Abstract
Now entering its fourth decade, research on the biological function, small molecule inhibition, and disease relevance of the three known isoforms of protein kinase D, PKD1, PKD2, and PKD3, has entered a mature development stage. This miniperspective focuses on the medicinal chemistry that provided a structurally diverse set of mainly active site inhibitors, which, for a brief time period, moved through preclinical development stages but have yet to be tested in clinical trials. In particular, between 2006 and 2012, a rapid expansion of synthetic efforts led to several moderately to highly PKD-selective chemotypes but did not yet achieve PKD subtype selectivity or resolve general toxicity and pharmacokinetic challenges. In addition to cancer, other unresolved medical needs in cardiovascular, inflammatory, and metabolic diseases would, however, benefit from a renewed focus on potent and selective PKD modulators.
Graphical Abstract
INTRODUCTION
Since the first report of Protein Kinase D1 (PKD1, also known as PKCμ) in 1994,1 two additional isoforms (PKD2 and PKD3) have been characterized, and extensive progress has been made on the disease-related biological processes that engage this family of diacylglycerol (DAG)-regulated serine-threonine protein kinases. PKDs belong to the Ca2+/calmodulin-dependent kinase (CaMK) superfamily based on homology in the catalytic domain. In addition to cancer and cardiovascular diseases, the methionine gatekeeper kinases PKD1–3 are implicated in inflammatory, metabolic, CNS, and angiogenesis-related disorders as well as immune dysregulation and even viral infections. Several drug discovery programs were launched with the goal to generate selective PKD inhibitors for clinical trials. PKD biology, function, and signaling mechanisms have been reviewed in considerable depth,2 and this perspective will therefore mainly focus on the structural classifications, syntheses, and potential for further development of small molecule PKD inhibitors, augmenting other recent medicinal chemistry reviews.3
Chart 1 provides an overview of the most important classes of PKD inhibitors that have exhibited nanomolar potencies in in vitro enzyme assays. Chemotypes I–X are introduced to classify the scaffolds, and specific lead examples 1–10 allow for a structure-guided comparison between chemotype classes that in many cases are representative for general kinase active site inhibitor designs. Chart 2 shows the timeline of significant discoveries in this now firmly established field, highlighting the most active development period from 2006 to 2012.
Chart 1. Overview of Major Chemotypes and Specific Exemplified Structures for Potent PKD1–3 Inhibitors and Representative In Vitro IC50 or KD Data for PKD1 Inhibitiona.
aThe chemotype scaffolds are highlighted in blue in the specific examples. (*) KD measurement.
Chart 2.
Timeline of the PKD Inhibitor Discovery and Development Wave, Peaking between 2006 and 2012
Shortly after the discovery of PKD1, Gschwendt et al. screened a series of indolocarbazoles including staurosporine (1), K252a, Gö6976, and Gö6983 (Figure 1).4 Remarkably, these four bisindoles spanned a 3000-fold potency range, with the IC50 of the unselective pan-kinase inhibitor K252a being in the single-digit nanomolar and the maleimide Gö6983 in the double-digit micromolar range. The latter compound proved to be useful as a negative control in kinase assays. Furthermore, in contrast to the lack of kinase isoform selectivity observed for K252a and 1, the structurally closely related otherwise classical DAG- and calcium-dependent PKCα,β,γ-selective Gö6976 inhibited the calcium-independent, DAG-regulated PKD1 also very effectively at 20 nM. These and other SAR comparisons suggested that the central benzene ring in 1, K252a, and Gö6976 was necessary for PKD1 inhibition but that indole nitrogen substituents could be readily exchanged for achieving kinase selectivity for PKD. Accordingly, the discovery of the PKD-inhibitory chemotype I validated this kinase as a suitable target for medicinal chemistry studies.
Figure 1.
Structures and PKD1 potencies for selected chemotype I inhibitors; data suggest that the fused aromatic system (shown in red) is necessary for high potency but that indole nitrogen substituents (shown in blue) can convey increased selectivity for PKD over other kinases.
Synthesis and Representative SAR Analyses of Chemotypes I–X.
A concise synthesis of (+)-K252a, a representative example for inhibitors of chemotype I, is shown in Scheme 1, but many other strategies have culminated in the successful preparation of these natural product scaffolds and their analogs.16 In a versatile regioselective approach in 1999, Kobayashi et al. reported the conversion of indole 11 to the target indolocarbazole alkaloid.17 The allyl ester derived from 11 was first brominated with NBS and then N-glycosylated to generate the β-furanosylated intermediate 12. The allyl ester protective group was removed with Pd(0) catalysis, and the acid was coupled to tryptamine with the water-soluble reagent 1-ethyl-3-(3-(dimethylamino)propyl)carbodiimide (EDCI) in high overall yield from 11. Amide 13 was then oxidized regioselectively with DDQ to introduce a ketone group at the more reactive indole side chain, followed by acetylation to generate imide 14. An intramolecular aldol reaction/dehydration utilized DBU in the presence of 4 Å molecular sieves, and the resulting lactam 15 was subjected to a photochemical 6π-electrocyclization to generate the benzene ring after elimination of HBr from the intermediate cyclohexadiene. Potassium hydroxide cleavage of the tolyl esters and selective conversion of the primary alcohol to the iodide led to key intermediate 16. Cyclization of this compound to introduce the seven-membered ring was problematic but ultimately accomplished by a multistep sequence starting with a conversion of the iodide to the selenide, acetylation of the secondary alcohol, oxidation to the selenoxide, and elimination with triethylamine to give alkene 17. Dihydropyran (DHP) was used to prevent an undesired addition of phenylselenous acid to the reactive enol ether. Treatment of 17 with iodine, potassium iodide, and DBU effected a smooth iodoglycosylation to close the desired seven-membered ring, and a radical-mediated deiodination, methanolysis of the acetate, and Swern oxidation provided ketone 18 in excellent overall yield.
