithKline at the 3D7 and resistant Dd2 strains. SAR expansion has been performed using structural knowledge of the most plausible parasite target. Seventeen new analogs have been synthesized and tested against the resistant K1 strain of Plasmodium falciparum (Pf). The cytotoxicity of the compds. was assessed in Vero and A549 cells and their selectivity towards human kinases including JAK2 and EGFR were undertaken. The authors identified 2 compds. as sub-micromolar inhibitors, with equiv. antimalarial activity to Chloroquine (CQ). Compds. I and II, μM inhibitors of Pf, displayed improved cytotoxicity with weak inhibition of the human kinases.
- 15Toviwek, B.; Phuangsawai, O.; Konsue, A.; Hannongbua, S.; Riley, J.; Mutter, N.; Anderson, M.; Webster, L.; Hallyburton, I.; Read, K. D.; Gleeson, M. P. Preparation, Biological & Cheminformatics-Based Assessment of N2,N4-Diphenylpyrimidine-2,4-Diamine as Potential Kinase-Targeted Antimalarials. Bioorg. Med. Chem. 2021, 46, 116348, DOI: 10.1016/j.bmc.2021.116348Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFemu7%252FM&md5=cf5c3c887dcbf02d1d8149692fd51ea4Preparation, biological & cheminformatics-based assessment of N2,N4-diphenylpyrimidine-2,4-diamine as potential Kinase-targeted antimalarialsToviwek, Borvornwat; Phuangsawai, Oraphan; Konsue, Adchatawut; Hannongbua, Supa; Riley, Jennifer; Mutter, Nicole; Anderson, Mark; Webster, Lauren; Hallyburton, Irene; Read, Kevin D.; Gleeson, M. PaulBioorganic & Medicinal Chemistry (2021), 46 (), 116348CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Twenty eight new N2,N4-diphenylpyrimidine-2,4-diamines have been prepd. in order to expand our understanding of the anti-malarial SAR of the scaffold. The aim of the study was to make structural modifications to improve the overall potency, selectivity and soly. of the series by varying the anilino groups attached to the 2- and 4-position. We evaluated the activity of the compds. against Plasmodium falciparum (Pf) 3D7, cytotoxicity against HepG2, % inhibition at a panel of 10 human kinases, soly., permeability and lipophilicity, and human and rat in vitro clearance. 11 was identified as a potent anti-malarial with an IC50 of 0.66μM at the 3D7 strain and a selectivity (SI) of ∼ 40 in terms of cytotoxicity against the HepG2 cell line. It also displayed low exptl. logD7.4 (2.27), reasonable soly. (124μg/mL), good metabolic stability, but low permeability. A proteo-chemometric workflow was employed to identify putative Pf targets of the most promising compds. Ligand-based similarity searching of the ChEMBL database led to the identification of most probable human targets. These were then used as input for sequence-based searching of the Pf proteome. Homol. modeling and mol. docking were used to evaluate whether compds. could indeed bind to these targets with valid binding modes. In vitro biol. testing against close human analogs of these targets was subsequently undertaken. This allowed us to identify potential Pf targets and human anti-targets that could be exploited in future development.
- 16Bohmer, M. J.; Wang, J.; Istvan, E. S.; Luth, M. R.; Collins, J. E.; Huttlin, E. L.; Wang, L.; Mittal, N.; Hao, M.; Kwiatkowski, N. P.; Gygi, S. P.; Chakrabarti, R.; Deng, X.; Goldberg, D. E.; Winzeler, E. A.; Gray, N. S.; Chakrabarti, D. Human Polo-like Kinase Inhibitors as Antiplasmodials. ACS Infect. Dis. 2023, 9, 1004– 1021, DOI: 10.1021/acsinfecdis.3c00025Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXltFSitrc%253D&md5=9c8a807461ba96403b6577fce1712aeeHuman Polo-like Kinase Inhibitors as AntiplasmodialsBohmer, Monica J.; Wang, Jinhua; Istvan, Eva S.; Luth, Madeline R.; Collins, Jennifer E.; Huttlin, Edward L.; Wang, Lushun; Mittal, Nimisha; Hao, Mingfeng; Kwiatkowski, Nicholas P.; Gygi, Steven P.; Chakrabarti, Ratna; Deng, Xianming; Goldberg, Daniel E.; Winzeler, Elizabeth A.; Gray, Nathanael S.; Chakrabarti, DebopamACS Infectious Diseases (2023), 9 (4), 1004-1021CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Protein kinases have proven to be a very productive class of therapeutic targets, and over 90 inhibitors are currently in clin. use primarily for the treatment of cancer. Repurposing these inhibitors as antimalarials could provide an accelerated path to drug development. In this study, we identified BI-2536 (compd. 1), a known potent human polo-like kinase 1 inhibitor, with low nanomolar antiplasmodial activity. Screening of addnl. PLK1 inhibitors revealed further antiplasmodial candidates despite the lack of an obvious orthologue of PLKs in Plasmodium. A subset of these inhibitors was profiled for their in vitro killing profile, and commonalities between the killing rate and inhibition of nuclear replication were noted. A kinase panel screen identified PfNEK3 as a shared target of these PLK1 inhibitors; however, phosphoproteome anal. confirmed distinct signaling pathways were disrupted by two structurally distinct inhibitors, suggesting PfNEK3 may not be the sole target. Genomic anal. of BI-2536-resistant parasites revealed mutations in genes assocd. with the starvation-induced stress response, suggesting BI-2536 may also inhibit an aminoacyl-tRNA synthetase.
- 17Ramsden, N.; Perrin, J.; Ren, Z.; Lee, B. D.; Zinn, N.; Dawson, V. L.; Tam, D.; Bova, M.; Lang, M.; Drewes, G.; Bantscheff, M.; Bard, F.; Dawson, T. M.; Hopf, C. Chemoproteomics-Based Design of Potent LRRK2-Selective Lead Compounds That Attenuate Parkinson’s Disease-Related Toxicity in Human Neurons. ACS Chem. Biol. 2011, 6, 1021– 1028, DOI: 10.1021/cb2002413Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVSrtLzP&md5=9537f562f2097dd244d77c21333c8c47Chemoproteomics-Based Design of Potent LRRK2-Selective Lead Compounds That Attenuate Parkinson's Disease-Related Toxicity in Human NeuronsRamsden, Nigel; Perrin, Jessica; Ren, Zhao; Lee, Byoung Dae; Zinn, Nico; Dawson, Valina L.; Tam, Danny; Bova, Michael; Lang, Manja; Drewes, Gerard; Bantscheff, Marcus; Bard, Frederique; Dawson, Ted M.; Hopf, CarstenACS Chemical Biology (2011), 6 (10), 1021-1028CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Leucine-rich repeat kinase-2 (LRRK2) mutations are the most important cause of familial Parkinson's disease, and non-selective inhibitors are protective in rodent disease models. Because of their poor potency and selectivity, the neuroprotective mechanism of these tool compds. has remained elusive so far, and it is still unknown whether selective LRRK2 inhibition can attenuate mutant LRRK2-dependent toxicity in human neurons. Here, we employ a chemoproteomics strategy to identify potent, selective, and metabolically stable LRRK2 inhibitors. We demonstrate that CZC-25146 (I) prevents mutant LRRK2-induced injury of cultured rodent and human neurons with mid-nanomolar potency. These precise chem. probes further validate this emerging therapeutic strategy. They will enable more detailed studies of LRRK2-dependent signaling and pathogenesis and accelerate drug discovery.
- 18Jester, B. W.; Cox, K. J.; Gaj, A.; Shomin, C. D.; Porter, J. R.; Ghosh, I. A Coiled-Coil Enabled Split-Luciferase Three-Hybrid System: Applied Toward Profiling Inhibitors of Protein Kinases. J. Am. Chem. Soc. 2010, 132, 11727– 11735, DOI: 10.1021/ja104491hGoogle Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpsV2mtL0%253D&md5=b6b422ada61cbc678e0156eeb07ed9a9A Coiled-Coil Enabled Split-Luciferase Three-Hybrid System: Applied Toward Profiling Inhibitors of Protein KinasesJester, Benjamin W.; Cox, Kurt J.; Gaj, Alicia; Shomin, Carolyn D.; Porter, Jason R.; Ghosh, IndraneelJournal of the American Chemical Society (2010), 132 (33), 11727-11735CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The 518 protein kinases encoded in the human genome are exquisitely regulated and their aberrant function(s) are often assocd. with human disease. Thus, to advance therapeutics and to probe signal transduction cascades, there is considerable interest in the development of inhibitors that can selectively target protein kinases. However, identifying specific compds. against such a large array of protein kinases is difficult to routinely achieve using traditional activity assays, where purified protein kinases are necessary. Toward a simple, rapid, and practical method for identifying specific inhibitors, the authors describe the development and application of a split-protein methodol. using a coiled-coil-assisted three-hybrid system. In this approach, a protein kinase of interest is attached to the C-terminal fragment of split-firefly luciferase and the coiled-coil Fos, which is specific for the coiled-coil Jun, is attached to the N-terminal fragment. Upon addn. of Jun conjugated to a pan-kinase inhibitor such as staurosporine, a three-hybrid complex was established with concomitant reassembly of the split-luciferase enzyme. An inhibitor can be potentially identified by the commensurate loss in split-luciferase activity by displacement of the modified staurosporine. This new three-hybrid approach is potentially general by testing protein kinases from the different kinase families. To interrogate whether this method allows for screening inhibitors, the authors tested six different protein kinases against a library of 80 known protein kinase inhibitors. Finally, this three-hybrid system can potentially provide a rapid method for structure/function anal. as well as aid in the identification of allosteric inhibitors.
- 19Reininger, L.; Wilkes, J. M.; Bourgade, H.; Miranda-Saavedra, D.; Doerig, C. An Essential Aurora-Related Kinase Transiently Associates with Spindle Pole Bodies during Plasmodium Falciparum Erythrocytic Schizogony. Mol. Microbiol. 2011, 79, 205– 221, DOI: 10.1111/j.1365-2958.2010.07442.xGoogle Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFGruro%253D&md5=6b7136888ba61e1d74140e3ef7348216An essential Aurora-related kinase transiently associates with spindle pole bodies during Plasmodium falciparum erythrocytic schizogonyReininger, Luc; Wilkes, Jonathan M.; Bourgade, Helene; Miranda-Saavedra, Diego; Doerig, ChristianMolecular Microbiology (2011), 79 (1), 205-221CODEN: MOMIEE; ISSN:0950-382X. (Wiley-Blackwell)Aurora kinases compose a family of conserved Ser/Thr protein kinases playing essential roles in eukaryotic cell division. To date, Aurora homologues remain uncharacterized in the protozoan phylum Apicompexa. In malaria parasites, the characterization of Aurora kinases may help understand the cell cycle control during erythrocytic schizogony where asynchronous nuclear divisions occur. In this study, we revisited the kinome of Plasmodium falciparum and identified three Aurora-related kinases, Pfark-1, -2, -3. Among these, Pfark-1 is highly conserved in malaria parasites and also appears to be conserved across Apicomplexa. By tagging the endogenous Pfark-1 gene with the green fluorescent protein (GFP) in live parasites, we show that the Pfark-1-GFP protein forms paired dots assocd. with only a subset of nuclei within individual schizonts. Immunofluorescence anal. using an anti-α-tubulin antibody strongly suggests a recruitment of Pfark-1 at duplicated spindle pole bodies at the entry of the M phase of the cell cycle. Unsuccessful attempts at disrupting the Pfark-1 gene with a knockout construct further indicate that Pfark-1 is required for parasite growth in red blood cells. This study provides new insights into the cell cycle control of malaria parasites and reports the importance of Aurora kinases as potential targets for new antimalarials.
- 20Berry, L.; Chen, C.-T.; Reininger, L.; Carvalho, T. G.; El Hajj, H.; Morlon-Guyot, J.; Bordat, Y.; Lebrun, M.; Gubbels, M.-J.; Doerig, C.; Daher, W. The Conserved Apicomplexan Aurora Kinase TgArk3 Is Involved in Endodyogeny, Duplication Rate and Parasite Virulence. Cell. Microbiol. 2016, 18, 1106– 1120, DOI: 10.1111/cmi.12571Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XivVWnt7Y%253D&md5=dcf723b943366896f05adce74df38be8The conserved apicomplexan Aurora kinase TgArk3 is involved in endodyogeny, duplication rate and parasite virulenceBerry, Laurence; Chen, Chun-Ti; Reininger, Luc; Carvalho, Teresa G.; El Hajj, Hiba; Morlon-Guyot, Juliette; Bordat, Yann; Lebrun, Maryse; Gubbels, Marc-Jan; Doerig, Christian; Daher, WassimCellular Microbiology (2016), 18 (8), 1106-1120CODEN: CEMIF5; ISSN:1462-5814. (Wiley-Blackwell)Aurora kinases are eukaryotic serine/threonine protein kinases that regulate key events assocd. with chromatin condensation, centrosome and spindle function and cytokinesis. Elucidating the roles of Aurora kinases in apicomplexan parasites is crucial to understand the cell cycle control during Plasmodium schizogony or Toxoplasma endodyogeny. Here, the authors report on the localization of two previously uncharacterized Toxoplasma Aurora-related kinases (Ark2 and Ark3) in tachyzoites and of the uncharacterized Ark3 ortholog in Plasmodium falciparum erythrocytic stages. In Toxoplasma gondii, TgArk2 and TgArk3 conc. at specific subcellular structures linked to parasite division: the mitotic spindle and intranuclear mitotic structures (TgArk2), and the outer core of the centrosome and the budding daughter cells cytoskeleton (TgArk3). By tagging the endogenous PfArk3 gene with the green fluorescent protein in live parasites, PfArk3 protein expression was shown to peak late in schizogony and localize at the periphery of budding schizonts. Disruption of the TgArk2 gene reveals no essential function for tachyzoite propagation in vitro, which is surprising giving that the P. falciparum and P. berghei orthologs are essential for erythrocyte schizogony. In contrast, knock-down of TgArk3 protein results in pronounced defects in parasite division and a major growth deficiency. TgArk3-depleted parasites display several defects, such as reduced parasite growth rate, delayed egress and parasite duplication, defect in rosette formation, reduced parasite size and invasion efficiency and lack of virulence in mice. The authors' study provides new insights into cell cycle control in Toxoplasma and malaria parasites and highlights Aurora kinase 3 as potential drug target.
- 21Patel, G.; Roncal, N. E.; Lee, P. J.; Leed, S. E.; Erath, J.; Rodriguez, A.; Sciotti, R. J.; Pollastri, M. P. Repurposing Human Aurora Kinase Inhibitors as Leads for Anti-Protozoan Drug Discovery. Med. Chem. Commun. 2014, 5, 655– 658, DOI: 10.1039/C4MD00045EGoogle ScholarThere is no corresponding record for this reference.
- 22Morahan, B. J.; Abrie, C.; Al-Hasani, K.; Batty, M. B.; Corey, V.; Cowell, A. N.; Niemand, J.; Winzeler, E. A.; Birkholtz, L.-M.; Doerig, C.; Garcia-Bustos, J. F. Human Aurora Kinase Inhibitor Hesperadin Reveals Epistatic Interaction between Plasmodium Falciparum PfArk1 and PfNek1 Kinases. Commun. Biol. 2020, 3, 701, DOI: 10.1038/s42003-020-01424-zGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXis12isb%252FP&md5=8f36349abe695fe5b2e7c10e774b3309Human Aurora kinase inhibitor Hesperadin reveals epistatic interaction between Plasmodium falciparum PfArk1 and PfNek1 kinasesMorahan, Belinda J.; Abrie, Clarissa; Al-Hasani, Keith; Batty, Mitchell B.; Corey, Victoria; Cowell, Anne N.; Niemand, Jandeli; Winzeler, Elizabeth A.; Birkholtz, Lyn-Marie; Doerig, Christian; Garcia-Bustos, Jose F.Communications Biology (2020), 3 (1), 701CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)Abstr.: Mitosis has been validated by numerous anti-cancer drugs as being a druggable process, and selective inhibition of parasite proliferation provides an obvious opportunity for therapeutic intervention against malaria. Mitosis is controlled through the interplay between several protein kinases and phosphatases. We show here that inhibitors of human mitotic kinases belonging to the Aurora family inhibit P. falciparum proliferation in vitro with various potencies, and that a genetic selection for mutant parasites resistant to one of the drugs, Hesperadin, identifies a resistance mechanism mediated by a member of a different kinase family, PfNek1 (PF3D7_1228300). Intriguingly, loss of PfNek1 catalytic activity provides protection against drug action. This points to an undescribed functional interaction between Ark and Nek kinases and shows that existing inhibitors can be used to validate addnl. essential and druggable kinase functions in the parasite.
- 23Lye, Y. M.; Chan, M.; Sim, T.-S. Pfnek3: An Atypical Activator of a MAP Kinase in Plasmodium Falciparum. FEBS Lett. 2006, 580, 6083– 6092, DOI: 10.1016/j.febslet.2006.10.003Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFOhsr7E&md5=bfdbb15a199ff5a926db887b0c908c5ePfnek3: An atypical activator of a MAP kinase in Plasmodium falciparumLye, Yu Min; Chan, Maurice; Sim, Tiow-SuanFEBS Letters (2006), 580 (26), 6083-6092CODEN: FEBLAL; ISSN:0014-5793. (Elsevier B.V.)The canonical mitogen-activated protein kinase (MAPK) signal cascade was previously suggested to be atypical in the malaria parasite. This raises queries on the existence of alternative mediators of plasmodial MAPK pathways. This study describes, Pfnek3, a malarial protein kinase belonging to the NIMA (Never in Mitosis, Aspergillus) family. Endogenous Pfnek3 is expressed during late asexual to gametocyte stages and lacks some classical protein kinase sequence motifs. Moreover, Pfnek3 is phylogenetically distant from mammalian NIMA-kinases. Recombinant Pfnek3 was able to phosphorylate and stimulate a malarial MAPK (Pfmap2). Contrastingly, this was not obsd. with two other kinases, Pfmap1 and human MAPK1, suggesting that the Pfnek3-Pfmap2 interaction may be specific for Pfmap2 regulation. In summary, our data reveal a malarial NIMA-kinase with the potential to regulate a MAPK. Possessing biochem. properties divergent from classical mammalian NIMA-kinases, Pfnek3 could potentially be an attractive target for parasite-selective anti-malarials.
- 24Low, H.; Lye, Y. M.; Sim, T.-S. Pfnek3 Functions as an Atypical MAPKK in Plasmodium Falciparum. Biochem. Biophys. Res. Commun. 2007, 361, 439– 444, DOI: 10.1016/j.bbrc.2007.07.047Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXosF2jsLo%253D&md5=9f2fe3b56f19b67bb316d700456ad155Pfnek3 functions as an atypical MAPKK in Plasmodium falciparumLow, Huiyu; Lye, Yu Min; Sim, Tiow-SuanBiochemical and Biophysical Research Communications (2007), 361 (2), 439-444CODEN: BBRCA9; ISSN:0006-291X. (Elsevier)Eukaryotes generally rely on signal transduction by mitogen-activated protein kinases (MAPKs) for activating their regulatory pathways. However, the presence of a complete MAPK cascade in Plasmodium falciparum is debatable because a search of the entire genome did not portray known MAPK kinase (MAPKK) sequences. Via homol. PCR expts., only two copies of plasmodial MAPK homologs (Pfmap1 and Pfmap2) have been identified but their upstream activators remain unknown. In an earlier expt., Pfnek3 was found to be an unusual activator of Pfmap2 in vitro expts., despite its mol. identity as a malarial protein kinase from the NIMA (Never in Mitosis, Aspergillus) family. In this study, the role of Pfnek3 as a likely upstream MAPKK is defined through mol. and biochem. characterization. Since a previous report proposes a TSH motif as an activation site of Pfmap2, its site-directed mutants, T290A, S291A, and H292K were constructed to elucidate the involvement of Pfnek3 in phosphorylating and activating Pfmap2 in a battery of kinase assays. The results suggested that residue T290 is the site of phosphorylation by Pfnek3. This supposition was further supported by liq. chromatog. mass spectrometry. Although P. falciparum does not appear to possess a conventional MAPK cascade, they may rely on other kinases such as Pfnek3 to carry out similar phosphorylation to activate its signaling pathways.