Scheme 1.
Regioselective Synthesis of (+)-K252a17
The α-hydroxy ester in (+)-K252a was introduced via formation of the cyanohydrin of 18, O-acetylation, and conversion of the nitrile to the amide 19 with gaseous HCl in formic acid. Interestingly, a single, kinetically favored cyanohydrin acetate was formed when the cyanohydrin formation was performed with a solution of hydrogen cyanide in pyridine. Finally, 19 was hydrolyzed under basic conditions, and the acid was converted to the methyl ester (+)-K252a by treatment with diazomethane. This linear but high-yielding synthetic route is also potentially useful for the preparation of a variety of interesting analogs of (+)-K252a.
For the preparation of simplified (+)-K252a analogs related to the potent checkpoint kinase inhibitor Gö6976, Roy et al. used the N-alkylated glyoxylyl chloride 20 and indole-3-acetic acid 21 to form the anhydride 22 in 31% yield (Scheme 2).18 Treatment of 22 with hexamethyldisilazane (HDMS) and methanol gave the corresponding imide in 96% yield, and oxidation with palladium(II) trifluoroacetate led to analog 23 in 30% yield.
Scheme 2.
Preparation of Gö6976 Analogs18
Subsequent to the discovery of PKD and the characterization of staurosporine-type inhibitors of this PKC variant, a trypanocidal drug, suramin (Figure 2), was shown to differentially affect the activity of several PKC isoforms19 and to activate PKD.20 However, the pharmacological properties of this hexasulfonated rather promiscuous compound are complex,21 and its properties or those of its analogs as modulators of PKD have not been further evaluated.
Figure 2.
Structure of the PKC activity modulator suramin.
In 2008, the first potent and selective cell-active small molecule inhibitor of PKD, benzoxoloazepinolone CID755673, was reported as part of a high-throughput assay of 196 173 compounds from the National Institutes of Health small molecule repository.22 In part due to its ready availability, CID755673 has become a probe for PKD and a potential therapeutic molecule for inflammation- and metabolic dysregulation-driven pathologies.23,24
CID755673 introduced chemotype II as a novel class of selective PKD inhibitors and inspired the discovery of numerous potent analogs, including 9-hydroxy-3,4-dihydrobenzo[4,5]-thieno[2,3-f][1,4]thiazepin-5(2H)-one (2, kb-NB142-70) (Scheme 3).6 Several approaches to construct the fused tricyclic core system of these analogs have been reported. Benzylation of commercially available 3-hydroxycinnamic acid 24 and a Higa cyclization25 with thionyl chloride provided the benzo-[b]-thiophene acid chloride intermediate, which was subsequently converted to methyl ester 25 with triethylamine and methanol. Condensation of 25 with cysteamine hydrochloride (26) in the presence of DBU resulted in the formation of the desired benzothienothiazepinone 27, and debenzylation with boron tribromide led to kb-NB142-70 in good yield. Interestingly, the replacement of the furan and methylene group in CID755673 with a thiophene and a sulfur atom, respectively, in 2 increased the potency more than 6-fold: CID755673 showed an IC50 of 182 nM against PKD1, while 2 had an IC50 of 28 nM.
Scheme 3.
Structure of HTS Hit CID755673, and Preparation of the Potent and Selective PKD Inhibitor kb-NB142-706
To improve the plasma stability of 2, the pyrimidine kmg-NB4–23 was also prepared (Scheme 4).6 In this analog, an electron-deficient pyrimidine methyl ether was used in place of the phenol in 2 to reduce the rate of phase I and II metabolism of the latter moiety. Starting with commercially available 28, annulation of the pyrimidine was accomplished with potassium cyanate in acetic acid and base treatment to generate diol 29. The two hydroxy groups were converted to chlorides with POCl3, and a palladium-catalyzed reductive dechlorination occurred exclusively at the C-4 position to yield 30. The methanolysis of the C-2 chloride generated a methyl ether intermediate that was brominated to obtain the desired 31. Selective deprotonation at C-6 with Knochel’s magnesium tetramethylpiperidite base and treatment with Mander’s reagent led to methyl ester 32, which was cyclized with 26 and completed the formation of kmg-NB4-23. In spite of the significant structural modifications compared to kb-NB142–70, this compound retained an IC50 of 124 nM in the PKD1 assay.
Scheme 4.
Preparation of the Pyrimidine Analog kmg-NB4-236
The SAR of these benzofuroazepine, benzothienothiazepine, and pyrimidothienothiazepine PKD inhibitors covers several orders of magnitude. Chart 3 illustrates the strong dependence of the PKD1 inhibitory activity on the nature of the heterocycle, the ring size of the fused lactam, and the phenolic substituent. Exchanging the furan with a pyrrole slightly improves the activity from 0.18 μM in CID755673 to 0.13 μM in kb-NB123-57. The thiophene analog kb-NB142-70 that also contains a thioether in the lactam tether is significantly more potent with an IC50 of 28 nM. In contrast, enlarging the furan to a pyran core decreases the activity ca. 10-fold (CID755673 vs CID797718) and ca. 5-fold when the seven-membered lactam is enlarged to an eight-membered analog (kb-NB96-53). The methyl ether analog of CID755673, kb-NB77-56, also drops ca. 10-fold in activity, but the inhibitory potency is partly regained in the benzothiophene kb-NB184-02 and the pyrimidothiophene kmg-NB4-23.5 The latter modification is significant because it prevents O-glucoronidation and rapid elimination of the active component in in vivo experiments.7
Chart 3.
Chemical Structures and PKD1 Inhibitory Activities (IC50’s) of Representative Chemotype II Analogs
Several other lead compounds that directly inhibit PKD or influence its activity were also reported in the patent literature between 2007 and 2009, and some of these leads were further developed or inspired the design of related structures (see, for example, chemotypes III and IV) (Figure 3).26-29
Figure 3.
Representative examples of PKC/PKD inhibitors claimed in the early patent literature.
Also, during this time period of vivid interest in the therapeutic potential of PKD in cardiovascular diseases, Novartis and Myogen (Gilead) reported several heterocyclic scaffolds with strong PKD-inhibitory effects and claims covering a broad range of indications,30-32 including cancer and heart failure33 and, more recently, neurodegenerative disorders such as Friedreich’s Ataxia (FRDA).34 For example, the bipyridyl PKD inhibitor 3 (BPKDi, 2′-(cyclohexylamino)-6-(1-piperazinyl)[2,4′-bipyridine]-4-carboxamide) was based on a hit from a time-resolved fluorescence resonance transfer (TR-FRET) library screen of >650 000 compounds from full-length, human PKD1 overexpressed in insect cells (Scheme 5).8 An earlier 2,6-naphthyridine scaffold analog proved potent and orally bioavailable but showed moderate kinase selectivity and comparative nanomolar off-target activity vs GSK3β.35 In contrast, 3 inhibited PKD1, PKD2, and PKD3 with IC50 values of 1, 9, and 1 nM, respectively, but did not inhibit other related kinases.8 Typical for SAR analyses of this chemotype, citrazinic acid (33) was converted to dibromide 34 followed by selective aminolysis and Suzuki coupling to generate the bipyridine backbone 38 (Scheme 5). Ester aminolysis, an SNAr reaction with cyclohexylamine, and deprotection of the Boc group completed the synthesis of the lead compound, BPKDi (3).
Scheme 5.
Preparation of the Bipyridine BPKDi (3)8
Cancer Research Technology’s compound CRT0066101 (47, CRT101, (S)-2-(4-((2-aminobutyl)amino)pyrimidin-2-yl)-4-(1-methyl-1H-pyrazol-4-yl)phenol) represents another example of a chemotype III lead structure.26,36 A large number of analogs of this lead structure were prepared using variations of the synthetic pathway shown in Scheme 6. For the modular assembly of the parent compound, aldehyde 39 was converted to nitrile 40 followed by aminolysis with lithium hexamethyldisilazide (LiHMDS) to give amidine intermediate 41. Condensation with ethyl propiolate and saponification provided hydroxypyrimidine 42, which was converted to the chloride, demethylated, and combined with Boc-protected 44 to attach the diamine side chain. Suzuki coupling of 45 with boronic acid 46 and Boc cleavage then led to 47.
Scheme 6.
Preparation of the 2-Arylpyrimidine 4726
Meta-substituted aromatics are a popular pharmacophore in medicinal chemistry, and several of the PKD-inhibitory scaffolds take advantage of this motif. The orally bioavailable 3,5-diarylisoxazole 4 represents chemotype IV and was shown to inhibit PKD selectively with a low nanomolar EC50, demonstrating ca. 9- and 3-fold selectivity for PKD1 versus PKD2 and PKD3.9 For the preparation of 4 and related 3,5-diarylazoles, a nitrile oxide cycloaddition of chloroimidate 48 and alkyne 49 was used to access the isoxazole moiety in 50 (Scheme 7). Radical bromination of the methyl group and substitution of the benzylic bromide with amine 51 provided intermediate 52, which was saponified and coupled with alanine amide 53. Treatment of amide 54 with trifluoroacetic anhydride (TFAA) led to a nitrile containing a trifluoroacetyl group at the benzylic amine. The trifluoroacetamide was then removed chemoselectively by treatment with sodium borohydride to yield inhibitor 4.
Scheme 7.