- 25Ong, H. W.; Truong, A.; Kwarcinski, F.; de Silva, C.; Avalani, K.; Havener, T. M.; Chirgwin, M.; Galal, K. A.; Willis, C.; Krämer, A.; Liu, S.; Knapp, S.; Derbyshire, E. R.; Zutshi, R.; Drewry, D. H. Discovery of Potent Plasmodium Falciparum Protein Kinase 6 (PfPK6) Inhibitors with a Type II Inhibitor Pharmacophore. Eur. J. Med. Chem. 2023, 249, 115043, DOI: 10.1016/j.ejmech.2022.115043Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXisV2ntL8%253D&md5=a6d54605fec3347c4cd57a46f397cdb1Discovery of potent Plasmodium falciparum protein kinase 6 (PfPK6) inhibitors with a type II inhibitor pharmacophoreOng, Han Wee; Truong, Anna; Kwarcinski, Frank; de Silva, Chandi; Avalani, Krisha; Havener, Tammy M.; Chirgwin, Michael; Galal, Kareem A.; Willis, Caleb; Kramer, Andreas; Liu, Shubin; Knapp, Stefan; Derbyshire, Emily R.; Zutshi, Reena; Drewry, David H.European Journal of Medicinal Chemistry (2023), 249 (), 115043CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Malaria is a devastating disease that causes significant global morbidity and mortality. The rise of drug resistance against artemisinin-based combination therapy demonstrates the necessity to develop alternative antimalarials with novel mechanisms of action. We report the discovery of Ki8751 as an inhibitor of essential kinase PfPK6. 79 derivs. were designed, synthesized and evaluated for PfPK6 inhibition and antiplasmodial activity. Using group efficiency analyses, we established the importance of key groups on the scaffold consistent with a type II inhibitor pharmacophore. We highlight modifications on the tail group that contribute to antiplasmodial activity, cumulating in the discovery of compd. 67, a PfPK6 inhibitor (IC50 = 13 nM) active against the P. falciparum blood stage (EC50 = 160 nM), and compd. 79, a PfPK6 inhibitor (IC50 < 5 nM) with dual-stage antiplasmodial activity against P. falciparum blood stage (EC50 = 39 nM) and against P. berghei liver stage (EC50 = 220 nM).
- 26Philip, N.; Haystead, T. A. Characterization of a UBC13 Kinase in Plasmodium Falciparum. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 7845– 7850, DOI: 10.1073/pnas.0611601104Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtFCit7g%253D&md5=952f7a70bf86207c61596d9cd2bd8bcfCharacterization of a UBC13 kinase in Plasmodium falciparumPhilip, Nisha; Haystead, Timothy A.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (19), 7845-7850CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Protein kinases are generally recognized as attractive drug targets to treat a variety of human diseases. Recent anal. of the Plasmodium falciparum kinome identified several kinases that are entirely unique to Plasmodium species. The specific functions and targets of most of these enzymes remain largely unknown. Here, we have identified a P. falciparum kinase (PfPK9/PF13_0085 ORF) that does not cluster with any of the typical eukaryotic protein kinases. PfPK9 protein expression was induced ≈18 h after red blood cell infection, and was mainly localized to the parasitophorous vacuolar membrane as well as the cytosol. Recombinant PfPK9 autophosphorylated in vitro and specifically phosphorylated the exogenous substrate histone H1, indicating that it is catalytically active. Phosphopeptide mapping studies showed that autophosphorylation occurred at three residues: T082, T265, and T269. We identified a P. falciparum homolog of the E2 ubiquitin-conjugating enzyme 13 (UBC13) as an endogenous substrate for PfPK9. PfPK9 phosphorylated UBC13 at S106, a highly conserved residue among eukaryotic E2s, and suppressed its ubiquitin-conjugating activity. Our findings not only describe a previously uncharacterized Plasmodium kinase and its likely in vivo target, but also suggest that modulation of UBC13 activity by phosphorylation may be a common regulatory mechanism in eukaryotes.
- 27Raphemot, R.; Eubanks, A. L.; Toro-Moreno, M.; Geiger, R. A.; Hughes, P. F.; Lu, K.-Y.; Haystead, T. A. J.; Derbyshire, E. R. Plasmodium PK9 Inhibitors Promote Growth of Liver-Stage Parasites. Cell Chem. Biol. 2019, 26, 411– 419, DOI: 10.1016/j.chembiol.2018.11.003Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntl2q&md5=a9179165387521f8a7f77ba0a532e25aPlasmodium PK9 Inhibitors Promote Growth of Liver-Stage ParasitesRaphemot, Rene; Eubanks, Amber Leigh; Toro-Moreno, Maria; Geiger, Rechel Anne; Hughes, Philip Floyd; Lu, Kuan-Yi; Haystead, Timothy Arthur James; Derbyshire, Emily RoseCell Chemical Biology (2019), 26 (3), 411-419.e7CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)There is a scarcity of pharmacol. tools to interrogate protein kinase function in Plasmodium parasites, the causative agent of malaria. Among Plasmodiums protein kinases, those characterized as atypical represent attractive drug targets as they lack sequence similarity to human proteins. Here, we describe takinib as a small mol. to bind the atypical P. falciparum protein kinase 9 (PfPK9). PfPK9 phosphorylates the Plasmodium E2 ubiquitin-conjugating enzyme PfUBC13, which mediates K63-linkage-specific polyubiquitination. Takinib is a potent human TAK1 inhibitor, thus we developed the Plasmodium-selective takinib analog HS220. We demonstrate that takinib and HS220 decrease K63-linked ubiquitination in P. falciparum, suggesting PfPK9 inhibition in cells. Takinib and HS220 induce a unique phenotype where parasite size in hepatocytes increases, yet high compd. concns. decrease the no. of parasites. Our studies highlight the role of PK9 in regulating parasite development and the potential of targeting Plasmodium kinases for malaria control.
- 28Maneekesorn, S.; Knuepfer, E.; Green, J. L.; Prommana, P.; Uthaipibull, C.; Srichairatanakool, S.; Holder, A. A. Deletion of Plasmodium Falciparum Ubc13 Increases Parasite Sensitivity to the Mutagen, Methyl Methanesulfonate and Dihydroartemisinin. Sci. Rep. 2021, 11, 21791, DOI: 10.1038/s41598-021-01267-6Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVegt73P&md5=f4c0e790f9fd30975e686c4452a60c73Deletion of Plasmodium falciparum ubc13 increases parasite sensitivity to the mutagen, methyl methanesulfonate and dihydroartemisininManeekesorn, Supawadee; Knuepfer, Ellen; Green, Judith L.; Prommana, Parichat; Uthaipibull, Chairat; Srichairatanakool, Somdet; Holder, Anthony A.Scientific Reports (2021), 11 (1), 21791CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)The inducible Di-Cre system was used to delete the putative ubiquitin-conjugating enzyme 13 gene (ubc13) of Plasmodium falciparum to study its role in ubiquitylation and the functional consequence during the parasite asexual blood stage. Deletion resulted in a significant redn. of parasite growth in vitro, reduced ubiquitylation of the Lys63 residue of ubiquitin attached to protein substrates, and an increased sensitivity of the parasite to both the mutagen, Me methanesulfonate and the antimalarial drug dihydroartemisinin (DHA), but not chloroquine. The parasite was also sensitive to the UBC13 inhibitor NSC697923. The data suggest that this gene does code for an ubiquitin conjugating enzyme responsible for K63 ubiquitylation, which is important in DNA repair pathways as was previously demonstrated in other organisms. The increased parasite sensitivity to DHA in the absence of ubc13 function indicates that DHA may act primarily through this pathway and that inhibitors of UBC13 may both enhance the efficacy of this antimalarial drug and directly inhibit parasite growth.
- 29Kumar, A.; Vaid, A.; Syin, C.; Sharma, P. PfPKB, a Novel Protein Kinase B-like Enzyme from Plasmodium Falciparum I. Identification, Characterization, and Possible Role in Parasite Development. J. Biol. Chem. 2004, 279, 24255– 24264, DOI: 10.1074/jbc.M312855200Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksV2isbk%253D&md5=2bbbbaa7b0b76a037173eb41d298f09cPfPKB, a Novel Protein Kinase B-like Enzyme from Plasmodium falciparum: I. Identification, characterization, and possible role in parasite developmentKumar, Amit; Vaid, Ankush; Syin, Chiang; Sharma, PushkarJournal of Biological Chemistry (2004), 279 (23), 24255-24264CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Extracellular signals control various important functions of a eukaryotic cell, which is often achieved by regulating a battery of protein kinases and phosphatases. Protein Kinase B (PKB) is an important member of the phosphatidylinositol 3-kinase-dependent signaling pathways in several eukaryotes, but the role of PKB in protozoan parasites is not known. We have identified a protein kinase B homolog in Plasmodium falciparum (PfPKB) that is expressed mainly in the schizonts and merozoites. Even though PfPKB shares high sequence homol. with PKB catalytic domain, it lacks a pleckstrin homol. domain typically found at the N terminus of the mammalian enzyme. Biochem. studies performed to understand the mechanism of PfPKB catalytic activation suggested that (i) its activation is dependent on autophosphorylation of a serine residue (Ser-271) in its activation loop region and (ii) PfPKB has an unusual N-terminal region that was found to neg. regulate its catalytic activity. We also identified an inhibitor of PfPKB activity that also inhibits P. falciparum growth, suggesting that this enzyme may be important for the development of the parasite.
- 30Baum, J.; Richard, D.; Healer, J.; Rug, M.; Krnajski, Z.; Gilberger, T.-W.; Green, J. L.; Holder, A. A.; Cowman, A. F. A Conserved Molecular Motor Drives Cell Invasion and Gliding Motility across Malaria Life Cycle Stages and Other Apicomplexan Parasites. J. Biol. Chem. 2006, 281, 5197– 5208, DOI: 10.1074/jbc.M509807200Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhs1ektL0%253D&md5=c5e9deb26afdf2281cfc259d354380d8A Conserved Molecular Motor Drives Cell Invasion and Gliding Motility across Malaria Life Cycle Stages and Other Apicomplexan ParasitesBaum, Jake; Richard, Dave; Healer, Julie; Rug, Melanie; Krnajski, Zita; Gilberger, Tim-Wolf; Green, Judith L.; Holder, Anthony A.; Cowman, Alan F.Journal of Biological Chemistry (2006), 281 (8), 5197-5208CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Apicomplexan parasites constitute one of the most significant groups of pathogens infecting humans and animals. The liver stage sporozoites of Plasmodium spp. and tachyzoites of Toxoplasma gondii, the causative agents of malaria and toxoplasmosis, resp., use a unique mode of locomotion termed gliding motility to invade host cells and cross cell substrates. This amoeboid-like movement uses a parasite adhesin from the thrombospondin-related anonymous protein (TRAP) family and a set of proteins linking the extracellular adhesin, via an actin-myosin motor, to the inner membrane complex. The Plasmodium blood stage merozoite, however, does not exhibit gliding motility. Here we show that homologs of the key proteins that make up the motor complex, including the recently identified glideosome-assocd. proteins 45 and 50 (GAP45 and GAP50), are present in P. falciparum merozoites and appear to function in erythrocyte invasion. Furthermore, we identify a merozoite TRAP homolog, termed MTRAP, a micronemal protein that shares key features with TRAP, including a thrombospondin repeat domain, a putative rhomboid-protease cleavage site, and a cytoplasmic tail that, in vitro, binds the actin-binding protein aldolase. Anal. of other parasite genomes shows that the components of this motor complex are conserved across diverse Apicomplexan genera. Conservation of the motor complex suggests that a common mol. mechanism underlies all Apicomplexan motility, which, given its unique properties, highlights a no. of novel targets for drug intervention to treat major diseases of humans and livestock.
- 31Jones, M. L.; Kitson, E. L.; Rayner, J. C. Plasmodium Falciparum Erythrocyte Invasion: A Conserved Myosin Associated Complex. Mol. Biochem. Parasitol. 2006, 147, 74– 84, DOI: 10.1016/j.molbiopara.2006.01.009Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjvVWmsrk%253D&md5=3e7db83cf98a9fc7081650857af81ad9Plasmodium falciparum erythrocyte invasion: A conserved myosin associated complexJones, Matthew L.; Kitson, Erika L.; Rayner, Julian C.Molecular & Biochemical Parasitology (2006), 147 (1), 74-84CODEN: MBIPDP; ISSN:0166-6851. (Elsevier Ltd.)Host cell invasion by apicomplexan parasites is powered by an actin/myosin motor complex that has been most thoroughly described in Toxoplasma gondii tachyzoites. In T. gondii, two inner membrane complex (IMC) proteins, the peripheral protein TgGAP45 and the transmembrane protein TgGAP50, form a complex with the myosin, TgMyoA, and its light chain, TgMLC1. This complex, referred to as the glideosome, anchors the invasion motor to the IMC. We have identified and characterized orthologues of TgMLC1, TgGAP45 and TgGAP50 in blood-stages of the major human pathogen Plasmodium falciparum, supporting the idea that the same basic complex drives host cell invasion across the apicomplexan phylum. The P. falciparum glideosome proteins are transcribed, expressed and localized in a manner consistent with a role in erythrocyte invasion. Furthermore, PfMyoA interacts with PfMTIP through broadly conserved mechanisms described in other eukaryotes, and forms a complex with PfGAP45 and PfGAP50 in late schizonts and merozoites. P. falciparum is known to use multiple alternative invasion pathways to enter erythrocytes, hampering vaccine development efforts targeting erythrocyte invasion. Our data suggests that the same invasion motor underpins all alternative invasion pathways, making it an attractive target for the development of novel intervention strategies.
- 32Thomas, D. C.; Ahmed, A.; Gilberger, T. W.; Sharma, P. Regulation of Plasmodium Falciparum Glideosome Associated Protein 45 (PfGAP45) Phosphorylation. PLoS One 2012, 7, e35855 DOI: 10.1371/journal.pone.0035855Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmvFOntrc%253D&md5=69fc93944aa2521edfc2e6f5abb3cb42Regulation of Plasmodium falciparum Glideosome Associated Protein 45 (PfGAP45) phosphorylationThomas, Divya Catherine; Ahmed, Anwar; Gilberger, Tim Wolf; Sharma, PushkarPLoS One (2012), 7 (4), e35855CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The actomyosin motor complex of the glideosome provides the force needed by apicomplexan parasites such as Toxoplasma gondii (Tg) and Plasmodium falciparum (Pf) to invade their host cells and for gliding motility of their motile forms. Glideosome Assocd. Protein 45 (PfGAP45) is an essential component of the glideosome complex as it facilitates anchoring and effective functioning of the motor. Dissection of events that regulate PfGAP45 may provide insights into how the motor and the glideosome operate. We found that PfGAP45 is phosphorylated in response to Phospholipase C (PLC) and calcium signaling. It is phosphorylated by P. falciparum kinases Protein Kinase B (PfPKB) and Calcium Dependent Protein Kinase 1 (PfCDPK1), which are calcium dependent enzymes, at S89, S103 and S149. The Phospholipase C pathway influenced the phosphorylation of S103 and S149. The phosphorylation of PfGAP45 at these sites is differentially regulated during parasite development. The localization of PfGAP45 and its assocn. may be independent of the phosphorylation of these sites. PfGAP45 regulation in response to calcium fits in well with the previously described role of calcium in host cell invasion by malaria parasite.
- 33Vaid, A.; Thomas, D. C.; Sharma, P. Role of Ca2+/Calmodulin-PfPKB Signaling Pathway in Erythrocyte Invasion by Plasmodium Falciparum. J. Biol. Chem. 2008, 283, 5589– 5597, DOI: 10.1074/jbc.M708465200Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXit1Omt7g%253D&md5=0a854ef8a85f8a080a74ec4b20c17ff8Role of Ca2+/calmodulin-PfPKB signaling pathway in erythrocyte invasion by Plasmodium falciparumVaid, Ankush; Thomas, Divya C.; Sharma, PushkarJournal of Biological Chemistry (2008), 283 (9), 5589-5597CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Mol. mechanisms by which signaling pathways operate in the malaria parasite and control its development are promiscuous. Recently, we reported the identification of a signaling pathway in Plasmodium falciparum, which involves activation of protein kinase B-like enzyme (PfPKB) by calcium/calmodulin. Studies carried out to elucidate the function of this pathway suggested that it may be important for erythrocyte invasion. Blocking the function of the upstream activators of this pathway, calmodulin and phospholipase C, resulted in impaired invasion. To evaluate if this signaling cascade controls invasion by regulating PfPKB, inhibitors against this kinase were developed. PfPKB inhibitors dramatically reduced the ability of the parasite to invade erythrocytes. Furthermore, we demonstrate that PfPKB assocs. with actin-myosin motor and phosphorylates PfGAP45 (glideosome-assocd. protein 45), one of the important components of the motor complex, which may help explain its role in erythrocyte invasion.
- 34Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S. A. A.; Ballard, A. J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A. W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly Accurate Protein Structure Prediction with AlphaFold. Nature 2021, 596, 583– 589, DOI: 10.1038/s41586-021-03819-2Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaktrrL&md5=25964ab1157cd5b74a437333dd86650dHighly accurate protein structure prediction with AlphaFoldJumper, John; Evans, Richard; Pritzel, Alexander; Green, Tim; Figurnov, Michael; Ronneberger, Olaf; Tunyasuvunakool, Kathryn; Bates, Russ; Zidek, Augustin; Potapenko, Anna; Bridgland, Alex; Meyer, Clemens; Kohl, Simon A. A.; Ballard, Andrew J.; Cowie, Andrew; Romera-Paredes, Bernardino; Nikolov, Stanislav; Jain, Rishub; Adler, Jonas; Back, Trevor; Petersen, Stig; Reiman, David; Clancy, Ellen; Zielinski, Michal; Steinegger, Martin; Pacholska, Michalina; Berghammer, Tamas; Bodenstein, Sebastian; Silver, David; Vinyals, Oriol; Senior, Andrew W.; Kavukcuoglu, Koray; Kohli, Pushmeet; Hassabis, DemisNature (London, United Kingdom) (2021), 596 (7873), 583-589CODEN: NATUAS; ISSN:0028-0836. (Nature Portfolio)Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous exptl. effort, the structures of around 100,000 unique proteins have been detd., but this represents a small fraction of the billions of known protein sequences. Structural coverage is bottlenecked by the months to years of painstaking effort required to det. a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence-the structure prediction component of the 'protein folding problem'-has been an important open research problem for more than 50 years. Despite recent progress, existing methods fall far short of at. accuracy, esp. when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with at. accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Crit. Assessment of protein Structure Prediction (CASP14), demonstrating accuracy competitive with exptl. structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates phys. and biol. knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm.