Preparation of the 3,5-Diarylisoxazole 49
The SAR of 4 reveals a strong dependence of the inhibitory activity on the end groups of the meta-linked arene triad (Chart 4). The enantiomeric aminonitrile in 4′ increases the IC50 ca. 15-fold, establishing a strong stereoselectivity in the PKD1 binding mode. The polar nitrile function is also required for low nanomolar inhibition since the isopropylamide 4a is ca. 35 times less active than 4. In contrast, exchanging the 4-aminopyran in 4 with the isopropylamine in 4b has only a minor effect on the IC50. Isoxazole 4b and pyrazole 4c also have single-digit nanomolar activity, but similar to the isoxazole series, removing the nitrile from the terminal amide in isopropylamide 4d diminishes inhibition ca. 25-fold. Pyrazoles 4e and 4f have even lower activities, with IC50’s of 1.0 and 2.5 μM, respectively.9 The structurally related furan CID2011756 has similarly moderate activity for PKD1 but ca. 5-fold increased activity for the other PKD isoforms with IC50 values of 3.2, 0.6, and 0.7 μM, respectively, for PKD1, PKD2, and PKD3.37 CID2011756 might therefore be an interesting lead structure for the design of PKD2/3-selective inhibitors.
Chart 4.
Chemical Structures and PKD1 Inhibitory Activities (IC50’s) of Representative Chemotype IV Analogs
The pyridine benzamide CRT0066051 (61, CRT51, CRT5, 3-(6-amino-5-(6-ethoxynaphthalen-2-yl)pyridin-3-yl)-N-(2-(dimethylamino)ethyl)benzamide) is another example of a structural variation of chemotype IV (Scheme 8).38 Coupling of acid 55 with diamine 56 in the presence of EDCI provided amide 57 in 63% yield. The meta-substituted pharmacophore was then readily assembled by two Suzuki couplings first with bromide 58 and then boronate 60 to give the target inhibitor 61. Both CRT0066051 (61) and CRT0066101 (47) have low-nanomolar pan-PKD inhibitory activities and have been used to characterize the role of PKD in various biological processes and disease models. A potential concern for in vivo applications of CRT0066051 is the low aqueous solubility at pH 7 for this compound, calculated at log S = −5.9, versus log S = −3.4 for CRT0066101,39 which might also limit dosing regimens and bioavailability.
Scheme 8.
Preparation of the Pyridine Benzamide 6127
In 2017, CRT0066101 and CRT0066051 were reported to inhibit the replication of HRV, PV, and FMDV, suggesting that PKD may also present opportunities in antiviral drug discovery.40 Furthermore, in 2019, an industrial collaboration disclosed the structure-based design of potent, conformationally restricted imidazole inhibitors that broadly fit into this chemotype (Chart 5).41 While Im-1 inhibited PKD1 in an ADP FP assay with high stereoselectivity over its enantiomer Im-1′, it was also a potent inhibitor of NF-κB-inducing kinase (NIK). Similarly, oxazole Im-2 in spite of its increased PKD1 activity was still very potent in the NIK assay. High selectivity for PKD1 over NIK was observed with the cyclopropyl-substituted Im-3. While the ether-bridged Im-4 proved to be selective for NIK over PKD1, the desired preference for PKD1 was regained by introducing the cyclopropyl group in Im-5. In fact, among 220 kinases tested, Im-5 only inhibited PKD1 >30% at 0.1 μM concentration. Interestingly, Im-5 has the (S)-configuration, which was the less-preferred stereochemistry for the PKD1 inhibitory potency of Im-1 but provided a 10-fold increased selectivity over NIK. Unfortunately, PKD2 and PKD3 inhibitory data are not available for these compounds.
Chart 5. Chemical Structures and PKD1 Inhibitory Activities (Ki’s) of Conformationally Rigidified Chemotype IV Analogsa.
aThe ratio of NIK/PKD1 Ki’s is shown in parentheses. The higher the ratio, the greater the selectivity for PKD1.41
A screen of a diverse collection of kinase-biased inhibitors in 2012 revealed additional scaffolds for PKD inhibition (Figure 4).10 In particular, the 4-azaindole chemotype V exemplified by compound 139 (5) distinguished itself as a solid ATP-competitive lead structure, responsive to structural modifications, with a low double-digit nanomolar potency and robust cell activity. Furthermore, this chemotype offers the hitherto untapped opportunity for dual PKD-p38 MAP kinase inhibitors.42 In addition to 5, 4-azaindoles 68 and 69 demonstrated a moderate to high level of inhibition of PKD1 with the indole NH being a prerequisite for the highest degree of inhibition. Lead compound 5 also lacked activity for undesired kinase targets (PKCα, PKCδ, and CAMKIIα).10
Figure 4.
Chemical structures of novel PKD1 small molecule inhibitors found as primary hits in a radiometric PKD1 assay. Hits were selected based on their ability to inhibit PKD1 at or above 50% at 1 μM (percent inhibition in parentheses).10
The preparation of 4-azaindole inhibitor 5 used dimethylacetal 70 for a chain extension with 4-fluorobenzyl bromide to generate N-oxide 71 after oxidation with methyltrioxorhenium (Scheme 9). Condensation with aminopyridine 72 and Pd-catalyzed Heck cyclization of the enamine 73 provided the azaindole 74, which was chlorinated and treated with amine 76 to complete the synthesis of 5.42
Scheme 9.