- 35Varadi, M.; Anyango, S.; Deshpande, M.; Nair, S.; Natassia, C.; Yordanova, G.; Yuan, D.; Stroe, O.; Wood, G.; Laydon, A.; Žídek, A.; Green, T.; Tunyasuvunakool, K.; Petersen, S.; Jumper, J.; Clancy, E.; Green, R.; Vora, A.; Lutfi, M.; Figurnov, M.; Cowie, A.; Hobbs, N.; Kohli, P.; Kleywegt, G.; Birney, E.; Hassabis, D.; Velankar, S. AlphaFold Protein Structure Database: Massively Expanding the Structural Coverage of Protein-Sequence Space with High-Accuracy Models. Nucleic Acids Res. 2022, 50, D439– D444, DOI: 10.1093/nar/gkab1061Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1Churw%253D&md5=1e36970ae4b901a4acd62fd2d8ff23bfAlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy modelsVaradi, Mihaly; Anyango, Stephen; Deshpande, Mandar; Nair, Sreenath; Natassia, Cindy; Yordanova, Galabina; Yuan, David; Stroe, Oana; Wood, Gemma; Laydon, Agata; Zidek, Augustin; Green, Tim; Tunyasuvunakool, Kathryn; Petersen, Stig; Jumper, John; Clancy, Ellen; Green, Richard; Vora, Ankur; Lutfi, Mira; Figurnov, Michael; Cowie, Andrew; Hobbs, Nicole; Kohli, Pushmeet; Kleywegt, Gerard; Birney, Ewan; Hassabis, Demis; Velankar, SameerNucleic Acids Research (2022), 50 (D1), D439-D444CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The AlphaFold Protein Structure Database is an openly accessible, extensive database of high-accuracy protein-structure predictions. Powered by AlphaFold v2.0 of DeepMind, it has enabled an unprecedented expansion of the structural coverage of the known protein-sequence space. AlphaFold DB provides programmatic access to and interactive visualization of predicted at. coordinates, per-residue and pairwise model-confidence ests. and predicted aligned errors. The initial release of AlphaFold DB contains over 360,000 predicted structures across 21 model-organism proteomes, which will soon be expanded to cover most of the (over 100 million) representative sequences from the UniRef90 data set.
- 36van Linden, O. P. J.; Kooistra, A. J.; Leurs, R.; de Esch, I. J. P.; de Graaf, C. KLIFS: A Knowledge-Based Structural Database To Navigate Kinase–Ligand Interaction Space. J. Med. Chem. 2014, 57, 249– 277, DOI: 10.1021/jm400378wGoogle Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1Ojur7P&md5=8519942e1703ba04f64797e07df4b712KLIFS: A Knowledge-Based Structural Database To Navigate Kinase-Ligand Interaction Spacevan Linden, Oscar P. J.; Kooistra, Albert J.; Leurs, Rob; de Esch, Iwan J. P.; de Graaf, ChrisJournal of Medicinal Chemistry (2014), 57 (2), 249-277CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Protein kinases regulate the majority of signal transduction pathways in cells and have become important targets for the development of designer drugs. We present a systematic anal. of kinase-ligand interactions in all regions of the catalytic cleft of all 1252 human kinase-ligand cocrystal structures present in the Protein Data Bank (PDB). The kinase-ligand interaction fingerprints and structure database (KLIFS) contains a consistent alignment of 85 kinase ligand binding site residues that enables the identification of family specific interaction features and classification of ligands according to their binding modes. We illustrate how systematic mining of kinase-ligand interaction space gives new insights into how conserved and selective kinase interaction hot spots can accommodate the large diversity of chem. scaffolds in kinase ligands. These analyses lead to an improved understanding of the structural requirements of kinase binding that will be useful in ligand discovery and design studies.
- 37Lategahn, J.; Tumbrink, H. L.; Schultz-Fademrecht, C.; Heimsoeth, A.; Werr, L.; Niggenaber, J.; Keul, M.; Parmaksiz, F.; Baumann, M.; Menninger, S.; Zent, E.; Landel, I.; Weisner, J.; Jeyakumar, K.; Heyden, L.; Russ, N.; Müller, F.; Lorenz, C.; Brägelmann, J.; Spille, I.; Grabe, T.; Müller, M. P.; Heuckmann, J. M.; Klebl, B. M.; Nussbaumer, P.; Sos, M. L.; Rauh, D. Insight into Targeting Exon20 Insertion Mutations of the Epidermal Growth Factor Receptor with Wild Type-Sparing Inhibitors. J. Med. Chem. 2022, 65, 6643– 6655, DOI: 10.1021/acs.jmedchem.1c02080Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFems7vO&md5=242416ee3cac2aa17a2536bb8e8670d0Insight into Targeting Exon20 Insertion Mutations of the Epidermal Growth Factor Receptor with Wild Type-Sparing InhibitorsLategahn, Jonas; Tumbrink, Hannah L.; Schultz-Fademrecht, Carsten; Heimsoeth, Alena; Werr, Lisa; Niggenaber, Janina; Keul, Marina; Parmaksiz, Fatma; Baumann, Matthias; Menninger, Sascha; Zent, Eldar; Landel, Ina; Weisner, Joern; Jeyakumar, Kirujan; Heyden, Leonie; Russ, Nicole; Mueller, Fabienne; Lorenz, Carina; Braegelmann, Johannes; Spille, Inga; Grabe, Tobias; Mueller, Matthias P.; Heuckmann, Johannes M.; Klebl, Bert M.; Nussbaumer, Peter; Sos, Martin L.; Rauh, DanielJournal of Medicinal Chemistry (2022), 65 (9), 6643-6655CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Despite the clin. efficacy of epidermal growth factor receptor (EGFR) inhibitors, a subset of patients with non-small cell lung cancer displays insertion mutations in exon20 in EGFR and Her2 with limited treatment options. Here, we present the development and characterization of the novel covalent inhibitors LDC8201 and LDC0496 based on a 1H-pyrrolo[2,3-b]pyridine scaffold. They exhibited intense inhibitory potency toward EGFR and Her2 exon20 insertion mutations as well as selectivity over wild type EGFR and within the kinome. Complex crystal structures with the inhibitors and biochem. and cellular on-target activity document their favorable binding characteristics. Ultimately, we obsd. tumor shrinkage in mice engrafted with patient-derived EGFR-H773_V774insNPH mutant cells during treatment with LDC8201. Together, these results highlight the potential of covalent pyrrolopyridines as inhibitors to target exon20 insertion mutations.
- 38Alam, M.; Beevers, R. E.; Ceska, T.; Davenport, R. J.; Dickson, K. M.; Fortunato, M.; Gowers, L.; Haughan, A. F.; James, L. A.; Jones, M. W.; Kinsella, N.; Lowe, C.; Meissner, J. W. G.; Nicolas, A. L.; Perry, B. G.; Phillips, D. J.; Pitt, W. R.; Platt, A.; Ratcliffe, A. J.; Sharpe, A.; Tait, L. J. Synthesis and SAR of Aminopyrimidines as Novel C-Jun N-Terminal Kinase (JNK) Inhibitors. Bioorg. Med. Chem. Lett. 2007, 17, 3463– 3467, DOI: 10.1016/j.bmcl.2007.03.078Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtValsb4%253D&md5=15d7fb1836a7a11eb94d57a4e1a4265aSynthesis and SAR of aminopyrimidines as novel c-Jun N-terminal kinase (JNK) inhibitorsAlam, Mahbub; Beevers, Rebekah E.; Ceska, Tom; Davenport, Richard J.; Dickson, Karen M.; Fortunato, Mara; Gowers, Lewis; Haughan, Alan F.; James, Lynwen A.; Jones, Mark W.; Kinsella, Natasha; Lowe, Christopher; Meissner, Johannes W. G.; Nicolas, Anne-Lise; Perry, Benjamin G.; Phillips, David J.; Pitt, William R.; Platt, Adam; Ratcliffe, Andrew J.; Sharpe, Andrew; Tait, Laura J.Bioorganic & Medicinal Chemistry Letters (2007), 17 (12), 3463-3467CODEN: BMCLE8; ISSN:0960-894X. (Elsevier Ltd.)The development of a series of aminopyrimidines, e.g., I, as inhibitors of c-Jun N-terminal kinases is described. The synthesis, in vitro inhibitory values for JNK1, JNK2 and CDK2, and the in vitro inhibitory value for a c-Jun cellular assay were discussed.
- 39Lange, A.; Günther, M.; Büttner, F. M.; Zimmermann, M. O.; Heidrich, J.; Hennig, S.; Zahn, S.; Schall, C.; Sievers-Engler, A.; Ansideri, F.; Koch, P.; Laemmerhofer, M.; Stehle, T.; Laufer, S. A.; Boeckler, F. M. Targeting the Gatekeeper MET146 of C-Jun N-Terminal Kinase 3 Induces a Bivalent Halogen/Chalcogen Bond. J. Am. Chem. Soc. 2015, 137, 14640– 14652, DOI: 10.1021/jacs.5b07090Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslegt73O&md5=d2205de85d5be4dc3f97d008ed838251Targeting the Gatekeeper MET146 of C-Jun N-Terminal Kinase 3 Induces a Bivalent Halogen/Chalcogen BondLange, Andreas; Guenther, Marcel; Buettner, Felix Michael; Zimmermann, Markus O.; Heidrich, Johannes; Hennig, Susanne; Zahn, Stefan; Schall, Christoph; Sievers-Engler, Adrian; Ansideri, Francesco; Koch, Pierre; Laemmerhofer, Michael; Stehle, Thilo; Laufer, Stefan A.; Boeckler, Frank M.Journal of the American Chemical Society (2015), 137 (46), 14640-14652CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We target the gatekeeper MET146 of c-Jun N-terminal kinase 3 (JNK3) to exemplify the applicability of X···S halogen bonds in mol. design using computational, synthetic, structural and biophys. techniques. In a designed series of aminopyrimidine-based inhibitors, we unexpectedly encounter a plateau of affinity. Compared to their QM-calcd. interaction energies, particularly bromine and iodine fail to reach the full potential according to the size of their σ-hole. Instead, mutation of the gatekeeper residue into leucine, alanine, or threonine reveals that the heavier halides can significantly influence selectivity in the human kinome. Thus, we demonstrate that, although the choice of halogen may not always increase affinity, it can still be relevant for inducing selectivity. Detg. the crystal structure of the iodine deriv. in complex with JNK3 (4X21) reveals an unusual bivalent halogen/chalcogen bond donated by the ligand and the back-pocket residue MET115. Incipient repulsion from the too short halogen bond increases the flexibility of Cε of MET146, whereas the rest of the residue fails to adapt being fixed by the chalcogen bond. This effect can be useful to induce selectivity, as the necessary combination of methionine residues only occurs in 9.3% of human kinases, while methionine is the predominant gatekeeper (39%).
- 40Hole, A. J.; Baumli, S.; Shao, H.; Shi, S.; Huang, S.; Pepper, C.; Fischer, P. M.; Wang, S.; Endicott, J. A.; Noble, M. E. Comparative Structural and Functional Studies of 4-(Thiazol-5-Yl)-2-(Phenylamino)Pyrimidine-5-Carbonitrile CDK9 Inhibitors Suggest the Basis for Isotype Selectivity. J. Med. Chem. 2013, 56, 660– 670, DOI: 10.1021/jm301495vGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVKhurjE&md5=8c09b13b5f828b7a013607cab64b0b80Comparative Structural and Functional Studies of 4-(Thiazol-5-yl)-2-(phenylamino)pyrimidine-5-carbonitrile CDK9 Inhibitors Suggest the Basis for Isotype SelectivityHole, Alison J.; Baumli, Sonja; Shao, Hao; Shi, Shenhua; Huang, Shiliang; Pepper, Chris; Fischer, Peter M.; Wang, Shudong; Endicott, Jane A.; Noble, Martin E.Journal of Medicinal Chemistry (2013), 56 (3), 660-670CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Cyclin-dependent kinase 9/cyclin T, the protein kinase heterodimer that constitutes pos. transcription elongation factor b, is a well-validated target for treatment of several diseases, including cancer and cardiac hypertrophy. In order to aid inhibitor design and rationalize the basis for CDK9 selectivity, we have studied the CDK-binding properties of six different members of a 4-(thiazol-5-yl)-2-(phenylamino)pyrimidine-5-carbonitrile series that bind to both CDK9/cyclin T and CDK2/cyclin A. We find that for a given CDK, the melting temp. of a CDK/cyclin/inhibitor complex correlates well with inhibitor potency, suggesting that differential scanning fluorometry (DSF) is a useful orthogonal measure of inhibitory activity for this series. We have used DSF to demonstrate that the binding of these compds. is independent of the presence or absence of the C-terminal tail region of CDK9, unlike the binding of the CDK9-selective inhibitor 5,6-dichlorobenzimidazone-1-β-D-ribofuranoside (DRB). Finally, on the basis of 11 cocrystal structures bound to CDK9/cyclin T or CDK2/cyclin A, we conclude that selective inhibition of CDK9/cyclin T by members of the 4-(thiazol-5-yl)-2-(phenylamino)pyrimidine-5-carbonitrile series results from the relative malleability of the CDK9 active site rather than from the formation of specific polar contacts.
- 41Galkin, A. V.; Melnick, J. S.; Kim, S.; Hood, T. L.; Li, N.; Li, L.; Xia, G.; Steensma, R.; Chopiuk, G.; Jiang, J.; Wan, Y.; Ding, P.; Liu, Y.; Sun, F.; Schultz, P. G.; Gray, N. S.; Warmuth, M. Identification of NVP-TAE684, a Potent, Selective, and Efficacious Inhibitor of NPM-ALK. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 270– 275, DOI: 10.1073/pnas.0609412103Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjt12jsQ%253D%253D&md5=4a3441ee637a67dc496c80730bd0e184Identification of NVP-TAE684, a potent, selective, and efficacious inhibitor of NPM-ALKGalkin, Anna V.; Melnick, Jonathon S.; Kim, Sunjoon; Hood, Tami L.; Li, Nanxin; Li, Lintong; Xia, Gang; Steensma, Ruo; Chopiuk, Greg; Wan, Yongqin; Ding, Peter; Liu, Yi; Sun, Fangxian; Schultz, Peter G.; Gray, Nathanael S.; Warmuth, MarkusProceedings of the National Academy of Sciences of the United States of America (2007), 104 (1), 270-275CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Constitutive overexpression and activation of NPM-ALK fusion protein [t(2:5)(p23;q35)] is a key oncogenic event that drives the survival and proliferation of anaplastic large cell lymphomas (ALCLs). We have identified a highly potent and selective small-mol. ALK inhibitor, NVP-TAE684, which blocked the growth of ALCL-derived and ALK-dependent cell lines with IC50 values between 2 and 10 nM. NVP-TAE684 treatment resulted in a rapid and sustained inhibition of phosphorylation of NPM-ALK and its downstream effectors and subsequent induction of apoptosis and cell cycle arrest. In vivo, NVP-TAE684 suppressed lymphomagenesis in two independent models of ALK-pos. ALCL and induced regression of established Karpas-299 lymphomas. NVP-TAE684 also induced down-regulation of CD30 expression, suggesting that CD30 may be used as a biomarker of therapeutic NPM-ALK kinase activity inhibition.
- 42Kothe, M.; Kohls, D.; Low, S.; Coli, R.; Rennie, G. R.; Feru, F.; Kuhn, C.; Ding, Y.-H. Selectivity-Determining Residues in Plk1. Chem. Biol. Drug Des. 2007, 70, 540– 546, DOI: 10.1111/j.1747-0285.2007.00594.xGoogle Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVGhsbrJ&md5=0f976bb98fe7d0413e8d2314abc68e2bSelectivity-determining residues in Plk1Kothe, Michael; Kohls, Darcy; Low, Simon; Coli, Rocco; Rennie, Glen R.; Feru, Frederic; Kuhn, Cyrille; Ding, Yuan-HuaChemical Biology & Drug Design (2007), 70 (6), 540-546CODEN: CBDDAL; ISSN:1747-0277. (Blackwell Publishing Ltd.)Polo-like kinase 1 is an important regulator of cell cycle progression whose over-expression is often assocd. with oncogenesis. Polo-like kinase 1 hence represents an attractive target for cancer intervention. BI 2536 (Boehringer Ingelheim, Ingelheim, Germany), a Polo-like kinase 1 inhibitor currently in clin. trials, exhibits nanomolar potency against Polo-like kinase isoforms and high selectivity against other kinases. The authors have previously published the crystal structures of the Polo-like kinase 1 domain in complex with AMPPNP and an Aurora A inhibitor. In this work, the authors present the cocrystal structure of Polo-like kinase 1 with BI 2536. The structure, in combination with selectivity data for BI 2536 and related compds., illustrates important features for potency and selectivity. In particular, the authors show that the methoxy group of BI 2536 is an important specificity determinant against non-Polo-like kinases by taking advantage of a small pocket generated by Leu 132 in the hinge region of Polo-like kinase 1. The work presented here provides a framework for structure-based drug design of Polo-like kinase 1-specific inhibitors.
- 43Blake, J. F.; Burkard, M.; Chan, J.; Chen, H.; Chou, K.-J.; Diaz, D.; Dudley, D. A.; Gaudino, J. J.; Gould, S. E.; Grina, J.; Hunsaker, T.; Liu, L.; Martinson, M.; Moreno, D.; Mueller, L.; Orr, C.; Pacheco, P.; Qin, A.; Rasor, K.; Ren, L.; Robarge, K.; Shahidi-Latham, S.; Stults, J.; Sullivan, F.; Wang, W.; Yin, J.; Zhou, A.; Belvin, M.; Merchant, M.; Moffat, J.; Schwarz, J. B. Discovery of (S)-1-(1-(4-Chloro-3-Fluorophenyl)-2-Hydroxyethyl)-4-(2-((1-Methyl-1H-Pyrazol-5-Yl)Amino)Pyrimidin-4-Yl)Pyridin-2(1H)-One (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development. J. Med. Chem. 2016, 59, 5650– 5660, DOI: 10.1021/acs.jmedchem.6b00389Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XoslOrsLs%253D&md5=6d2d52dabea4d443ddee2959df6d45f1Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical DevelopmentBlake, James F.; Burkard, Michael; Chan, Jocelyn; Chen, Huifen; Chou, Kang-Jye; Diaz, Dolores; Dudley, Danette A.; Gaudino, John J.; Gould, Stephen E.; Grina, Jonas; Hunsaker, Thomas; Liu, Lichuan; Martinson, Matthew; Moreno, David; Mueller, Lars; Orr, Christine; Pacheco, Patricia; Qin, Ann; Rasor, Kevin; Ren, Li; Robarge, Kirk; Shahidi-Latham, Sheerin; Stults, Jeffrey; Sullivan, Francis; Wang, Weiru; Yin, Jianping; Zhou, Aihe; Belvin, Marcia; Merchant, Mark; Moffat, John; Schwarz, Jacob B.Journal of Medicinal Chemistry (2016), 59 (12), 5650-5660CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The extracellular signal-regulated kinases ERK1/2 represent an essential node within the RAS/RAF/MEK/ERK signaling cascade that is commonly activated by oncogenic mutations in BRAF or RAS or by upstream oncogenic signaling. While targeting upstream nodes with RAF and MEK inhibitors has proven effective clin., resistance frequently develops through reactivation of the pathway. Simultaneous targeting of multiple nodes in the pathway, such as MEK and ERK, offers the prospect of enhanced efficacy as well as reduced potential for acquired resistance. Described herein is the discovery and characterization of GDC-0994 (22), an orally bioavailable small mol. inhibitor selective for ERK kinase activity.
- 44Johnson, T. W.; Richardson, P. F.; Bailey, S.; Brooun, A.; Burke, B. J.; Collins, M. R.; Cui, J. J.; Deal, J. G.; Deng, Y.; Dinh, D.; Engstrom, L. D.; He, M.; Hoffman, J.; Hoffman, R. L.; Huang, Q.; Kania, R. S.; Kath, J. C.; Lam, H.; Lam, J. L.; Le, P. T.; Lingardo, L.; Liu, W.; McTigue, M.; Palmer, C. L.; Sach, N. W.; Smeal, T.; Smith, G. L.; Stewart, A. E.; Timofeevski, S.; Zhu, H.; Zhu, J.; Zou, H. Y.; Edwards, M. P. Discovery of (10R)-7-Amino-12-Fluoro-2,10,16-Trimethyl-15-Oxo-10,15,16,17-Tetrahydro- 2H −8,4-(Metheno)Pyrazolo[4,3-h][2,5,11]-Benzoxadiazacyclotetradecine-3-Carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-Ros. J. Med. Chem. 2014, 57, 4720– 4744, DOI: 10.1021/jm500261qGoogle Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnvVSgtL4%253D&md5=db21844f8e9a2f9ca5953bca56ad7b1cDiscovery of (10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-ros Oncogene 1 (ROS1) with Preclinical Brain Exposure and Broad-Spectrum Potency against ALK-Resistant MutationsJohnson, Ted W.; Richardson, Paul F.; Bailey, Simon; Brooun, Alexei; Burke, Benjamin J.; Collins, Michael R.; Cui, J. Jean; Deal, Judith G.; Deng, Ya-Li; Dinh, Dac; Engstrom, Lars D.; He, Mingying; Hoffman, Jacqui; Hoffman, Robert L.; Huang, Qinhua; Kania, Robert S.; Kath, John C.; Lam, Hieu; Lam, Justine L.; Le, Phuong T.; Lingardo, Laura; Liu, Wei; McTigue, Michele; Palmer, Cynthia L.; Sach, Neal W.; Smeal, Tod; Smith, Graham L.; Stewart, Albert E.; Timofeevski, Sergei; Zhu, Huichun; Zhu, Jinjiang; Zou, Helen Y.; Edwards, Martin P.Journal of Medicinal Chemistry (2014), 57 (11), 4720-4744CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Although crizotinib demonstrates robust efficacy in anaplastic lymphoma kinase (ALK)-pos. non-small-cell lung carcinoma patients, progression during treatment eventually develops. Resistant patient samples revealed a variety of point mutations in the kinase domain of ALK, including the L1196M gatekeeper mutation. In addn., some patients progress due to cancer metastasis in the brain. Using structure-based drug design, lipophilic efficiency, and phys.-property-based optimization, highly potent macrocyclic ALK inhibitors were prepd. with good absorption, distribution, metab., and excretion (ADME), low propensity for p-glycoprotein 1-mediated efflux, and good passive permeability. These structurally unusual macrocyclic inhibitors were potent against wild-type ALK and clin. reported ALK kinase domain mutations. Significant synthetic challenges were overcome, utilizing novel transformations to enable the use of these macrocycles in drug discovery paradigms. This work led to the discovery of 8k (PF-06463922), combining broad-spectrum potency, central nervous system ADME, and a high degree of kinase selectivity.
- 45Tan, L.; Akahane, K.; McNally, R.; Reyskens, K. M. S. E.; Ficarro, S. B.; Liu, S.; Herter-Sprie, G. S.; Koyama, S.; Pattison, M. J.; Labella, K.; Johannessen, L.; Akbay, E. A.; Wong, K.-K.; Frank, D. A.; Marto, J. A.; Look, T. A.; Arthur, J. S. C.; Eck, M. J.; Gray, N. S. Development of Selective Covalent Janus Kinase 3 Inhibitors. J. Med. Chem. 2015, 58, 6589– 6606, DOI: 10.1021/acs.jmedchem.5b00710Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht12ksLjM&md5=af334f0cdd1d4f58462182f138619099Development of Selective Covalent Janus Kinase 3 InhibitorsTan, Li; Akahane, Koshi; McNally, Randall; Reyskens, Kathleen M. S. E.; Ficarro, Scott B.; Liu, Suhu; Herter-Sprie, Grit S.; Koyama, Shohei; Pattison, Michael J.; Labella, Katherine; Johannessen, Liv; Akbay, Esra A.; Wong, Kwok-Kin; Frank, David A.; Marto, Jarrod A.; Look, Thomas A.; Arthur, J. Simon C.; Eck, Michael J.; Gray, Nathanael S.Journal of Medicinal Chemistry (2015), 58 (16), 6589-6606CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The Janus kinases (JAKs) and their downstream effectors, signal transducer and activator of transcription proteins (STATs), form a crit. immune cell signaling circuit, which is of fundamental importance in innate immunity, inflammation, and hematopoiesis, and dysregulation is frequently obsd. in immune disease and cancer. The high degree of structural conservation of the JAK ATP binding pockets has posed a considerable challenge to medicinal chemists seeking to develop highly selective inhibitors as pharmacol. probes and as clin. drugs. Here we report the discovery and optimization of 2,4-substituted pyrimidines as covalent JAK3 inhibitors that exploit a unique cysteine (Cys909) residue in JAK3. Investigation of structure-activity relationship (SAR) utilizing biochem. and transformed Ba/F3 cellular assays resulted in identification of potent and selective inhibitors such as compds. 9 and 45. A 2.9 Å cocrystal structure of JAK3 in complex with 9 confirms the covalent interaction. Compd. 9 exhibited decent pharmacokinetic properties and is suitable for use in vivo. These inhibitors provide a set of useful tools to pharmacol. interrogate JAK3-dependent biol.
- 46Smilkstein, M.; Sriwilaijaroen, N.; Kelly, J. X.; Wilairat, P.; Riscoe, M. Simple and Inexpensive Fluorescence-Based Technique for High-Throughput Antimalarial Drug Screening. Antimicrob. Agents Chemother. 2004, 48, 1803– 1806, DOI: 10.1128/AAC.48.5.1803-1806.2004Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjslGltLw%253D&md5=84a9721da1d873dfca16d1f2570ed304Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screeningSmilkstein, Martin; Sriwilaijaroen, Nongluk; Kelly, Jane Xu; Wilairat, Prapon; Riscoe, MichaelAntimicrobial Agents and Chemotherapy (2004), 48 (5), 1803-1806CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Radioisotopic assays involve expense, multistep protocols, equipment, and radioactivity safety requirements which are problematic in high-throughput drug testing. This study reports an alternative, simple, robust, inexpensive, one-step fluorescence assay for use in antimalarial drug screening. Parasite growth is detd. by using SYBR Green 1, a dye with marked fluorescence enhancement upon contact with Plasmodium DNA. A side-by-side comparison of this fluorescence assay and a std. radioisotopic method was performed by testing known antimalarial agents against Plasmodium falciparum strain D6. Both assay methods were used to det. the effective concn. of drug that resulted in a 50% redn. in the obsd. counts (EC50) after 48 h of parasite growth in the presence of each drug. The EC50s of chloroquine, quinine, mefloquine, artemisinin, and 3,6-bis-.vepsiln.-(N,N-diethylamino)-amyloxyanthone were similar or identical by both techniques. The results obtained with this new fluorescence assay suggest that it may be an ideal method for high-throughput antimalarial drug screening.
- 47Rakotoarivelo, N. V.; Perio, P.; Najahi, E.; Nepveu, F. Interaction between Antimalarial 2-Aryl-3H-Indol-3-One Derivatives and Human Serum Albumin. J. Phys. Chem. B 2014, 118, 13477– 13485, DOI: 10.1021/jp507569eGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVClsbzF&md5=e36b416d37d4fe81317daa90bd20a3d2Interaction between Antimalarial 2-Aryl-3H-indol-3-one Derivatives and Human Serum AlbuminRakotoarivelo, Nambinina V.; Perio, Pierre; Najahi, Ennaji; Nepveu, FrancoiseJournal of Physical Chemistry B (2014), 118 (47), 13477-13485CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Binding of drugs to plasma proteins, such as albumin, is a major factor which dets. their pharmacokinetics and pharmacol. effects. Therefore, the interactions between human serum albumin (HSA) and four antimalarial compds. selected in the 2-aryl-3H-indol-3-one series have been investigated using UV-visible, fluorescence and CD spectroscopies. Compds. produced a static quenching of the intrinsic fluorescence of HSA. The thermodn. parameters have shown that the binding reaction is endothermic for three compds. while exothermic for the 2-phenyl-3H-indol-3-one, 3. The interaction is entropically driven with predominant hydrophobic forces with binding affinities of the order of 104 M-1. The highest binding const. is obsd. for 3 (Kλ=280nm = 4.53 × 104 M-1) which is also the less active compd. against Plasmodium falciparum. Synchronous fluorescence gave qual. information on the conformational changes of HSA while quant. data were obtained with CD. Displacement expts. with site markers indicated that drugs bind to HSA at site I (subdomain IIA). In addn., the apparent binding const. and the binding site no. were calcd. in the presence of different ions.
Cited By
This article has not yet been cited by other publications.
Article Views
Altmetric
Citations
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 47 other publications.
- 1World Malaria Report 2022; World Health Organization: Geneva, 2022.There is no corresponding record for this reference.
- 2Brown, J. R.; Drewry, D.; Gamo, F.-J.; Garcia-Bustos, J. F. Kinase Inhibitors Among Hits from Malaria Cellular Screens. In Protein Phosphorylation in Parasites; Wiley-VCH: Weinheim, Germany, 2013; pp 261– 291.There is no corresponding record for this reference.
- 3Lucet, I. S.; Tobin, A.; Drewry, D.; Wilks, A. F.; Doerig, C. Plasmodium Kinases as Targets for New-Generation Antimalarials. Future Med. Chem. 2012, 4, 2295– 2310, DOI: 10.4155/fmc.12.1833https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVamtrvN&md5=86dfa73b9b939679b7f80af3eb8f0b43Plasmodium kinases as targets for new-generation antimalarialsLucet, Isabelle S.; Tobin, Andrew; Drewry, David; Wilks, Andrew F.; Doerig, ChristianFuture Medicinal Chemistry (2012), 4 (18), 2295-2310CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. There is an urgent need for the development of new antimalarial drugs with novel modes of actions. The malarial parasite, Plasmodium falciparum, has a relatively small kinome of <100 kinases, with many members exhibiting a high degree of structural divergence from their host counterparts. A no. of Plasmodium kinases have recently been shown by reverse genetics to be essential for various parts of the complex parasitic life cycle, and are thus genetically validated as potential targets. Implementation of mass spectrometry-based phosphoproteomics approaches has informed on key phospho-signalling pathways in the parasite. In addn., global phenotypic screens have revealed a large no. of putative protein kinase inhibitors with antimalarial potency. Taken together, these investigations point to the Plasmodium kinome as a rich source of potential new targets. In this review, we highlight recent progress made towards this goal.
- 4Arendse, L. B.; Wyllie, S.; Chibale, K.; Gilbert, I. H. Plasmodium Kinases as Potential Drug Targets for Malaria: Challenges and Opportunities. ACS Infect. Dis. 2021, 7, 518– 534, DOI: 10.1021/acsinfecdis.0c007244https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFOqurs%253D&md5=d58c6825f248e5acb8351f78e68de65fPlasmodium Kinases as Potential Drug Targets for Malaria: Challenges and OpportunitiesArendse, Lauren B.; Wyllie, Susan; Chibale, Kelly; Gilbert, Ian H.ACS Infectious Diseases (2021), 7 (3), 518-534CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A review. Protein and phosphoinositide kinases have been successfully exploited as drug targets in various disease areas, principally in oncol. In malaria, several protein kinases are under investigation as potential drug targets, and an inhibitor of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4KIIIβ) is currently in phase 2 clin. studies. In this Perspective, we review the potential of kinases as drug targets for the treatment of malaria. Kinases are known to be readily druggable, and many are essential for parasite survival. A key challenge in the design of Plasmodium kinase inhibitors is obtaining selectivity over the corresponding human ortholog(s) and other human kinases due to the highly conserved nature of the shared ATP binding site. Notwithstanding this, there are some notable differences between the Plasmodium and human kinome that may be exploitable. There is also the potential for designed polypharmacol., where several Plasmodium kinases are inhibited by the same drug. Prior to starting the drug discovery process, it is important to carefully assess potential kinase targets to ensure that the inhibition of the desired kinase will kill the parasites in the required life-cycle stages with a sufficiently fast rate of kill. Here, we highlight key target attributes and exptl. approaches to consider and summarize the progress that has been made targeting Plasmodium PI4KIIIβ, cGMP-dependent protein kinase, and cyclin-dependent-like kinase 3.
- 5Ong, H. W.; Adderley, J.; Tobin, A. B.; Drewry, D. H.; Doerig, C. Parasite and Host Kinases as Targets for Antimalarials. Expert Opin. Ther. Targets 2023, 27, 151– 169, DOI: 10.1080/14728222.2023.21855115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXmt1ynurg%253D&md5=f226d5ba4e0e8836e62fa62fb0058e95Parasite and host kinases as targets for antimalarialsOng, Han Wee; Adderley, Jack; Tobin, Andrew B.; Drewry, David H.; Doerig, ChristianExpert Opinion on Therapeutic Targets (2023), 27 (2), 151-169CODEN: EOTTAO; ISSN:1472-8222. (Taylor & Francis Ltd.)A review. The deployment of Artemisinin-based combination therapies and transmission control measures led to a decrease in the global malaria burden over the recent decades. Unfortunately, this trend is now reversing, in part due to resistance against available treatments, calling for the development of new drugs against untapped targets to prevent cross-resistance. In view of their demonstrated druggability in noninfectious diseases, protein kinases represent attractive targets. Kinase-focussed antimalarial drug discovery is facilitated by the availability of kinase-targeting scaffolds and large libraries of inhibitors, as well as high-throughput phenotypic and biochem. assays. We present an overview of validated Plasmodium kinase targets and their inhibitors, and briefly discuss the potential of host cell kinases as targets for host-directed therapy. We propose priority research areas, including (i) diversification of Plasmodium kinase targets (at present most efforts focus on a very small no. of targets); (ii) polypharmacol. as an avenue to limit resistance (kinase inhibitors are highly suitable in this respect); and (iii) preemptive limitation of resistance through host-directed therapy (targeting host cell kinases that are required for parasite survival) and transmission-blocking through targeting sexual stage-specific kinases as a strategy to protect curative drugs from the spread of resistance.
- 6Ward, P.; Equinet, L.; Packer, J.; Doerig, C. Protein Kinases of the Human Malaria Parasite Plasmodium Falciparum: The Kinome of a Divergent Eukaryote. BMC Genomics 2004, 5, 79, DOI: 10.1186/1471-2164-5-796https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2crlvValtQ%253D%253D&md5=1d5ff6155dbb978b6b2b42c44de18b90Protein kinases of the human malaria parasite Plasmodium falciparum: the kinome of a divergent eukaryoteWard Pauline; Equinet Leila; Packer Jeremy; Doerig ChristianBMC genomics (2004), 5 (), 79 ISSN:.BACKGROUND: Malaria, caused by the parasitic protist Plasmodium falciparum, represents a major public health problem in the developing world. The P. falciparum genome has been sequenced, which provides new opportunities for the identification of novel drug targets. Eukaryotic protein kinases (ePKs) form a large family of enzymes with crucial roles in most cellular processes; hence malarial ePKS represent potential drug targets. We report an exhaustive analysis of the P. falciparum genomic database (PlasmoDB) aimed at identifying and classifying all ePKs in this organism. RESULTS: Using a variety of bioinformatics tools, we identified 65 malarial ePK sequences and constructed a phylogenetic tree to position these sequences relative to the seven established ePK groups. Predominant features of the tree were: (i) that several malarial sequences did not cluster within any of the known ePK groups; (ii) that the CMGC group, whose members are usually involved in the control of cell proliferation, had the highest number of malarial ePKs; and (iii) that no malarial ePK clustered with the tyrosine kinase (TyrK) or STE groups, pointing to the absence of three-component MAPK modules in the parasite. A novel family of 20 ePK-related sequences was identified and called FIKK, on the basis of a conserved amino acid motif. The FIKK family seems restricted to Apicomplexa, with 20 members in P. falciparum and just one member in some other Apicomplexan species. CONCLUSION: The considerable phylogenetic distance between Apicomplexa and other Eukaryotes is reflected by profound divergences between the kinome of malaria parasites and that of yeast or mammalian cells.
- 7Anamika; Srinivasan, N.; Krupa, A. A Genomic Perspective of Protein Kinases in Plasmodium Falciparum. Proteins Struct. Funct. Bioinforma. 2005, 58, 180– 189, DOI: 10.1002/prot.202787https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2cnhvFWgsg%253D%253D&md5=24c832b005212c072ca8dac6143336e6A genomic perspective of protein kinases in Plasmodium falciparumAnamika; Srinivasan N; Krupa AProteins (2005), 58 (1), 180-9 ISSN:.Protein kinases are central to regulation of cellular signaling in the eukaryotes. Well-conserved and lineage-specific protein kinases have previously been identified from various completely sequenced genomes of eukaryotes. The current work describes a genome-wide analysis for protein kinases encoded in the Plasmodium falciparum genome. Using a few different profile matching methods, we have identified 99 protein kinases or related proteins in the parasite genome. We have classified these kinases into subfamilies and analyzed them in the context of noncatalytic domains that occur in these catalytic kinase domain-containing proteins. Compared to most eukaryotic protein kinases, these sequences vary significantly in terms of their lengths, inserts in catalytic domains, and co-occurring domains. Catalytic and noncatalytic domains contain long stretches of repeats of positively charged and other polar amino acids. Various components of the cell cycle, including 4 cyclin-dependent kinase (CDK) homologues, 2 cyclins, 1 CDK regulatory subunit, and 1 kinase-associated phosphatase, are identified. Identification of putative mitogen-activated protein (MAP) Kinase and MAP Kinase Kinase of P. falciparum suggests a new paradigm in the highly conserved signaling pathway of eukaryotes. The calcium-dependent kinase family, well represented in P. falciparum, shows varying domain combinations with EF-hands and pleckstrin homology domains. The analysis reveals a new subfamily of protein kinases having limited sequence similarity with previously known subfamilies. A new transmembrane kinase with 6 membrane-spanning regions is identified. Putative apicoplast targeting sequences have been detected in some of these protein kinases, suggesting their export to the apicoplast.
- 8Solyakov, L.; Halbert, J.; Alam, M. M.; Semblat, J.-P.; Dorin-Semblat, D.; Reininger, L.; Bottrill, A. R.; Mistry, S.; Abdi, A.; Fennell, C.; Holland, Z.; Demarta, C.; Bouza, Y.; Sicard, A.; Nivez, M.-P.; Eschenlauer, S.; Lama, T.; Thomas, D. C.; Sharma, P.; Agarwal, S.; Kern, S.; Pradel, G.; Graciotti, M.; Tobin, A. B.; Doerig, C. Global Kinomic and Phospho-Proteomic Analyses of the Human Malaria Parasite Plasmodium Falciparum. Nat. Commun. 2011, 2, 565, DOI: 10.1038/ncomms15588https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38%252Fjs1SrtQ%253D%253D&md5=8887de8eb952447f55afe5b773875981Global kinomic and phospho-proteomic analyses of the human malaria parasite Plasmodium falciparumSolyakov Lev; Halbert Jean; Alam Mahmood M; Semblat Jean-Philippe; Dorin-Semblat Dominique; Reininger Luc; Bottrill Andrew R; Mistry Sharad; Abdi Abdirhaman; Fennell Clare; Holland Zoe; Demarta Claudia; Bouza Yvan; Sicard Audrey; Nivez Marie-Paule; Eschenlauer Sylvain; Lama Tenzing; Thomas Divya Catherine; Sharma Pushkar; Agarwal Shruti; Kern Selina; Pradel Gabriele; Graciotti Michele; Tobin Andrew B; Doerig ChristianNature communications (2011), 2 (), 565 ISSN:.The role of protein phosphorylation in the life cycle of malaria parasites is slowly emerging. Here we combine global phospho-proteomic analysis with kinome-wide reverse genetics to assess the importance of protein phosphorylation in Plasmodium falciparum asexual proliferation. We identify 1177 phosphorylation sites on 650 parasite proteins that are involved in a wide range of general cellular activities such as DNA synthesis, transcription and metabolism as well as key parasite processes such as invasion and cyto-adherence. Several parasite protein kinases are themselves phosphorylated on putative regulatory residues, including tyrosines in the activation loop of PfGSK3 and PfCLK3; we show that phosphorylation of PfCLK3 Y526 is essential for full kinase activity. A kinome-wide reverse genetics strategy identified 36 parasite kinases as likely essential for erythrocytic schizogony. These studies not only reveal processes that are regulated by protein phosphorylation, but also define potential anti-malarial drug targets within the parasite kinome.