Preparation of 4-Azaindole 542
In 2013, the generally well-known kinase-inhibitory pyrazolopyrimidine chemotype VI was also found to act as a strong inhibitor of PKD1–3.11 In particular 1-naphthyl-PP1 (6, 1-NA-PP1, 1-(tert-butyl)-3-(naphthalen-1-yl)-1H-pyrazolo[3,4-d]-pyrimidin-4-amine) was found to inhibit PKD isoforms at about 100 nM as an ATP-competitive inhibitor. However, among 17 synthetic analogs of 6, only 83 provided a minor modulation of PKD1 activity, confirming that the sterically demanding 6 already provides a tight fit in the binding pocket. This property could potentially be used in the future for the development of analogs of 6 and 83 as PKD subtype-specific inhibitors. The synthesis of 83 and related compounds was accomplished starting with a Vilsmeier–Haack reaction of barbituric acid 77 to give the trichloropyrimidine 78 in 57% yield (Scheme 10). The pyrazole ring was closed by treatment of 78 with methyl hydrazine in the presence of triethylamine to yield 59% of the pyrazolo[3,4-d]pyrimidine 79. Selective aminolysis at C-6 with ammonia at room temperature led in 81% yield to 80, which was brominated to give the pyrazolopyrimidine 81 in 40% yield. A selective Suzuki coupling at C-3 with 1-naphthyl boronic acid provided intermediate 82, and the chlorine group was reduced by a catalytic hydrogen transfer process to give 83 in 68% yield over two steps.
Scheme 10.
Preparation of the Pyrazolopyrimidine 8311
More recently, an analog of 6, 3-indolylmethyl-pyrazolo[3,4-d]pyrimidine (3-IN-PP1), was found to be significantly more potent with an IC50 of 33 nM against PKD2.43
Shortly after the discovery of the PKD-inhibitory effects of the pyrazolopyrimidine chemotype, a targeted kinase inhibitor library screen revealed the pteridine chemotype VII 2-(5-chloro-2-fluorophenyl)-N-(pyridin-4-yl)pteridin-4-amine (7, SD-208) as an ATP-competitive, cell-active pan-PKD inhibitor.12 The SAR profile of 7 proved to be rather narrow, and among diverse substitutions, only analogs with the 4-aminopyridine moiety maintained a low nanomolar potency. For the preparation of the lead compound as well as most analogs, methyl 3-aminopyrazine-2-carboxylate (84) was converted to the imide 86 with an excess of acid chloride 85 (Scheme 11). Cyclization of 86 in the presence of ammonium hydroxide provided the pteridine 87 in high yield. Anilines such as 88 were then fused to the heterocyclic core in the presence of the coupling reagent PyBOP to give target compound 7.
Scheme 11.
Preparation of Pteridine 712
Chemotype VIII has not yet been rendered specific for PKD but was found to be a potent inhibitor of this kinase in an off-target panel assay of the aurora kinase inhibitor AT9283 (8).13 This pyrazol-4-yl-urea is best described as a multitargeted kinase inhibitor, and interestingly, several benzimidazole fragments also showed submicromolar PKD activity.3 Therefore, it is quite feasible that more PKD-selective analogs of 8 could be developed in future SAR campaigns.
Quite recently, a series of quinazolines such as 9 (NK-176, N-(2-(4-methylpyridin-2-yl)quinazolin-4-yl)benzo[d]oxazol-6-amine) was reported in a phenotypic assay for kinase regulators of lysosome function.14 Several of these quinazolines proved to have submicromolar PKD1 inhibitory activities. This chemotype IX is structurally related to the previously reported chemotypes III, VII, and VIII and has the potential to provide more potent analogs upon further optimization. Interestingly, 9 and its analogs have also been found as potent inhibitors of activin-like kinase (ALK5) with moderate activity on p38,44 thus further highlighting the structural connection between PKD and these kinases and the need to perform kinase panel studies in order to determine particular selective PKD and multitargeted kinase activity profiles. The SAR of chemotype IX is exemplified in Chart 6. Moving from the benzoxazole 9 to the benzothiazole NK-140 did not change the measured PKD1 KD value, but replacing the linker NH with an oxygen atom obliviated the kinase activity in NK-164, which therefore served as a negative control in the transcription factor E3 (TFE3) assays.14 Related benzofuran, benzoxazole, and indole analogs also showed decreased activity, suggesting that the position of the nitrogen atom as a hydrogen-bond acceptor in the benzazole and the position of the NH linker and hydrogen-bond donor are critical requirements for low nanomolar activity.
Chart 6.
Chemical Structures and PKD1 Inhibitory Activities (KD’s) of Representative Chemotype IX Analogs14
Protein kinases can be inhibited reversibly and irreversibly by alkylation of key cysteine residues with suitable Michael acceptors, as exemplified by chemotype X.45 This strategy has also been applied to PKD, which contains a conserved Cys residue in the ATP-binding site that is susceptible to Michael acceptors. The resorcyl lactone hypothemycin (10) demonstrated a 94% inhibition of PKD1 at 2 μM concentration in vitro;15 however, PKD1 was not readily inhibited by 10 in cells.46
Structural Studies of PKD Inhibitors.