- 9Zhang, M.; Wang, C.; Otto, T. D.; Oberstaller, J.; Liao, X.; Adapa, S. R.; Udenze, K.; Bronner, I. F.; Casandra, D.; Mayho, M.; Brown, J.; Li, S.; Swanson, J.; Rayner, J. C.; Jiang, R. H. Y.; Adams, J. H. Uncovering the Essential Genes of the Human Malaria Parasite Plasmodium Falciparum by Saturation Mutagenesis. Science 2018, 360, eaap7847 DOI: 10.1126/science.aap7847There is no corresponding record for this reference.
- 10Xing, L.; Rai, B.; Lunney, E. A. Scaffold Mining of Kinase Hinge Binders in Crystal Structure Database. J. Comput. Aided. Mol. Des. 2014, 28, 13– 23, DOI: 10.1007/s10822-013-9700-410https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXitVWhsLfO&md5=0dfcd290ca89041e699b610020e7109bScaffold mining of kinase hinge binders in crystal structure databaseXing, Li; Rai, Brajesh; Lunney, Elizabeth A.Journal of Computer-Aided Molecular Design (2014), 28 (1), 13-23CODEN: JCADEQ; ISSN:0920-654X. (Springer)Protein kinases are the second most prominent group of drug targets, after G-protein-coupled receptors. Despite their distinct inhibition mechanisms, the majority of kinase inhibitors engage the conserved hydrogen bond interactions with the backbone of hinge residues. The authors mined Pfizer internal crystal structure database (CSDb) comprising of several thousand of public as well as internal x-ray binary complexes to compile an inclusive list of hinge binding scaffolds. The min. ring scaffolds with directly attached hetero-atoms and functional groups were extd. from the full compds. by applying a rule-based filtering procedure employing a comprehensive annotation of ATP-binding site of the human kinase complements. The results indicated large no. of kinase inhibitors of diverse chem. structures are derived from a relatively small no. of common scaffolds, which serve as the crit. recognition elements for protein kinase interaction. Out of the nearly 4,000 kinase-inhibitor complexes in the CSDb the authors identified approx. 600 unique scaffolds. Hinge scaffolds are overwhelmingly flat with very little sp3 characteristics, and are less lipophilic than their corresponding parent compds. Examples of the most common as well as the uncommon hinge scaffolds are presented. Although the most common scaffolds are found in complex with multiple kinase targets, a large no. of them are uniquely bound to a specific kinase, suggesting certain scaffolds could be more promiscuous than the others. The compiled collection of hinge scaffolds along with their three-dimensional binding coordinates could serve as basis set for hinge hopping, a practice frequently employed to generate novel invention as well as to optimize existing leads in medicinal chem.
- 11Xing, L.; Klug-Mcleod, J.; Rai, B.; Lunney, E. A. Kinase Hinge Binding Scaffolds and Their Hydrogen Bond Patterns. Bioorg. Med. Chem. 2015, 23, 6520– 6527, DOI: 10.1016/j.bmc.2015.08.00611https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVentr3L&md5=048b4b99628c6e1ae273b00a47ab2377Kinase hinge binding scaffolds and their hydrogen bond patternsXing, Li; Klug-Mcleod, Jacquelyn; Rai, Brajesh; Lunney, Elizabeth A.Bioorganic & Medicinal Chemistry (2015), 23 (19), 6520-6527CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Protein kinases constitute a major class of intracellular signaling mols., and describe some of the most prominent drug targets. Kinase inhibitors commonly employ small chem. scaffolds that form hydrogen bonds with the kinase hinge residues connecting the N- and C-terminal lobes of the catalytic domain. In general the satisfied hydrogen bonds are required for potent inhibition, therefore constituting a conserved feature in the majority of inhibitor-kinase interactions. From systematically analyzing the kinase scaffolds extd. from Pfizer crystal structure database (CSDb) the authors recognize that large no. of kinase inhibitors of diverse chem. structures are derived from a relatively small no. of common scaffolds. Depending on specific substitution patterns, scaffolds may demonstrate versatile binding capacities to interact with kinase hinge. Afforded by thousands of ligand-protein binary complexes, the hinge hydrogen bond patterns were analyzed with a focus on their three-dimensional configurations. Most of the compds. engage H6 NH for hinge recognition. Dual hydrogen bonds are commonly obsd. with addnl. recruitment of H4 CO upstream and/or H6 CO downstream. Triple hydrogen bonds accounts for small no. of binary complexes. An unusual hydrogen bond with a non-canonical H5 conformation is obsd., requiring a peptide bond flip by a glycine residue at the H6 position. Addnl. hydrogen bonds to kinase hinge do not necessarily correlate with an increase in potency; conversely they appear to compromise kinase selectivity. Such learnings could enhance the prospect of successful therapy design.
- 12Gamo, F.-J.; Sanz, L. M.; Vidal, J.; de Cozar, C.; Alvarez, E.; Lavandera, J.-L.; Vanderwall, D. E.; Green, D. V. S.; Kumar, V.; Hasan, S.; Brown, J. R.; Peishoff, C. E.; Cardon, L. R.; Garcia-Bustos, J. F. Thousands of Chemical Starting Points for Antimalarial Lead Identification. Nature 2010, 465, 305– 310, DOI: 10.1038/nature0910712https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXmt1Ois7k%253D&md5=a1fd8c0b89a5650ecba31f5456928b42Thousands of chemical starting points for antimalarial lead identificationGamo, Francisco-Javier; Sanz, Laura M.; Vidal, Jaume; de Cozar, Cristina; Alvarez, Emilio; Lavandera, Jose-Luis; Vanderwall, Dana E.; Green, Darren V. S.; Kumar, Vinod; Hasan, Samiul; Brown, James R.; Peishoff, Catherine E.; Cardon, Lon R.; Garcia-Bustos, Jose F.Nature (London, United Kingdom) (2010), 465 (7296), 305-310CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Malaria is a devastating infection caused by protozoa of the genus Plasmodium. Drug resistance is widespread, no new chem. class of antimalarials has been introduced into clin. practice since 1996 and there is a recent rise of parasite strains with reduced sensitivity to the newest drugs. We screened nearly 2 million compds. in GlaxoSmithKline's chem. library for inhibitors of P. falciparum, of which 13,533 were confirmed to inhibit parasite growth by at least 80% at 2 μM concn. More than 8,000 also showed potent activity against the multidrug resistant strain Dd2. Most (82%) compds. originate from internal company projects and are new to the malaria community. Analyses using historic assay data suggest several novel mechanisms of antimalarial action, such as inhibition of protein kinases and host-pathogen interaction related targets. Chem. structures and assocd. data are hereby made public to encourage addnl. drug lead identification efforts and further research into this disease.
- 13Crowther, G. J.; Hillesland, H. K.; Keyloun, K. R.; Reid, M. C.; Lafuente-Monasterio, M. J.; Ghidelli-Disse, S.; Leonard, S. E.; He, P.; Jones, J. C.; Krahn, M. M.; Mo, J. S.; Dasari, K. S.; Fox, A. M. W.; Boesche, M.; El Bakkouri, M.; Rivas, K. L.; Leroy, D.; Hui, R.; Drewes, G.; Maly, D. J.; Van Voorhis, W. C.; Ojo, K. K. Biochemical Screening of Five Protein Kinases from Plasmodium Falciparum against 14,000 Cell-Active Compounds. PLoS One 2016, 11, e0149996 DOI: 10.1371/journal.pone.014999613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsFSjurk%253D&md5=a55e4cf30328bbe04218100c69705ebcBiochemical screening of five protein kinases from Plasmodium falciparum against 14,000 cell-active compoundsCrowther, Gregory J.; Hillesland, Heidi K.; Keyloun, Katelyn R.; Reid, Molly C.; Lafuente-Monasterio, Maria Jose; Ghidelli-Disse, Sonja; Leonard, Stephen E.; He, Panqing; Jones, Jackson C.; Krahn, Mallory M.; Mo, Jack S.; Dasari, Kartheek S.; Fox, Anna M. W.; Boesche, Markus; El Bakkouri, Majida; Rivas, Kasey L.; Leroy, Didier; Hui, Raymond; Drewes, Gerard; Maly, Dustin J.; Van Voorhis, Wesley C.; Ojo, Kayode K.PLoS One (2016), 11 (3), e0149996/1-e0149996/16CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)In 2010 the identities of thousands of anti-Plasmodium compds. were released publicly to facilitate malaria drug development. Understanding these compds. mechanisms of action, i.e., the specific mol. targets by which they kill the parasite, would further facilitate the drug development process. Given that kinases are promising anti-malaria targets, the authors screened ∼14,000 cell-active compds. for activity against five different protein kinases. Collections of cell-active compds. from GlaxoSmithKline (the ∼13,000-compd. Tres Cantos Antimalarial Set, or TCAMS), St. Jude Children's Research Hospital (260 compds.), and the Medicines for Malaria Venture (the 400-compd. Malaria Box) were screened in biochem. assays of Plasmodium falciparum calcium-dependent protein kinases 1 and 4 (CDPK1 and CDPK4), mitogen-assocd. protein kinase 2 (MAPK2/ MAP2), protein kinase 6 (PK6), and protein kinase 7 (PK7). Novel potent inhibitors (IC50 < 1 μM) were discovered for three of the kinases: CDPK1, CDPK4, and PK6. The PK6 inhibitors are the most potent yet discovered for this enzyme and deserve further scrutiny. Addnl., kinome-wide competition assays revealed a compd. that inhibits CDPK4 with few effects on ∼150 human kinases, and several related compds. that inhibit CDPK1 and CDPK4 yet have limited cytotoxicity to human (HepG2) cells. The authors' data suggest that inhibiting multiple Plasmodium kinase targets without harming human cells is challenging but feasible.
- 14Phuangsawai, O.; Beswick, P.; Ratanabunyong, S.; Tabtimmai, L.; Suphakun, P.; Obounchoey, P.; Srisook, P.; Horata, N.; Chuckowree, I.; Hannongbua, S.; Ward, S. E.; Choowongkomon, K.; Gleeson, M. P. Evaluation of the Anti-Malarial Activity and Cytotoxicity of 2,4-Diamino-Pyrimidine-Based Kinase Inhibitors. Eur. J. Med. Chem. 2016, 124, 896– 905, DOI: 10.1016/j.ejmech.2016.08.05514https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFyms7rM&md5=3c8e425740ebb6387ef42a66ae9bb75eEvaluation of the anti-malarial activity and cytotoxicity of 2,4-diamino-pyrimidine-based kinase inhibitorsPhuangsawai, Oraphan; Beswick, Paul; Ratanabunyong, Siriluk; Tabtimmai, Lueacha; Suphakun, Praphasri; Obounchoey, Phongphat; Srisook, Pimonwan; Horata, Natharinee; Chuckowree, Irina; Hannongbua, Supa; Ward, Simon E.; Choowongkomon, Kiattawee; Gleeson, M. PaulEuropean Journal of Medicinal Chemistry (2016), 124 (), 896-905CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A series of 2,4 diamino-pyrimidines have been identified from an anal. of open access high throughput antimalarial screening data reported by GlaxoSmithKline at the 3D7 and resistant Dd2 strains. SAR expansion has been performed using structural knowledge of the most plausible parasite target. Seventeen new analogs have been synthesized and tested against the resistant K1 strain of Plasmodium falciparum (Pf). The cytotoxicity of the compds. was assessed in Vero and A549 cells and their selectivity towards human kinases including JAK2 and EGFR were undertaken. The authors identified 2 compds. as sub-micromolar inhibitors, with equiv. antimalarial activity to Chloroquine (CQ). Compds. I and II, μM inhibitors of Pf, displayed improved cytotoxicity with weak inhibition of the human kinases.
- 15Toviwek, B.; Phuangsawai, O.; Konsue, A.; Hannongbua, S.; Riley, J.; Mutter, N.; Anderson, M.; Webster, L.; Hallyburton, I.; Read, K. D.; Gleeson, M. P. Preparation, Biological & Cheminformatics-Based Assessment of N2,N4-Diphenylpyrimidine-2,4-Diamine as Potential Kinase-Targeted Antimalarials. Bioorg. Med. Chem. 2021, 46, 116348, DOI: 10.1016/j.bmc.2021.11634815https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFemu7%252FM&md5=cf5c3c887dcbf02d1d8149692fd51ea4Preparation, biological & cheminformatics-based assessment of N2,N4-diphenylpyrimidine-2,4-diamine as potential Kinase-targeted antimalarialsToviwek, Borvornwat; Phuangsawai, Oraphan; Konsue, Adchatawut; Hannongbua, Supa; Riley, Jennifer; Mutter, Nicole; Anderson, Mark; Webster, Lauren; Hallyburton, Irene; Read, Kevin D.; Gleeson, M. PaulBioorganic & Medicinal Chemistry (2021), 46 (), 116348CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Twenty eight new N2,N4-diphenylpyrimidine-2,4-diamines have been prepd. in order to expand our understanding of the anti-malarial SAR of the scaffold. The aim of the study was to make structural modifications to improve the overall potency, selectivity and soly. of the series by varying the anilino groups attached to the 2- and 4-position. We evaluated the activity of the compds. against Plasmodium falciparum (Pf) 3D7, cytotoxicity against HepG2, % inhibition at a panel of 10 human kinases, soly., permeability and lipophilicity, and human and rat in vitro clearance. 11 was identified as a potent anti-malarial with an IC50 of 0.66μM at the 3D7 strain and a selectivity (SI) of ∼ 40 in terms of cytotoxicity against the HepG2 cell line. It also displayed low exptl. logD7.4 (2.27), reasonable soly. (124μg/mL), good metabolic stability, but low permeability. A proteo-chemometric workflow was employed to identify putative Pf targets of the most promising compds. Ligand-based similarity searching of the ChEMBL database led to the identification of most probable human targets. These were then used as input for sequence-based searching of the Pf proteome. Homol. modeling and mol. docking were used to evaluate whether compds. could indeed bind to these targets with valid binding modes. In vitro biol. testing against close human analogs of these targets was subsequently undertaken. This allowed us to identify potential Pf targets and human anti-targets that could be exploited in future development.
- 16Bohmer, M. J.; Wang, J.; Istvan, E. S.; Luth, M. R.; Collins, J. E.; Huttlin, E. L.; Wang, L.; Mittal, N.; Hao, M.; Kwiatkowski, N. P.; Gygi, S. P.; Chakrabarti, R.; Deng, X.; Goldberg, D. E.; Winzeler, E. A.; Gray, N. S.; Chakrabarti, D. Human Polo-like Kinase Inhibitors as Antiplasmodials. ACS Infect. Dis. 2023, 9, 1004– 1021, DOI: 10.1021/acsinfecdis.3c0002516https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXltFSitrc%253D&md5=9c8a807461ba96403b6577fce1712aeeHuman Polo-like Kinase Inhibitors as AntiplasmodialsBohmer, Monica J.; Wang, Jinhua; Istvan, Eva S.; Luth, Madeline R.; Collins, Jennifer E.; Huttlin, Edward L.; Wang, Lushun; Mittal, Nimisha; Hao, Mingfeng; Kwiatkowski, Nicholas P.; Gygi, Steven P.; Chakrabarti, Ratna; Deng, Xianming; Goldberg, Daniel E.; Winzeler, Elizabeth A.; Gray, Nathanael S.; Chakrabarti, DebopamACS Infectious Diseases (2023), 9 (4), 1004-1021CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Protein kinases have proven to be a very productive class of therapeutic targets, and over 90 inhibitors are currently in clin. use primarily for the treatment of cancer. Repurposing these inhibitors as antimalarials could provide an accelerated path to drug development. In this study, we identified BI-2536 (compd. 1), a known potent human polo-like kinase 1 inhibitor, with low nanomolar antiplasmodial activity. Screening of addnl. PLK1 inhibitors revealed further antiplasmodial candidates despite the lack of an obvious orthologue of PLKs in Plasmodium. A subset of these inhibitors was profiled for their in vitro killing profile, and commonalities between the killing rate and inhibition of nuclear replication were noted. A kinase panel screen identified PfNEK3 as a shared target of these PLK1 inhibitors; however, phosphoproteome anal. confirmed distinct signaling pathways were disrupted by two structurally distinct inhibitors, suggesting PfNEK3 may not be the sole target. Genomic anal. of BI-2536-resistant parasites revealed mutations in genes assocd. with the starvation-induced stress response, suggesting BI-2536 may also inhibit an aminoacyl-tRNA synthetase.
- 17Ramsden, N.; Perrin, J.; Ren, Z.; Lee, B. D.; Zinn, N.; Dawson, V. L.; Tam, D.; Bova, M.; Lang, M.; Drewes, G.; Bantscheff, M.; Bard, F.; Dawson, T. M.; Hopf, C. Chemoproteomics-Based Design of Potent LRRK2-Selective Lead Compounds That Attenuate Parkinson’s Disease-Related Toxicity in Human Neurons. ACS Chem. Biol. 2011, 6, 1021– 1028, DOI: 10.1021/cb200241317https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVSrtLzP&md5=9537f562f2097dd244d77c21333c8c47Chemoproteomics-Based Design of Potent LRRK2-Selective Lead Compounds That Attenuate Parkinson's Disease-Related Toxicity in Human NeuronsRamsden, Nigel; Perrin, Jessica; Ren, Zhao; Lee, Byoung Dae; Zinn, Nico; Dawson, Valina L.; Tam, Danny; Bova, Michael; Lang, Manja; Drewes, Gerard; Bantscheff, Marcus; Bard, Frederique; Dawson, Ted M.; Hopf, CarstenACS Chemical Biology (2011), 6 (10), 1021-1028CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Leucine-rich repeat kinase-2 (LRRK2) mutations are the most important cause of familial Parkinson's disease, and non-selective inhibitors are protective in rodent disease models. Because of their poor potency and selectivity, the neuroprotective mechanism of these tool compds. has remained elusive so far, and it is still unknown whether selective LRRK2 inhibition can attenuate mutant LRRK2-dependent toxicity in human neurons. Here, we employ a chemoproteomics strategy to identify potent, selective, and metabolically stable LRRK2 inhibitors. We demonstrate that CZC-25146 (I) prevents mutant LRRK2-induced injury of cultured rodent and human neurons with mid-nanomolar potency. These precise chem. probes further validate this emerging therapeutic strategy. They will enable more detailed studies of LRRK2-dependent signaling and pathogenesis and accelerate drug discovery.
- 18Jester, B. W.; Cox, K. J.; Gaj, A.; Shomin, C. D.; Porter, J. R.; Ghosh, I. A Coiled-Coil Enabled Split-Luciferase Three-Hybrid System: Applied Toward Profiling Inhibitors of Protein Kinases. J. Am. Chem. Soc. 2010, 132, 11727– 11735, DOI: 10.1021/ja104491h18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpsV2mtL0%253D&md5=b6b422ada61cbc678e0156eeb07ed9a9A Coiled-Coil Enabled Split-Luciferase Three-Hybrid System: Applied Toward Profiling Inhibitors of Protein KinasesJester, Benjamin W.; Cox, Kurt J.; Gaj, Alicia; Shomin, Carolyn D.; Porter, Jason R.; Ghosh, IndraneelJournal of the American Chemical Society (2010), 132 (33), 11727-11735CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The 518 protein kinases encoded in the human genome are exquisitely regulated and their aberrant function(s) are often assocd. with human disease. Thus, to advance therapeutics and to probe signal transduction cascades, there is considerable interest in the development of inhibitors that can selectively target protein kinases. However, identifying specific compds. against such a large array of protein kinases is difficult to routinely achieve using traditional activity assays, where purified protein kinases are necessary. Toward a simple, rapid, and practical method for identifying specific inhibitors, the authors describe the development and application of a split-protein methodol. using a coiled-coil-assisted three-hybrid system. In this approach, a protein kinase of interest is attached to the C-terminal fragment of split-firefly luciferase and the coiled-coil Fos, which is specific for the coiled-coil Jun, is attached to the N-terminal fragment. Upon addn. of Jun conjugated to a pan-kinase inhibitor such as staurosporine, a three-hybrid complex was established with concomitant reassembly of the split-luciferase enzyme. An inhibitor can be potentially identified by the commensurate loss in split-luciferase activity by displacement of the modified staurosporine. This new three-hybrid approach is potentially general by testing protein kinases from the different kinase families. To interrogate whether this method allows for screening inhibitors, the authors tested six different protein kinases against a library of 80 known protein kinase inhibitors. Finally, this three-hybrid system can potentially provide a rapid method for structure/function anal. as well as aid in the identification of allosteric inhibitors.