The crystal structure of PKD1 (or PKD2,3) is not yet available in the public domain, but several homology models have been built and used for docking studies with known inhibitors.47 Another strategy for studying the binding pose of an inhibitor and applying structure-based design strategies is to solve inhibitor cocrystal structures with surrogate kinases, such as Map4K4.41 In a recent approach, a 3D architecture of PKD1 was generated from a close homologue, the calcium/calmodulin-dependent kinase CaMK-1G (PDB ID: 2JAM), subjected to molecular dynamics (MD) simulations for optimization, and then used to screen drug-like molecules, resulting in a series of interesting new potential inhibitors that have yet to be experimentally verified.48
Figure 5 shows a typical docking analysis for two structurally closely related inhibitors, NPKDi and BPKDi, conducted using AutoDock Vina.49 The hydrophobic pyridines fit the adenosine binding region, replacing the adenosine of ATP in a binding mode typical for an ATP-competitive type I inhibitor. In addition to interactions with the gatekeeper residue M659, the hinge loop that forms the hydrophobic binding pocket II, K612, a highly conserved residue required for the binding of ATP in protein kinases, is also engaged. Modification of the 2,6-naphthyridine in NPKDi with functional groups that are able to hydrogen bond to K612 can result in a significant improvement in the specificity of BPKDi, as shown in the docking model Figure 5C and 5D.35,61
Figure 5.
Docking analyses of PKD1 inhibitors. (A) Structures of inhibitors NPKDi and BPKDi. (B) Docking modes in the ATP binding region. (C and D) Key PKD1 residue interactions.
Biochemical and Cellular Properties of PKD Inhibitors.
PKD small molecular inhibitors have been evaluated both as tool compounds and as potential clinical candidates in various biological systems and disease models. In the following sections, an overview of the in vitro and in vivo applications of the I–X chemotypes of PKD inhibitors is presented, highlighting the most significant findings and their implications for future drug development targeting PKD. This discussion mainly focuses on the cross-comparison of PKD inhibitors from different studies in an effort to reinvigorate PKD-targeted drug development and facilitate a selection of proof-of-concept inhibitors for future studies. Several inhibitor classes have also been reviewed in two previous reviews.3,50
Potency.
Table 1 compares the in vitro properties of representative PKD inhibitors in each chemotype. It is clear that several scaffolds have been optimized to reach single-digit nanomolar potencies, particularly in chemotype III, i.e., NPKDi,35 BPKDi,61 and CRT0066101.36,62 At cellular levels, submicromolar IC50 values have been demonstrated for the three Novartis PKD inhibitors (NPKDi, BPKDi, and 3,5-diarylisoxazole 4) in the range of 32–240 nM for inhibiting PKD1-mediated GFP-HDAC5 nuclear export in cardiac myocytes,9,35,61 resulting from a direct phosphorylation of HDAC5 by PKD1.51 However, despite the remarkable potencies at the biochemical level, the on-target inhibition of PKD1 autophosphorylation at S916 in intact cells stalls in the micromolar range. For example, CRT0066101 inhibits p-S916-PKD1 autophosphorylation with an IC50 of 6 μM in PANC-1 pancreatic cancer cells,3 displaying a remarkable >1000-fold difference between biochemical and cellular potencies. Note that this difference is not uncommon for ATP-competitive inhibitors, although it is yet to be determined whether CRT0066101 competes with ATP. In line with this pattern of activity shifts, the cellular biological activities of these inhibitors are also mostly in the micromolar range with only a few reported to reach submicromolar potency. For example, CRT0066101, the most potent PKD inhibitor in terms of anticancer activity in vitro, has been reported to inhibit tumor cell viability with an IC50 of 0.6–1.9 μM.62 Interestingly, chemotype II compounds (CID755673, kb-NB142-70, kmg-NB4-23), despite being noncompetitive with ATP, also exhibit a significant shift (<100-fold) in IC50 from biochemical to cellular environments.5,60 Binding to serum proteins might partially account for a potency drop; however, kb-NB142-70 was 78% plasma protein bound at both 0.3 and 3 μg/mL, and kb-NB165-09 was 88% and 94% bound to plasma protein at 0.3 and 3 μg/mL, respectively.7 These relatively low levels of protein binding imply that other factors in addition to ATP concentration and plasma protein binding affect the activity of PKD inhibitors in cells. These factors might primarily influence the bioavailability of the inhibitors and include general compound stability, solubility, size, and cell permeability properties as well as effects of efflux pumps. For example, the TPSA of CRT0066101 is 102 Å2,39 which suggests that the passive molecular transport of this molecule through cell membranes is likely a limiting factor. The impact of these ATP-independent properties could be evaluated by comparing the target binding affinity of the inhibitors in different experimental settings. Currently, there is scant information on the binding properties of PKD inhibitors; only two (compound 139 and NK-176) were evaluated in an active-site-directed competition binding assay (KINOMEscan) for kinase profiling. NK-176 was also examined using KiNativ, which measures small molecule binding to kinases in cell lysates, providing an opportunity to assess the impact of cellular factors other than ATP on the bioavailability of the inhibitor. Interestingly, despite the high binding affinity (KD = 53 nM) at the biochemical level, NK-176’s cellular binding activity was much lower, ranging from 17% to 59% for PKD1–3 at 2 μM (correlating to a probable >40× drop in binding affinity) measured in the KiNativ assay, suggesting that ATP-independent factors could significantly impact the efficacy of the PKD inhibitors in cells. In addition, tumor cells are also known to have higher levels of ATP, which can further reduce the cellular efficacy of inhibitors.