- 19Reininger, L.; Wilkes, J. M.; Bourgade, H.; Miranda-Saavedra, D.; Doerig, C. An Essential Aurora-Related Kinase Transiently Associates with Spindle Pole Bodies during Plasmodium Falciparum Erythrocytic Schizogony. Mol. Microbiol. 2011, 79, 205– 221, DOI: 10.1111/j.1365-2958.2010.07442.x19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFGruro%253D&md5=6b7136888ba61e1d74140e3ef7348216An essential Aurora-related kinase transiently associates with spindle pole bodies during Plasmodium falciparum erythrocytic schizogonyReininger, Luc; Wilkes, Jonathan M.; Bourgade, Helene; Miranda-Saavedra, Diego; Doerig, ChristianMolecular Microbiology (2011), 79 (1), 205-221CODEN: MOMIEE; ISSN:0950-382X. (Wiley-Blackwell)Aurora kinases compose a family of conserved Ser/Thr protein kinases playing essential roles in eukaryotic cell division. To date, Aurora homologues remain uncharacterized in the protozoan phylum Apicompexa. In malaria parasites, the characterization of Aurora kinases may help understand the cell cycle control during erythrocytic schizogony where asynchronous nuclear divisions occur. In this study, we revisited the kinome of Plasmodium falciparum and identified three Aurora-related kinases, Pfark-1, -2, -3. Among these, Pfark-1 is highly conserved in malaria parasites and also appears to be conserved across Apicomplexa. By tagging the endogenous Pfark-1 gene with the green fluorescent protein (GFP) in live parasites, we show that the Pfark-1-GFP protein forms paired dots assocd. with only a subset of nuclei within individual schizonts. Immunofluorescence anal. using an anti-α-tubulin antibody strongly suggests a recruitment of Pfark-1 at duplicated spindle pole bodies at the entry of the M phase of the cell cycle. Unsuccessful attempts at disrupting the Pfark-1 gene with a knockout construct further indicate that Pfark-1 is required for parasite growth in red blood cells. This study provides new insights into the cell cycle control of malaria parasites and reports the importance of Aurora kinases as potential targets for new antimalarials.
- 20Berry, L.; Chen, C.-T.; Reininger, L.; Carvalho, T. G.; El Hajj, H.; Morlon-Guyot, J.; Bordat, Y.; Lebrun, M.; Gubbels, M.-J.; Doerig, C.; Daher, W. The Conserved Apicomplexan Aurora Kinase TgArk3 Is Involved in Endodyogeny, Duplication Rate and Parasite Virulence. Cell. Microbiol. 2016, 18, 1106– 1120, DOI: 10.1111/cmi.1257120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XivVWnt7Y%253D&md5=dcf723b943366896f05adce74df38be8The conserved apicomplexan Aurora kinase TgArk3 is involved in endodyogeny, duplication rate and parasite virulenceBerry, Laurence; Chen, Chun-Ti; Reininger, Luc; Carvalho, Teresa G.; El Hajj, Hiba; Morlon-Guyot, Juliette; Bordat, Yann; Lebrun, Maryse; Gubbels, Marc-Jan; Doerig, Christian; Daher, WassimCellular Microbiology (2016), 18 (8), 1106-1120CODEN: CEMIF5; ISSN:1462-5814. (Wiley-Blackwell)Aurora kinases are eukaryotic serine/threonine protein kinases that regulate key events assocd. with chromatin condensation, centrosome and spindle function and cytokinesis. Elucidating the roles of Aurora kinases in apicomplexan parasites is crucial to understand the cell cycle control during Plasmodium schizogony or Toxoplasma endodyogeny. Here, the authors report on the localization of two previously uncharacterized Toxoplasma Aurora-related kinases (Ark2 and Ark3) in tachyzoites and of the uncharacterized Ark3 ortholog in Plasmodium falciparum erythrocytic stages. In Toxoplasma gondii, TgArk2 and TgArk3 conc. at specific subcellular structures linked to parasite division: the mitotic spindle and intranuclear mitotic structures (TgArk2), and the outer core of the centrosome and the budding daughter cells cytoskeleton (TgArk3). By tagging the endogenous PfArk3 gene with the green fluorescent protein in live parasites, PfArk3 protein expression was shown to peak late in schizogony and localize at the periphery of budding schizonts. Disruption of the TgArk2 gene reveals no essential function for tachyzoite propagation in vitro, which is surprising giving that the P. falciparum and P. berghei orthologs are essential for erythrocyte schizogony. In contrast, knock-down of TgArk3 protein results in pronounced defects in parasite division and a major growth deficiency. TgArk3-depleted parasites display several defects, such as reduced parasite growth rate, delayed egress and parasite duplication, defect in rosette formation, reduced parasite size and invasion efficiency and lack of virulence in mice. The authors' study provides new insights into cell cycle control in Toxoplasma and malaria parasites and highlights Aurora kinase 3 as potential drug target.
- 21Patel, G.; Roncal, N. E.; Lee, P. J.; Leed, S. E.; Erath, J.; Rodriguez, A.; Sciotti, R. J.; Pollastri, M. P. Repurposing Human Aurora Kinase Inhibitors as Leads for Anti-Protozoan Drug Discovery. Med. Chem. Commun. 2014, 5, 655– 658, DOI: 10.1039/C4MD00045EThere is no corresponding record for this reference.
- 22Morahan, B. J.; Abrie, C.; Al-Hasani, K.; Batty, M. B.; Corey, V.; Cowell, A. N.; Niemand, J.; Winzeler, E. A.; Birkholtz, L.-M.; Doerig, C.; Garcia-Bustos, J. F. Human Aurora Kinase Inhibitor Hesperadin Reveals Epistatic Interaction between Plasmodium Falciparum PfArk1 and PfNek1 Kinases. Commun. Biol. 2020, 3, 701, DOI: 10.1038/s42003-020-01424-z22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXis12isb%252FP&md5=8f36349abe695fe5b2e7c10e774b3309Human Aurora kinase inhibitor Hesperadin reveals epistatic interaction between Plasmodium falciparum PfArk1 and PfNek1 kinasesMorahan, Belinda J.; Abrie, Clarissa; Al-Hasani, Keith; Batty, Mitchell B.; Corey, Victoria; Cowell, Anne N.; Niemand, Jandeli; Winzeler, Elizabeth A.; Birkholtz, Lyn-Marie; Doerig, Christian; Garcia-Bustos, Jose F.Communications Biology (2020), 3 (1), 701CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)Abstr.: Mitosis has been validated by numerous anti-cancer drugs as being a druggable process, and selective inhibition of parasite proliferation provides an obvious opportunity for therapeutic intervention against malaria. Mitosis is controlled through the interplay between several protein kinases and phosphatases. We show here that inhibitors of human mitotic kinases belonging to the Aurora family inhibit P. falciparum proliferation in vitro with various potencies, and that a genetic selection for mutant parasites resistant to one of the drugs, Hesperadin, identifies a resistance mechanism mediated by a member of a different kinase family, PfNek1 (PF3D7_1228300). Intriguingly, loss of PfNek1 catalytic activity provides protection against drug action. This points to an undescribed functional interaction between Ark and Nek kinases and shows that existing inhibitors can be used to validate addnl. essential and druggable kinase functions in the parasite.
- 23Lye, Y. M.; Chan, M.; Sim, T.-S. Pfnek3: An Atypical Activator of a MAP Kinase in Plasmodium Falciparum. FEBS Lett. 2006, 580, 6083– 6092, DOI: 10.1016/j.febslet.2006.10.00323https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtFOhsr7E&md5=bfdbb15a199ff5a926db887b0c908c5ePfnek3: An atypical activator of a MAP kinase in Plasmodium falciparumLye, Yu Min; Chan, Maurice; Sim, Tiow-SuanFEBS Letters (2006), 580 (26), 6083-6092CODEN: FEBLAL; ISSN:0014-5793. (Elsevier B.V.)The canonical mitogen-activated protein kinase (MAPK) signal cascade was previously suggested to be atypical in the malaria parasite. This raises queries on the existence of alternative mediators of plasmodial MAPK pathways. This study describes, Pfnek3, a malarial protein kinase belonging to the NIMA (Never in Mitosis, Aspergillus) family. Endogenous Pfnek3 is expressed during late asexual to gametocyte stages and lacks some classical protein kinase sequence motifs. Moreover, Pfnek3 is phylogenetically distant from mammalian NIMA-kinases. Recombinant Pfnek3 was able to phosphorylate and stimulate a malarial MAPK (Pfmap2). Contrastingly, this was not obsd. with two other kinases, Pfmap1 and human MAPK1, suggesting that the Pfnek3-Pfmap2 interaction may be specific for Pfmap2 regulation. In summary, our data reveal a malarial NIMA-kinase with the potential to regulate a MAPK. Possessing biochem. properties divergent from classical mammalian NIMA-kinases, Pfnek3 could potentially be an attractive target for parasite-selective anti-malarials.
- 24Low, H.; Lye, Y. M.; Sim, T.-S. Pfnek3 Functions as an Atypical MAPKK in Plasmodium Falciparum. Biochem. Biophys. Res. Commun. 2007, 361, 439– 444, DOI: 10.1016/j.bbrc.2007.07.04724https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXosF2jsLo%253D&md5=9f2fe3b56f19b67bb316d700456ad155Pfnek3 functions as an atypical MAPKK in Plasmodium falciparumLow, Huiyu; Lye, Yu Min; Sim, Tiow-SuanBiochemical and Biophysical Research Communications (2007), 361 (2), 439-444CODEN: BBRCA9; ISSN:0006-291X. (Elsevier)Eukaryotes generally rely on signal transduction by mitogen-activated protein kinases (MAPKs) for activating their regulatory pathways. However, the presence of a complete MAPK cascade in Plasmodium falciparum is debatable because a search of the entire genome did not portray known MAPK kinase (MAPKK) sequences. Via homol. PCR expts., only two copies of plasmodial MAPK homologs (Pfmap1 and Pfmap2) have been identified but their upstream activators remain unknown. In an earlier expt., Pfnek3 was found to be an unusual activator of Pfmap2 in vitro expts., despite its mol. identity as a malarial protein kinase from the NIMA (Never in Mitosis, Aspergillus) family. In this study, the role of Pfnek3 as a likely upstream MAPKK is defined through mol. and biochem. characterization. Since a previous report proposes a TSH motif as an activation site of Pfmap2, its site-directed mutants, T290A, S291A, and H292K were constructed to elucidate the involvement of Pfnek3 in phosphorylating and activating Pfmap2 in a battery of kinase assays. The results suggested that residue T290 is the site of phosphorylation by Pfnek3. This supposition was further supported by liq. chromatog. mass spectrometry. Although P. falciparum does not appear to possess a conventional MAPK cascade, they may rely on other kinases such as Pfnek3 to carry out similar phosphorylation to activate its signaling pathways.
- 25Ong, H. W.; Truong, A.; Kwarcinski, F.; de Silva, C.; Avalani, K.; Havener, T. M.; Chirgwin, M.; Galal, K. A.; Willis, C.; Krämer, A.; Liu, S.; Knapp, S.; Derbyshire, E. R.; Zutshi, R.; Drewry, D. H. Discovery of Potent Plasmodium Falciparum Protein Kinase 6 (PfPK6) Inhibitors with a Type II Inhibitor Pharmacophore. Eur. J. Med. Chem. 2023, 249, 115043, DOI: 10.1016/j.ejmech.2022.11504325https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXisV2ntL8%253D&md5=a6d54605fec3347c4cd57a46f397cdb1Discovery of potent Plasmodium falciparum protein kinase 6 (PfPK6) inhibitors with a type II inhibitor pharmacophoreOng, Han Wee; Truong, Anna; Kwarcinski, Frank; de Silva, Chandi; Avalani, Krisha; Havener, Tammy M.; Chirgwin, Michael; Galal, Kareem A.; Willis, Caleb; Kramer, Andreas; Liu, Shubin; Knapp, Stefan; Derbyshire, Emily R.; Zutshi, Reena; Drewry, David H.European Journal of Medicinal Chemistry (2023), 249 (), 115043CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)Malaria is a devastating disease that causes significant global morbidity and mortality. The rise of drug resistance against artemisinin-based combination therapy demonstrates the necessity to develop alternative antimalarials with novel mechanisms of action. We report the discovery of Ki8751 as an inhibitor of essential kinase PfPK6. 79 derivs. were designed, synthesized and evaluated for PfPK6 inhibition and antiplasmodial activity. Using group efficiency analyses, we established the importance of key groups on the scaffold consistent with a type II inhibitor pharmacophore. We highlight modifications on the tail group that contribute to antiplasmodial activity, cumulating in the discovery of compd. 67, a PfPK6 inhibitor (IC50 = 13 nM) active against the P. falciparum blood stage (EC50 = 160 nM), and compd. 79, a PfPK6 inhibitor (IC50 < 5 nM) with dual-stage antiplasmodial activity against P. falciparum blood stage (EC50 = 39 nM) and against P. berghei liver stage (EC50 = 220 nM).
- 26Philip, N.; Haystead, T. A. Characterization of a UBC13 Kinase in Plasmodium Falciparum. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 7845– 7850, DOI: 10.1073/pnas.061160110426https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtFCit7g%253D&md5=952f7a70bf86207c61596d9cd2bd8bcfCharacterization of a UBC13 kinase in Plasmodium falciparumPhilip, Nisha; Haystead, Timothy A.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (19), 7845-7850CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Protein kinases are generally recognized as attractive drug targets to treat a variety of human diseases. Recent anal. of the Plasmodium falciparum kinome identified several kinases that are entirely unique to Plasmodium species. The specific functions and targets of most of these enzymes remain largely unknown. Here, we have identified a P. falciparum kinase (PfPK9/PF13_0085 ORF) that does not cluster with any of the typical eukaryotic protein kinases. PfPK9 protein expression was induced ≈18 h after red blood cell infection, and was mainly localized to the parasitophorous vacuolar membrane as well as the cytosol. Recombinant PfPK9 autophosphorylated in vitro and specifically phosphorylated the exogenous substrate histone H1, indicating that it is catalytically active. Phosphopeptide mapping studies showed that autophosphorylation occurred at three residues: T082, T265, and T269. We identified a P. falciparum homolog of the E2 ubiquitin-conjugating enzyme 13 (UBC13) as an endogenous substrate for PfPK9. PfPK9 phosphorylated UBC13 at S106, a highly conserved residue among eukaryotic E2s, and suppressed its ubiquitin-conjugating activity. Our findings not only describe a previously uncharacterized Plasmodium kinase and its likely in vivo target, but also suggest that modulation of UBC13 activity by phosphorylation may be a common regulatory mechanism in eukaryotes.
- 27Raphemot, R.; Eubanks, A. L.; Toro-Moreno, M.; Geiger, R. A.; Hughes, P. F.; Lu, K.-Y.; Haystead, T. A. J.; Derbyshire, E. R. Plasmodium PK9 Inhibitors Promote Growth of Liver-Stage Parasites. Cell Chem. Biol. 2019, 26, 411– 419, DOI: 10.1016/j.chembiol.2018.11.00327https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXntl2q&md5=a9179165387521f8a7f77ba0a532e25aPlasmodium PK9 Inhibitors Promote Growth of Liver-Stage ParasitesRaphemot, Rene; Eubanks, Amber Leigh; Toro-Moreno, Maria; Geiger, Rechel Anne; Hughes, Philip Floyd; Lu, Kuan-Yi; Haystead, Timothy Arthur James; Derbyshire, Emily RoseCell Chemical Biology (2019), 26 (3), 411-419.e7CODEN: CCBEBM; ISSN:2451-9448. (Cell Press)There is a scarcity of pharmacol. tools to interrogate protein kinase function in Plasmodium parasites, the causative agent of malaria. Among Plasmodiums protein kinases, those characterized as atypical represent attractive drug targets as they lack sequence similarity to human proteins. Here, we describe takinib as a small mol. to bind the atypical P. falciparum protein kinase 9 (PfPK9). PfPK9 phosphorylates the Plasmodium E2 ubiquitin-conjugating enzyme PfUBC13, which mediates K63-linkage-specific polyubiquitination. Takinib is a potent human TAK1 inhibitor, thus we developed the Plasmodium-selective takinib analog HS220. We demonstrate that takinib and HS220 decrease K63-linked ubiquitination in P. falciparum, suggesting PfPK9 inhibition in cells. Takinib and HS220 induce a unique phenotype where parasite size in hepatocytes increases, yet high compd. concns. decrease the no. of parasites. Our studies highlight the role of PK9 in regulating parasite development and the potential of targeting Plasmodium kinases for malaria control.
- 28Maneekesorn, S.; Knuepfer, E.; Green, J. L.; Prommana, P.; Uthaipibull, C.; Srichairatanakool, S.; Holder, A. A. Deletion of Plasmodium Falciparum Ubc13 Increases Parasite Sensitivity to the Mutagen, Methyl Methanesulfonate and Dihydroartemisinin. Sci. Rep. 2021, 11, 21791, DOI: 10.1038/s41598-021-01267-628https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVegt73P&md5=f4c0e790f9fd30975e686c4452a60c73Deletion of Plasmodium falciparum ubc13 increases parasite sensitivity to the mutagen, methyl methanesulfonate and dihydroartemisininManeekesorn, Supawadee; Knuepfer, Ellen; Green, Judith L.; Prommana, Parichat; Uthaipibull, Chairat; Srichairatanakool, Somdet; Holder, Anthony A.Scientific Reports (2021), 11 (1), 21791CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)The inducible Di-Cre system was used to delete the putative ubiquitin-conjugating enzyme 13 gene (ubc13) of Plasmodium falciparum to study its role in ubiquitylation and the functional consequence during the parasite asexual blood stage. Deletion resulted in a significant redn. of parasite growth in vitro, reduced ubiquitylation of the Lys63 residue of ubiquitin attached to protein substrates, and an increased sensitivity of the parasite to both the mutagen, Me methanesulfonate and the antimalarial drug dihydroartemisinin (DHA), but not chloroquine. The parasite was also sensitive to the UBC13 inhibitor NSC697923. The data suggest that this gene does code for an ubiquitin conjugating enzyme responsible for K63 ubiquitylation, which is important in DNA repair pathways as was previously demonstrated in other organisms. The increased parasite sensitivity to DHA in the absence of ubc13 function indicates that DHA may act primarily through this pathway and that inhibitors of UBC13 may both enhance the efficacy of this antimalarial drug and directly inhibit parasite growth.