Table 1.
Summary of Known PKD-Targeted SMIsa
chemotype | compound name | biochemical IC50 (nM) | cellular IC50b (μM) | kinases inhibitedc (#/#,d %,e μMf) | ref |
---|---|---|---|---|---|
I | staurosporine | 27 (PKD1) | n.d. | nonselective | 58 |
72 (PKD2) | |||||
18 (PKD3) | |||||
ATP competitive | |||||
K252a | 7 (PKD1) | n.d. | nonselective | 4 | |
ATP competitive | |||||
Gö6976 | 20 (PKD1) | n.d. | cPKC (no profiling data) | 4, 59 | |
ATP competitive | |||||
Gö6983 | 20 000 (PKD1) | n.d. | c/n/aPKC (no profiling data) | 4, 59 | |
20 000 (PKD1) | |||||
II | CID755673 | 182 (PKD1) | 11.8 (PKD1), PKD1 autophosphorylation at S916 in LNCaP cells | MK2, GSK-3/9, CK1δ, MK5/PRAK, CDK2, and ERK1 (6/48, ≥50%, 10 μM) | 5, 22, 60 |
280 (PKD2) | |||||
227 (PKD3) | |||||
non-ATP competitive | |||||
kb-NB142-70 | 28 (PKD1) | 2.2 (PKD1), PKD1 autophosphorylation at S916 in LNCaP cells | similar to CID755673 | 6, 60 | |
59 (PKD2) | |||||
53 (PKD3) | |||||
non-ATP competitive | |||||
kmg-NB4-23 | 120 (PKD1) | 6.8 (PKD1), PKD1 autophosphorylation at S916 in LNCaP cells | n.d. | 5, 6 | |
non-ATP competitive | |||||
III | NPKDi (13c) | 0.6 (PKD1) | 0.032 (PKD1), GFP-HDAC5 nuclear export in cardiac myocytes | GSK-3α/β, hCaMKIIδ, hMARK1/2 (8/197, > 80%, 1 μM) | 35, 61 |
BPKDi (12a) | 1 (PKD1) | 0.077 (PKD1), GFP-HDAC5 nuclear export in cardiac myocytes | none (3/197, > 80%, 1 μM) | 61 | |
9 (PKD2) | |||||
1 (PKD3) | |||||
CRT0066101 | 1 or 9 or 16 (PKD1) | 6.0 (PKD1), PKD1 autophosphorylation at S916 in PANC-1 cells | CDK5, DYRK2, Pim-1 (5/127, > 90%, 1 μM) |
36, 40, 62 |
|
2.5 or 8.1 or 9 (PKD2) | |||||
2 or 7.7 or 6 (PKD3) | |||||
IV | CRT0066051 | 1 or 9.2 (PKD1) | 6.6 (PKD1), PKD1 autophosphorylation at S916 in PANC-1 cells | BRK (3/127, >90%, 1 μM) | 38, 40 |
2 or 8.4 (PKD2) | |||||
1.5 or 7.6 (PKD3) | |||||
3,5-diarylisoxazole 4 | 5.5 (PKD1) | 0.24 (PKD1), GFP-HDAC5 nuclear export in cardiac myocytes | (4/230, ≥ 50%, 1 μM) | 9 | |
48 (PKD2) | |||||
17 (PKD3) | |||||
V | compound 139 | 16.8 (PKD1) | 1.5 (PKD1), PKD1 autophosphorylation at S916 in LNCaP cells | (43/353, ≥50%, 10 μM); p38α/β, JNK1/2, STK36/21, CK1ε (8/353, 99–100%, 10 μM)g | 10 |
VI | 1-NA-PP1 | 155 (PKD1) | 22.5 (PKD1), PKD1 autophosphorylation at S916 in LNCaP cells | p38α, CK1δ, Src, Lck, CSK, RIP2 (7–11/~70, >50%, 1 μM) | 11, 63 |
133 (PKD2) | |||||
104 (PKD3) | |||||
3-IN-PP1 | 108 (PKD1) | effective at 20 μM for blocking PDBu-stimulated cortactin phosphorylation at S298 | n.d. | 43, 62 | |
94 (PKD2) | |||||
108 (PKD3) | |||||
compound 17m | 35 (PKD1) | effective at 20 μM for blocking PDBu-stimulated cortactin phosphorylation at S298 | n.d. | 62 | |
35 (PKD2) | |||||
17 (PKD3) | |||||
VII | SD-208 | 107 (PKD1) | 17.0 (PKD1), PKD1 autophosphorylation at S916 in LNCaP cells | TGFβR1 (no profiling data) | 12 |
94 (PKD2) | |||||
105 (PKD3) | |||||
ATP competitive | |||||
VIII | AT9283 (8) | 10 (PKD1) | n.d. | aurora kinase A/B, JAK2/3, Abl (T315I), Flt-3, CDK2, etc. (28/144, IC50 > 30 nM) | 13 |
IX | NK-176 | 53 (PKD1) | n.d. | none (3/392, >90%, 1 μM) | 14 |
X | hypothemycin | 1200 (PKD1) | not effective up to 4 μM on PKD1 autophosphorylation | n.d. | 15, 46 |
covalent inhibitor |
The in vitro and cellular IC50 values of known PKD SMIs are listed.