- 29Kumar, A.; Vaid, A.; Syin, C.; Sharma, P. PfPKB, a Novel Protein Kinase B-like Enzyme from Plasmodium Falciparum I. Identification, Characterization, and Possible Role in Parasite Development. J. Biol. Chem. 2004, 279, 24255– 24264, DOI: 10.1074/jbc.M31285520029https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksV2isbk%253D&md5=2bbbbaa7b0b76a037173eb41d298f09cPfPKB, a Novel Protein Kinase B-like Enzyme from Plasmodium falciparum: I. Identification, characterization, and possible role in parasite developmentKumar, Amit; Vaid, Ankush; Syin, Chiang; Sharma, PushkarJournal of Biological Chemistry (2004), 279 (23), 24255-24264CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Extracellular signals control various important functions of a eukaryotic cell, which is often achieved by regulating a battery of protein kinases and phosphatases. Protein Kinase B (PKB) is an important member of the phosphatidylinositol 3-kinase-dependent signaling pathways in several eukaryotes, but the role of PKB in protozoan parasites is not known. We have identified a protein kinase B homolog in Plasmodium falciparum (PfPKB) that is expressed mainly in the schizonts and merozoites. Even though PfPKB shares high sequence homol. with PKB catalytic domain, it lacks a pleckstrin homol. domain typically found at the N terminus of the mammalian enzyme. Biochem. studies performed to understand the mechanism of PfPKB catalytic activation suggested that (i) its activation is dependent on autophosphorylation of a serine residue (Ser-271) in its activation loop region and (ii) PfPKB has an unusual N-terminal region that was found to neg. regulate its catalytic activity. We also identified an inhibitor of PfPKB activity that also inhibits P. falciparum growth, suggesting that this enzyme may be important for the development of the parasite.
- 30Baum, J.; Richard, D.; Healer, J.; Rug, M.; Krnajski, Z.; Gilberger, T.-W.; Green, J. L.; Holder, A. A.; Cowman, A. F. A Conserved Molecular Motor Drives Cell Invasion and Gliding Motility across Malaria Life Cycle Stages and Other Apicomplexan Parasites. J. Biol. Chem. 2006, 281, 5197– 5208, DOI: 10.1074/jbc.M50980720030https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhs1ektL0%253D&md5=c5e9deb26afdf2281cfc259d354380d8A Conserved Molecular Motor Drives Cell Invasion and Gliding Motility across Malaria Life Cycle Stages and Other Apicomplexan ParasitesBaum, Jake; Richard, Dave; Healer, Julie; Rug, Melanie; Krnajski, Zita; Gilberger, Tim-Wolf; Green, Judith L.; Holder, Anthony A.; Cowman, Alan F.Journal of Biological Chemistry (2006), 281 (8), 5197-5208CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Apicomplexan parasites constitute one of the most significant groups of pathogens infecting humans and animals. The liver stage sporozoites of Plasmodium spp. and tachyzoites of Toxoplasma gondii, the causative agents of malaria and toxoplasmosis, resp., use a unique mode of locomotion termed gliding motility to invade host cells and cross cell substrates. This amoeboid-like movement uses a parasite adhesin from the thrombospondin-related anonymous protein (TRAP) family and a set of proteins linking the extracellular adhesin, via an actin-myosin motor, to the inner membrane complex. The Plasmodium blood stage merozoite, however, does not exhibit gliding motility. Here we show that homologs of the key proteins that make up the motor complex, including the recently identified glideosome-assocd. proteins 45 and 50 (GAP45 and GAP50), are present in P. falciparum merozoites and appear to function in erythrocyte invasion. Furthermore, we identify a merozoite TRAP homolog, termed MTRAP, a micronemal protein that shares key features with TRAP, including a thrombospondin repeat domain, a putative rhomboid-protease cleavage site, and a cytoplasmic tail that, in vitro, binds the actin-binding protein aldolase. Anal. of other parasite genomes shows that the components of this motor complex are conserved across diverse Apicomplexan genera. Conservation of the motor complex suggests that a common mol. mechanism underlies all Apicomplexan motility, which, given its unique properties, highlights a no. of novel targets for drug intervention to treat major diseases of humans and livestock.
- 31Jones, M. L.; Kitson, E. L.; Rayner, J. C. Plasmodium Falciparum Erythrocyte Invasion: A Conserved Myosin Associated Complex. Mol. Biochem. Parasitol. 2006, 147, 74– 84, DOI: 10.1016/j.molbiopara.2006.01.00931https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XjvVWmsrk%253D&md5=3e7db83cf98a9fc7081650857af81ad9Plasmodium falciparum erythrocyte invasion: A conserved myosin associated complexJones, Matthew L.; Kitson, Erika L.; Rayner, Julian C.Molecular & Biochemical Parasitology (2006), 147 (1), 74-84CODEN: MBIPDP; ISSN:0166-6851. (Elsevier Ltd.)Host cell invasion by apicomplexan parasites is powered by an actin/myosin motor complex that has been most thoroughly described in Toxoplasma gondii tachyzoites. In T. gondii, two inner membrane complex (IMC) proteins, the peripheral protein TgGAP45 and the transmembrane protein TgGAP50, form a complex with the myosin, TgMyoA, and its light chain, TgMLC1. This complex, referred to as the glideosome, anchors the invasion motor to the IMC. We have identified and characterized orthologues of TgMLC1, TgGAP45 and TgGAP50 in blood-stages of the major human pathogen Plasmodium falciparum, supporting the idea that the same basic complex drives host cell invasion across the apicomplexan phylum. The P. falciparum glideosome proteins are transcribed, expressed and localized in a manner consistent with a role in erythrocyte invasion. Furthermore, PfMyoA interacts with PfMTIP through broadly conserved mechanisms described in other eukaryotes, and forms a complex with PfGAP45 and PfGAP50 in late schizonts and merozoites. P. falciparum is known to use multiple alternative invasion pathways to enter erythrocytes, hampering vaccine development efforts targeting erythrocyte invasion. Our data suggests that the same invasion motor underpins all alternative invasion pathways, making it an attractive target for the development of novel intervention strategies.
- 32Thomas, D. C.; Ahmed, A.; Gilberger, T. W.; Sharma, P. Regulation of Plasmodium Falciparum Glideosome Associated Protein 45 (PfGAP45) Phosphorylation. PLoS One 2012, 7, e35855 DOI: 10.1371/journal.pone.003585532https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmvFOntrc%253D&md5=69fc93944aa2521edfc2e6f5abb3cb42Regulation of Plasmodium falciparum Glideosome Associated Protein 45 (PfGAP45) phosphorylationThomas, Divya Catherine; Ahmed, Anwar; Gilberger, Tim Wolf; Sharma, PushkarPLoS One (2012), 7 (4), e35855CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)The actomyosin motor complex of the glideosome provides the force needed by apicomplexan parasites such as Toxoplasma gondii (Tg) and Plasmodium falciparum (Pf) to invade their host cells and for gliding motility of their motile forms. Glideosome Assocd. Protein 45 (PfGAP45) is an essential component of the glideosome complex as it facilitates anchoring and effective functioning of the motor. Dissection of events that regulate PfGAP45 may provide insights into how the motor and the glideosome operate. We found that PfGAP45 is phosphorylated in response to Phospholipase C (PLC) and calcium signaling. It is phosphorylated by P. falciparum kinases Protein Kinase B (PfPKB) and Calcium Dependent Protein Kinase 1 (PfCDPK1), which are calcium dependent enzymes, at S89, S103 and S149. The Phospholipase C pathway influenced the phosphorylation of S103 and S149. The phosphorylation of PfGAP45 at these sites is differentially regulated during parasite development. The localization of PfGAP45 and its assocn. may be independent of the phosphorylation of these sites. PfGAP45 regulation in response to calcium fits in well with the previously described role of calcium in host cell invasion by malaria parasite.
- 33Vaid, A.; Thomas, D. C.; Sharma, P. Role of Ca2+/Calmodulin-PfPKB Signaling Pathway in Erythrocyte Invasion by Plasmodium Falciparum. J. Biol. Chem. 2008, 283, 5589– 5597, DOI: 10.1074/jbc.M70846520033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXit1Omt7g%253D&md5=0a854ef8a85f8a080a74ec4b20c17ff8Role of Ca2+/calmodulin-PfPKB signaling pathway in erythrocyte invasion by Plasmodium falciparumVaid, Ankush; Thomas, Divya C.; Sharma, PushkarJournal of Biological Chemistry (2008), 283 (9), 5589-5597CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Mol. mechanisms by which signaling pathways operate in the malaria parasite and control its development are promiscuous. Recently, we reported the identification of a signaling pathway in Plasmodium falciparum, which involves activation of protein kinase B-like enzyme (PfPKB) by calcium/calmodulin. Studies carried out to elucidate the function of this pathway suggested that it may be important for erythrocyte invasion. Blocking the function of the upstream activators of this pathway, calmodulin and phospholipase C, resulted in impaired invasion. To evaluate if this signaling cascade controls invasion by regulating PfPKB, inhibitors against this kinase were developed. PfPKB inhibitors dramatically reduced the ability of the parasite to invade erythrocytes. Furthermore, we demonstrate that PfPKB assocs. with actin-myosin motor and phosphorylates PfGAP45 (glideosome-assocd. protein 45), one of the important components of the motor complex, which may help explain its role in erythrocyte invasion.
- 34Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S. A. A.; Ballard, A. J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A. W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly Accurate Protein Structure Prediction with AlphaFold. Nature 2021, 596, 583– 589, DOI: 10.1038/s41586-021-03819-234https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaktrrL&md5=25964ab1157cd5b74a437333dd86650dHighly accurate protein structure prediction with AlphaFoldJumper, John; Evans, Richard; Pritzel, Alexander; Green, Tim; Figurnov, Michael; Ronneberger, Olaf; Tunyasuvunakool, Kathryn; Bates, Russ; Zidek, Augustin; Potapenko, Anna; Bridgland, Alex; Meyer, Clemens; Kohl, Simon A. A.; Ballard, Andrew J.; Cowie, Andrew; Romera-Paredes, Bernardino; Nikolov, Stanislav; Jain, Rishub; Adler, Jonas; Back, Trevor; Petersen, Stig; Reiman, David; Clancy, Ellen; Zielinski, Michal; Steinegger, Martin; Pacholska, Michalina; Berghammer, Tamas; Bodenstein, Sebastian; Silver, David; Vinyals, Oriol; Senior, Andrew W.; Kavukcuoglu, Koray; Kohli, Pushmeet; Hassabis, DemisNature (London, United Kingdom) (2021), 596 (7873), 583-589CODEN: NATUAS; ISSN:0028-0836. (Nature Portfolio)Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous exptl. effort, the structures of around 100,000 unique proteins have been detd., but this represents a small fraction of the billions of known protein sequences. Structural coverage is bottlenecked by the months to years of painstaking effort required to det. a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence-the structure prediction component of the 'protein folding problem'-has been an important open research problem for more than 50 years. Despite recent progress, existing methods fall far short of at. accuracy, esp. when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with at. accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Crit. Assessment of protein Structure Prediction (CASP14), demonstrating accuracy competitive with exptl. structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates phys. and biol. knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm.
- 35Varadi, M.; Anyango, S.; Deshpande, M.; Nair, S.; Natassia, C.; Yordanova, G.; Yuan, D.; Stroe, O.; Wood, G.; Laydon, A.; Žídek, A.; Green, T.; Tunyasuvunakool, K.; Petersen, S.; Jumper, J.; Clancy, E.; Green, R.; Vora, A.; Lutfi, M.; Figurnov, M.; Cowie, A.; Hobbs, N.; Kohli, P.; Kleywegt, G.; Birney, E.; Hassabis, D.; Velankar, S. AlphaFold Protein Structure Database: Massively Expanding the Structural Coverage of Protein-Sequence Space with High-Accuracy Models. Nucleic Acids Res. 2022, 50, D439– D444, DOI: 10.1093/nar/gkab106135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1Churw%253D&md5=1e36970ae4b901a4acd62fd2d8ff23bfAlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy modelsVaradi, Mihaly; Anyango, Stephen; Deshpande, Mandar; Nair, Sreenath; Natassia, Cindy; Yordanova, Galabina; Yuan, David; Stroe, Oana; Wood, Gemma; Laydon, Agata; Zidek, Augustin; Green, Tim; Tunyasuvunakool, Kathryn; Petersen, Stig; Jumper, John; Clancy, Ellen; Green, Richard; Vora, Ankur; Lutfi, Mira; Figurnov, Michael; Cowie, Andrew; Hobbs, Nicole; Kohli, Pushmeet; Kleywegt, Gerard; Birney, Ewan; Hassabis, Demis; Velankar, SameerNucleic Acids Research (2022), 50 (D1), D439-D444CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)The AlphaFold Protein Structure Database is an openly accessible, extensive database of high-accuracy protein-structure predictions. Powered by AlphaFold v2.0 of DeepMind, it has enabled an unprecedented expansion of the structural coverage of the known protein-sequence space. AlphaFold DB provides programmatic access to and interactive visualization of predicted at. coordinates, per-residue and pairwise model-confidence ests. and predicted aligned errors. The initial release of AlphaFold DB contains over 360,000 predicted structures across 21 model-organism proteomes, which will soon be expanded to cover most of the (over 100 million) representative sequences from the UniRef90 data set.
- 36van Linden, O. P. J.; Kooistra, A. J.; Leurs, R.; de Esch, I. J. P.; de Graaf, C. KLIFS: A Knowledge-Based Structural Database To Navigate Kinase–Ligand Interaction Space. J. Med. Chem. 2014, 57, 249– 277, DOI: 10.1021/jm400378w36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1Ojur7P&md5=8519942e1703ba04f64797e07df4b712KLIFS: A Knowledge-Based Structural Database To Navigate Kinase-Ligand Interaction Spacevan Linden, Oscar P. J.; Kooistra, Albert J.; Leurs, Rob; de Esch, Iwan J. P.; de Graaf, ChrisJournal of Medicinal Chemistry (2014), 57 (2), 249-277CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Protein kinases regulate the majority of signal transduction pathways in cells and have become important targets for the development of designer drugs. We present a systematic anal. of kinase-ligand interactions in all regions of the catalytic cleft of all 1252 human kinase-ligand cocrystal structures present in the Protein Data Bank (PDB). The kinase-ligand interaction fingerprints and structure database (KLIFS) contains a consistent alignment of 85 kinase ligand binding site residues that enables the identification of family specific interaction features and classification of ligands according to their binding modes. We illustrate how systematic mining of kinase-ligand interaction space gives new insights into how conserved and selective kinase interaction hot spots can accommodate the large diversity of chem. scaffolds in kinase ligands. These analyses lead to an improved understanding of the structural requirements of kinase binding that will be useful in ligand discovery and design studies.
- 37Lategahn, J.; Tumbrink, H. L.; Schultz-Fademrecht, C.; Heimsoeth, A.; Werr, L.; Niggenaber, J.; Keul, M.; Parmaksiz, F.; Baumann, M.; Menninger, S.; Zent, E.; Landel, I.; Weisner, J.; Jeyakumar, K.; Heyden, L.; Russ, N.; Müller, F.; Lorenz, C.; Brägelmann, J.; Spille, I.; Grabe, T.; Müller, M. P.; Heuckmann, J. M.; Klebl, B. M.; Nussbaumer, P.; Sos, M. L.; Rauh, D. Insight into Targeting Exon20 Insertion Mutations of the Epidermal Growth Factor Receptor with Wild Type-Sparing Inhibitors. J. Med. Chem. 2022, 65, 6643– 6655, DOI: 10.1021/acs.jmedchem.1c0208037https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFems7vO&md5=242416ee3cac2aa17a2536bb8e8670d0Insight into Targeting Exon20 Insertion Mutations of the Epidermal Growth Factor Receptor with Wild Type-Sparing InhibitorsLategahn, Jonas; Tumbrink, Hannah L.; Schultz-Fademrecht, Carsten; Heimsoeth, Alena; Werr, Lisa; Niggenaber, Janina; Keul, Marina; Parmaksiz, Fatma; Baumann, Matthias; Menninger, Sascha; Zent, Eldar; Landel, Ina; Weisner, Joern; Jeyakumar, Kirujan; Heyden, Leonie; Russ, Nicole; Mueller, Fabienne; Lorenz, Carina; Braegelmann, Johannes; Spille, Inga; Grabe, Tobias; Mueller, Matthias P.; Heuckmann, Johannes M.; Klebl, Bert M.; Nussbaumer, Peter; Sos, Martin L.; Rauh, DanielJournal of Medicinal Chemistry (2022), 65 (9), 6643-6655CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Despite the clin. efficacy of epidermal growth factor receptor (EGFR) inhibitors, a subset of patients with non-small cell lung cancer displays insertion mutations in exon20 in EGFR and Her2 with limited treatment options. Here, we present the development and characterization of the novel covalent inhibitors LDC8201 and LDC0496 based on a 1H-pyrrolo[2,3-b]pyridine scaffold. They exhibited intense inhibitory potency toward EGFR and Her2 exon20 insertion mutations as well as selectivity over wild type EGFR and within the kinome. Complex crystal structures with the inhibitors and biochem. and cellular on-target activity document their favorable binding characteristics. Ultimately, we obsd. tumor shrinkage in mice engrafted with patient-derived EGFR-H773_V774insNPH mutant cells during treatment with LDC8201. Together, these results highlight the potential of covalent pyrrolopyridines as inhibitors to target exon20 insertion mutations.
- 38Alam, M.; Beevers, R. E.; Ceska, T.; Davenport, R. J.; Dickson, K. M.; Fortunato, M.; Gowers, L.; Haughan, A. F.; James, L. A.; Jones, M. W.; Kinsella, N.; Lowe, C.; Meissner, J. W. G.; Nicolas, A. L.; Perry, B. G.; Phillips, D. J.; Pitt, W. R.; Platt, A.; Ratcliffe, A. J.; Sharpe, A.; Tait, L. J. Synthesis and SAR of Aminopyrimidines as Novel C-Jun N-Terminal Kinase (JNK) Inhibitors. Bioorg. Med. Chem. Lett. 2007, 17, 3463– 3467, DOI: 10.1016/j.bmcl.2007.03.07838https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtValsb4%253D&md5=15d7fb1836a7a11eb94d57a4e1a4265aSynthesis and SAR of aminopyrimidines as novel c-Jun N-terminal kinase (JNK) inhibitorsAlam, Mahbub; Beevers, Rebekah E.; Ceska, Tom; Davenport, Richard J.; Dickson, Karen M.; Fortunato, Mara; Gowers, Lewis; Haughan, Alan F.; James, Lynwen A.; Jones, Mark W.; Kinsella, Natasha; Lowe, Christopher; Meissner, Johannes W. G.; Nicolas, Anne-Lise; Perry, Benjamin G.; Phillips, David J.; Pitt, William R.; Platt, Adam; Ratcliffe, Andrew J.; Sharpe, Andrew; Tait, Laura J.Bioorganic & Medicinal Chemistry Letters (2007), 17 (12), 3463-3467CODEN: BMCLE8; ISSN:0960-894X. (Elsevier Ltd.)The development of a series of aminopyrimidines, e.g., I, as inhibitors of c-Jun N-terminal kinases is described. The synthesis, in vitro inhibitory values for JNK1, JNK2 and CDK2, and the in vitro inhibitory value for a c-Jun cellular assay were discussed.
- 39Lange, A.; Günther, M.; Büttner, F. M.; Zimmermann, M. O.; Heidrich, J.; Hennig, S.; Zahn, S.; Schall, C.; Sievers-Engler, A.; Ansideri, F.; Koch, P.; Laemmerhofer, M.; Stehle, T.; Laufer, S. A.; Boeckler, F. M. Targeting the Gatekeeper MET146 of C-Jun N-Terminal Kinase 3 Induces a Bivalent Halogen/Chalcogen Bond. J. Am. Chem. Soc. 2015, 137, 14640– 14652, DOI: 10.1021/jacs.5b0709039https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslegt73O&md5=d2205de85d5be4dc3f97d008ed838251Targeting the Gatekeeper MET146 of C-Jun N-Terminal Kinase 3 Induces a Bivalent Halogen/Chalcogen BondLange, Andreas; Guenther, Marcel; Buettner, Felix Michael; Zimmermann, Markus O.; Heidrich, Johannes; Hennig, Susanne; Zahn, Stefan; Schall, Christoph; Sievers-Engler, Adrian; Ansideri, Francesco; Koch, Pierre; Laemmerhofer, Michael; Stehle, Thilo; Laufer, Stefan A.; Boeckler, Frank M.Journal of the American Chemical Society (2015), 137 (46), 14640-14652CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We target the gatekeeper MET146 of c-Jun N-terminal kinase 3 (JNK3) to exemplify the applicability of X···S halogen bonds in mol. design using computational, synthetic, structural and biophys. techniques. In a designed series of aminopyrimidine-based inhibitors, we unexpectedly encounter a plateau of affinity. Compared to their QM-calcd. interaction energies, particularly bromine and iodine fail to reach the full potential according to the size of their σ-hole. Instead, mutation of the gatekeeper residue into leucine, alanine, or threonine reveals that the heavier halides can significantly influence selectivity in the human kinome. Thus, we demonstrate that, although the choice of halogen may not always increase affinity, it can still be relevant for inducing selectivity. Detg. the crystal structure of the iodine deriv. in complex with JNK3 (4X21) reveals an unusual bivalent halogen/chalcogen bond donated by the ligand and the back-pocket residue MET115. Incipient repulsion from the too short halogen bond increases the flexibility of Cε of MET146, whereas the rest of the residue fails to adapt being fixed by the chalcogen bond. This effect can be useful to induce selectivity, as the necessary combination of methionine residues only occurs in 9.3% of human kinases, while methionine is the predominant gatekeeper (39%).
- 40Hole, A. J.; Baumli, S.; Shao, H.; Shi, S.; Huang, S.; Pepper, C.; Fischer, P. M.; Wang, S.; Endicott, J. A.; Noble, M. E. Comparative Structural and Functional Studies of 4-(Thiazol-5-Yl)-2-(Phenylamino)Pyrimidine-5-Carbonitrile CDK9 Inhibitors Suggest the Basis for Isotype Selectivity. J. Med. Chem. 2013, 56, 660– 670, DOI: 10.1021/jm301495v40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVKhurjE&md5=8c09b13b5f828b7a013607cab64b0b80Comparative Structural and Functional Studies of 4-(Thiazol-5-yl)-2-(phenylamino)pyrimidine-5-carbonitrile CDK9 Inhibitors Suggest the Basis for Isotype SelectivityHole, Alison J.; Baumli, Sonja; Shao, Hao; Shi, Shenhua; Huang, Shiliang; Pepper, Chris; Fischer, Peter M.; Wang, Shudong; Endicott, Jane A.; Noble, Martin E.Journal of Medicinal Chemistry (2013), 56 (3), 660-670CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Cyclin-dependent kinase 9/cyclin T, the protein kinase heterodimer that constitutes pos. transcription elongation factor b, is a well-validated target for treatment of several diseases, including cancer and cardiac hypertrophy. In order to aid inhibitor design and rationalize the basis for CDK9 selectivity, we have studied the CDK-binding properties of six different members of a 4-(thiazol-5-yl)-2-(phenylamino)pyrimidine-5-carbonitrile series that bind to both CDK9/cyclin T and CDK2/cyclin A. We find that for a given CDK, the melting temp. of a CDK/cyclin/inhibitor complex correlates well with inhibitor potency, suggesting that differential scanning fluorometry (DSF) is a useful orthogonal measure of inhibitory activity for this series. We have used DSF to demonstrate that the binding of these compds. is independent of the presence or absence of the C-terminal tail region of CDK9, unlike the binding of the CDK9-selective inhibitor 5,6-dichlorobenzimidazone-1-β-D-ribofuranoside (DRB). Finally, on the basis of 11 cocrystal structures bound to CDK9/cyclin T or CDK2/cyclin A, we conclude that selective inhibition of CDK9/cyclin T by members of the 4-(thiazol-5-yl)-2-(phenylamino)pyrimidine-5-carbonitrile series results from the relative malleability of the CDK9 active site rather than from the formation of specific polar contacts.
- 41Galkin, A. V.; Melnick, J. S.; Kim, S.; Hood, T. L.; Li, N.; Li, L.; Xia, G.; Steensma, R.; Chopiuk, G.; Jiang, J.; Wan, Y.; Ding, P.; Liu, Y.; Sun, F.; Schultz, P. G.; Gray, N. S.; Warmuth, M. Identification of NVP-TAE684, a Potent, Selective, and Efficacious Inhibitor of NPM-ALK. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 270– 275, DOI: 10.1073/pnas.060941210341https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjt12jsQ%253D%253D&md5=4a3441ee637a67dc496c80730bd0e184Identification of NVP-TAE684, a potent, selective, and efficacious inhibitor of NPM-ALKGalkin, Anna V.; Melnick, Jonathon S.; Kim, Sunjoon; Hood, Tami L.; Li, Nanxin; Li, Lintong; Xia, Gang; Steensma, Ruo; Chopiuk, Greg; Wan, Yongqin; Ding, Peter; Liu, Yi; Sun, Fangxian; Schultz, Peter G.; Gray, Nathanael S.; Warmuth, MarkusProceedings of the National Academy of Sciences of the United States of America (2007), 104 (1), 270-275CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Constitutive overexpression and activation of NPM-ALK fusion protein [t(2:5)(p23;q35)] is a key oncogenic event that drives the survival and proliferation of anaplastic large cell lymphomas (ALCLs). We have identified a highly potent and selective small-mol. ALK inhibitor, NVP-TAE684, which blocked the growth of ALCL-derived and ALK-dependent cell lines with IC50 values between 2 and 10 nM. NVP-TAE684 treatment resulted in a rapid and sustained inhibition of phosphorylation of NPM-ALK and its downstream effectors and subsequent induction of apoptosis and cell cycle arrest. In vivo, NVP-TAE684 suppressed lymphomagenesis in two independent models of ALK-pos. ALCL and induced regression of established Karpas-299 lymphomas. NVP-TAE684 also induced down-regulation of CD30 expression, suggesting that CD30 may be used as a biomarker of therapeutic NPM-ALK kinase activity inhibition.
- 42Kothe, M.; Kohls, D.; Low, S.; Coli, R.; Rennie, G. R.; Feru, F.; Kuhn, C.; Ding, Y.-H. Selectivity-Determining Residues in Plk1. Chem. Biol. Drug Des. 2007, 70, 540– 546, DOI: 10.1111/j.1747-0285.2007.00594.x42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhsVGhsbrJ&md5=0f976bb98fe7d0413e8d2314abc68e2bSelectivity-determining residues in Plk1Kothe, Michael; Kohls, Darcy; Low, Simon; Coli, Rocco; Rennie, Glen R.; Feru, Frederic; Kuhn, Cyrille; Ding, Yuan-HuaChemical Biology & Drug Design (2007), 70 (6), 540-546CODEN: CBDDAL; ISSN:1747-0277. (Blackwell Publishing Ltd.)Polo-like kinase 1 is an important regulator of cell cycle progression whose over-expression is often assocd. with oncogenesis. Polo-like kinase 1 hence represents an attractive target for cancer intervention. BI 2536 (Boehringer Ingelheim, Ingelheim, Germany), a Polo-like kinase 1 inhibitor currently in clin. trials, exhibits nanomolar potency against Polo-like kinase isoforms and high selectivity against other kinases. The authors have previously published the crystal structures of the Polo-like kinase 1 domain in complex with AMPPNP and an Aurora A inhibitor. In this work, the authors present the cocrystal structure of Polo-like kinase 1 with BI 2536. The structure, in combination with selectivity data for BI 2536 and related compds., illustrates important features for potency and selectivity. In particular, the authors show that the methoxy group of BI 2536 is an important specificity determinant against non-Polo-like kinases by taking advantage of a small pocket generated by Leu 132 in the hinge region of Polo-like kinase 1. The work presented here provides a framework for structure-based drug design of Polo-like kinase 1-specific inhibitors.
- 43Blake, J. F.; Burkard, M.; Chan, J.; Chen, H.; Chou, K.-J.; Diaz, D.; Dudley, D. A.; Gaudino, J. J.; Gould, S. E.; Grina, J.; Hunsaker, T.; Liu, L.; Martinson, M.; Moreno, D.; Mueller, L.; Orr, C.; Pacheco, P.; Qin, A.; Rasor, K.; Ren, L.; Robarge, K.; Shahidi-Latham, S.; Stults, J.; Sullivan, F.; Wang, W.; Yin, J.; Zhou, A.; Belvin, M.; Merchant, M.; Moffat, J.; Schwarz, J. B. Discovery of (S)-1-(1-(4-Chloro-3-Fluorophenyl)-2-Hydroxyethyl)-4-(2-((1-Methyl-1H-Pyrazol-5-Yl)Amino)Pyrimidin-4-Yl)Pyridin-2(1H)-One (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development. J. Med. Chem. 2016, 59, 5650– 5660, DOI: 10.1021/acs.jmedchem.6b0038943https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XoslOrsLs%253D&md5=6d2d52dabea4d443ddee2959df6d45f1Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical DevelopmentBlake, James F.; Burkard, Michael; Chan, Jocelyn; Chen, Huifen; Chou, Kang-Jye; Diaz, Dolores; Dudley, Danette A.; Gaudino, John J.; Gould, Stephen E.; Grina, Jonas; Hunsaker, Thomas; Liu, Lichuan; Martinson, Matthew; Moreno, David; Mueller, Lars; Orr, Christine; Pacheco, Patricia; Qin, Ann; Rasor, Kevin; Ren, Li; Robarge, Kirk; Shahidi-Latham, Sheerin; Stults, Jeffrey; Sullivan, Francis; Wang, Weiru; Yin, Jianping; Zhou, Aihe; Belvin, Marcia; Merchant, Mark; Moffat, John; Schwarz, Jacob B.Journal of Medicinal Chemistry (2016), 59 (12), 5650-5660CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The extracellular signal-regulated kinases ERK1/2 represent an essential node within the RAS/RAF/MEK/ERK signaling cascade that is commonly activated by oncogenic mutations in BRAF or RAS or by upstream oncogenic signaling. While targeting upstream nodes with RAF and MEK inhibitors has proven effective clin., resistance frequently develops through reactivation of the pathway. Simultaneous targeting of multiple nodes in the pathway, such as MEK and ERK, offers the prospect of enhanced efficacy as well as reduced potential for acquired resistance. Described herein is the discovery and characterization of GDC-0994 (22), an orally bioavailable small mol. inhibitor selective for ERK kinase activity.
- 44Johnson, T. W.; Richardson, P. F.; Bailey, S.; Brooun, A.; Burke, B. J.; Collins, M. R.; Cui, J. J.; Deal, J. G.; Deng, Y.; Dinh, D.; Engstrom, L. D.; He, M.; Hoffman, J.; Hoffman, R. L.; Huang, Q.; Kania, R. S.; Kath, J. C.; Lam, H.; Lam, J. L.; Le, P. T.; Lingardo, L.; Liu, W.; McTigue, M.; Palmer, C. L.; Sach, N. W.; Smeal, T.; Smith, G. L.; Stewart, A. E.; Timofeevski, S.; Zhu, H.; Zhu, J.; Zou, H. Y.; Edwards, M. P. Discovery of (10R)-7-Amino-12-Fluoro-2,10,16-Trimethyl-15-Oxo-10,15,16,17-Tetrahydro- 2H −8,4-(Metheno)Pyrazolo[4,3-h][2,5,11]-Benzoxadiazacyclotetradecine-3-Carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-Ros. J. Med. Chem. 2014, 57, 4720– 4744, DOI: 10.1021/jm500261q44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXnvVSgtL4%253D&md5=db21844f8e9a2f9ca5953bca56ad7b1cDiscovery of (10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-ros Oncogene 1 (ROS1) with Preclinical Brain Exposure and Broad-Spectrum Potency against ALK-Resistant MutationsJohnson, Ted W.; Richardson, Paul F.; Bailey, Simon; Brooun, Alexei; Burke, Benjamin J.; Collins, Michael R.; Cui, J. Jean; Deal, Judith G.; Deng, Ya-Li; Dinh, Dac; Engstrom, Lars D.; He, Mingying; Hoffman, Jacqui; Hoffman, Robert L.; Huang, Qinhua; Kania, Robert S.; Kath, John C.; Lam, Hieu; Lam, Justine L.; Le, Phuong T.; Lingardo, Laura; Liu, Wei; McTigue, Michele; Palmer, Cynthia L.; Sach, Neal W.; Smeal, Tod; Smith, Graham L.; Stewart, Albert E.; Timofeevski, Sergei; Zhu, Huichun; Zhu, Jinjiang; Zou, Helen Y.; Edwards, Martin P.Journal of Medicinal Chemistry (2014), 57 (11), 4720-4744CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Although crizotinib demonstrates robust efficacy in anaplastic lymphoma kinase (ALK)-pos. non-small-cell lung carcinoma patients, progression during treatment eventually develops. Resistant patient samples revealed a variety of point mutations in the kinase domain of ALK, including the L1196M gatekeeper mutation. In addn., some patients progress due to cancer metastasis in the brain. Using structure-based drug design, lipophilic efficiency, and phys.-property-based optimization, highly potent macrocyclic ALK inhibitors were prepd. with good absorption, distribution, metab., and excretion (ADME), low propensity for p-glycoprotein 1-mediated efflux, and good passive permeability. These structurally unusual macrocyclic inhibitors were potent against wild-type ALK and clin. reported ALK kinase domain mutations. Significant synthetic challenges were overcome, utilizing novel transformations to enable the use of these macrocycles in drug discovery paradigms. This work led to the discovery of 8k (PF-06463922), combining broad-spectrum potency, central nervous system ADME, and a high degree of kinase selectivity.
- 45Tan, L.; Akahane, K.; McNally, R.; Reyskens, K. M. S. E.; Ficarro, S. B.; Liu, S.; Herter-Sprie, G. S.; Koyama, S.; Pattison, M. J.; Labella, K.; Johannessen, L.; Akbay, E. A.; Wong, K.-K.; Frank, D. A.; Marto, J. A.; Look, T. A.; Arthur, J. S. C.; Eck, M. J.; Gray, N. S. Development of Selective Covalent Janus Kinase 3 Inhibitors. J. Med. Chem. 2015, 58, 6589– 6606, DOI: 10.1021/acs.jmedchem.5b0071045https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht12ksLjM&md5=af334f0cdd1d4f58462182f138619099Development of Selective Covalent Janus Kinase 3 InhibitorsTan, Li; Akahane, Koshi; McNally, Randall; Reyskens, Kathleen M. S. E.; Ficarro, Scott B.; Liu, Suhu; Herter-Sprie, Grit S.; Koyama, Shohei; Pattison, Michael J.; Labella, Katherine; Johannessen, Liv; Akbay, Esra A.; Wong, Kwok-Kin; Frank, David A.; Marto, Jarrod A.; Look, Thomas A.; Arthur, J. Simon C.; Eck, Michael J.; Gray, Nathanael S.Journal of Medicinal Chemistry (2015), 58 (16), 6589-6606CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The Janus kinases (JAKs) and their downstream effectors, signal transducer and activator of transcription proteins (STATs), form a crit. immune cell signaling circuit, which is of fundamental importance in innate immunity, inflammation, and hematopoiesis, and dysregulation is frequently obsd. in immune disease and cancer. The high degree of structural conservation of the JAK ATP binding pockets has posed a considerable challenge to medicinal chemists seeking to develop highly selective inhibitors as pharmacol. probes and as clin. drugs. Here we report the discovery and optimization of 2,4-substituted pyrimidines as covalent JAK3 inhibitors that exploit a unique cysteine (Cys909) residue in JAK3. Investigation of structure-activity relationship (SAR) utilizing biochem. and transformed Ba/F3 cellular assays resulted in identification of potent and selective inhibitors such as compds. 9 and 45. A 2.9 Å cocrystal structure of JAK3 in complex with 9 confirms the covalent interaction. Compd. 9 exhibited decent pharmacokinetic properties and is suitable for use in vivo. These inhibitors provide a set of useful tools to pharmacol. interrogate JAK3-dependent biol.
- 46Smilkstein, M.; Sriwilaijaroen, N.; Kelly, J. X.; Wilairat, P.; Riscoe, M. Simple and Inexpensive Fluorescence-Based Technique for High-Throughput Antimalarial Drug Screening. Antimicrob. Agents Chemother. 2004, 48, 1803– 1806, DOI: 10.1128/AAC.48.5.1803-1806.200446https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjslGltLw%253D&md5=84a9721da1d873dfca16d1f2570ed304Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screeningSmilkstein, Martin; Sriwilaijaroen, Nongluk; Kelly, Jane Xu; Wilairat, Prapon; Riscoe, MichaelAntimicrobial Agents and Chemotherapy (2004), 48 (5), 1803-1806CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Radioisotopic assays involve expense, multistep protocols, equipment, and radioactivity safety requirements which are problematic in high-throughput drug testing. This study reports an alternative, simple, robust, inexpensive, one-step fluorescence assay for use in antimalarial drug screening. Parasite growth is detd. by using SYBR Green 1, a dye with marked fluorescence enhancement upon contact with Plasmodium DNA. A side-by-side comparison of this fluorescence assay and a std. radioisotopic method was performed by testing known antimalarial agents against Plasmodium falciparum strain D6. Both assay methods were used to det. the effective concn. of drug that resulted in a 50% redn. in the obsd. counts (EC50) after 48 h of parasite growth in the presence of each drug. The EC50s of chloroquine, quinine, mefloquine, artemisinin, and 3,6-bis-.vepsiln.-(N,N-diethylamino)-amyloxyanthone were similar or identical by both techniques. The results obtained with this new fluorescence assay suggest that it may be an ideal method for high-throughput antimalarial drug screening.
- 47Rakotoarivelo, N. V.; Perio, P.; Najahi, E.; Nepveu, F. Interaction between Antimalarial 2-Aryl-3H-Indol-3-One Derivatives and Human Serum Albumin. J. Phys. Chem. B 2014, 118, 13477– 13485, DOI: 10.1021/jp507569e47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVClsbzF&md5=e36b416d37d4fe81317daa90bd20a3d2Interaction between Antimalarial 2-Aryl-3H-indol-3-one Derivatives and Human Serum AlbuminRakotoarivelo, Nambinina V.; Perio, Pierre; Najahi, Ennaji; Nepveu, FrancoiseJournal of Physical Chemistry B (2014), 118 (47), 13477-13485CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)Binding of drugs to plasma proteins, such as albumin, is a major factor which dets. their pharmacokinetics and pharmacol. effects. Therefore, the interactions between human serum albumin (HSA) and four antimalarial compds. selected in the 2-aryl-3H-indol-3-one series have been investigated using UV-visible, fluorescence and CD spectroscopies. Compds. produced a static quenching of the intrinsic fluorescence of HSA. The thermodn. parameters have shown that the binding reaction is endothermic for three compds. while exothermic for the 2-phenyl-3H-indol-3-one, 3. The interaction is entropically driven with predominant hydrophobic forces with binding affinities of the order of 104 M-1. The highest binding const. is obsd. for 3 (Kλ=280nm = 4.53 × 104 M-1) which is also the less active compd. against Plasmodium falciparum. Synchronous fluorescence gave qual. information on the conformational changes of HSA while quant. data were obtained with CD. Displacement expts. with site markers indicated that drugs bind to HSA at site I (subdomain IIA). In addn., the apparent binding const. and the binding site no. were calcd. in the presence of different ions.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.3c00354.
Percent identity and similarity for PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB; Use of additional molecular docking constraints; comparison of 1 docked to the structures of PfARK1, PfNEK3, PfPK9, and PfPKB; summary of 1H-13C HMBC results, methods; experimental spectra for CZC-54252 analogues; supplemental references (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.