Cellular IC50, inhibition of PKD1 activity (autophosphorylation at S916), or PKD1-regulated downstream event (HDAC5 nuclear export) in intact cells.
PKD is not listed but included when counting the number of hits.
Number of hits (including PKD)/total number of kinases profiled.
Percent inhibition as cutoff in kinase profiling.
Concentration used in kinase profiling.
Kinase profiling was performed on compound 140,10 which is similar in structure and activity to compound 139. n.d. not determined.
Selectivity.
Another key aspect of protein kinase inhibitor development is selectivity. Kinase profiling data at variable scale levels are available for several PKD inhibitors. Among them, NK-176 appears to be a remarkably selective PKD inhibitor. In a large-scale kinase profiling analysis encompassing 392 kinases, PKD1–3 emerged as the sole hits with >90% inhibition at 1 μM, demonstrating an almost exclusive specificity for the PKD family.14 Additionally, the selectivity profile of 3,5-diarylisoxazole 4 was also impressive.9 In a kinase profiling analysis of 230 kinases, only 4 hits with over 50% inhibition at 1 μM were identified.9 CRT0066101 and CRT0066051 also show high selectivity for PKD with only three (CDK5, DYRK2, Pim-1) and one (BRK) additional targets inhibited >90% at 1 μM besides PKD in a profiling involving 127 kinases. BPKDi exhibited an improved selectivity profile compared to NPKDi with no additional kinases inhibited >80% at 1 μM besides PKD1–3 in a panel of 197 kinases, as compared to five (GSK-3α/β, hCaMKIIδ, hMARK1/2) hits for NPKDi inhibited at >80% out of 197 kinases examined at 1 μM.35,61 The 4-azaindole PKD inhibitors compounds 139 and 140, also reported as potent p38α/β inhibitors, similarly exhibited an attractive selectivity profile with eight (p38α/β, JNK1/2, STK36/21, CK1ε) additional kinases besides PKD inhibited at the 99–100% level at 10 μM in a large-scale profiling involving 353 kinases.10 Compared to these highly selective PKD inhibitors that belong to chemotypes III, IV, V, and IX, inhibitors classified in chemotypes II, VI, and VIII are relatively less selective based on the number of off-target kinases, the profiling scale, and the concentration of inhibitor used in the analysis. Structural studies of chemotypes with favorable selectivity profiles may therefore provide additional insights to aid in the design of more selective PKD inhibitors.
In summary, extensive in vitro analyses have been conducted on PKD inhibitors over several decades. It is clear from these studies that highly potent and selective PKD inhibitors of distinct structural classes have been successfully identified. Some of these inhibitors exhibit nanomolar potencies for PKD and on-target cellular PKD inhibition in cells in the low micromolar range. They also exert a wide variety of biological activities at cellular levels that are in agreement with the multitude of PKD biological functions in different biological systems. Future work should be directed at side-by-side analyses of these inhibitors at biochemical and cellular levels to validate the results reported in a diverse assay systems and performed by different research groups. In particular, kb-NB142-70, CID2011756, CRT0066101, CRT0066051, compound 139, Im-5, and NK-176 would form an attractive panel for such a cross-comparison.
In Vivo Evaluation of PKD Inhibitors.
PKD plays an important role in the pathogenesis of multiple diseases, including stress-induced cardiac hypertrophy, inflammatory diseases, metabolic disorders, and cancer. A total of six different PKD inhibitors from chemotypes I (Gö6976), II (CID755673), III (NPKDi, BPKDi, CRT0066101), and VII (SD-208) have been evaluated in vivo. These preclinical studies can be separated into two cohorts based on the disease models being (1) acute, which only requires short-term inhibitor application, or (2) chronic, which requires repeated long-term administration of inhibitors. In the following sections, inhibitors that have been tested in these two types of in vivo applications will be discussed. Table 2 provides a detailed comparison of the application of PKD inhibitors in these animal models, detailing their doses, routes, and schedules of administration, the animal species used, the outcome of the evaluation, and their underlying mechanisms of actions.
Table 2.
In Vivo Application of PKD Inhibitorsa
chemotype | compound | disease | dose, route, schedule and species | in vivo efficacy | mechanisms | ref |
---|---|---|---|---|---|---|
I | Gö6976 | bone repair | 2.35 mg/kg, ip, once daily for 4 days before growth plate tissue collection at day 10 in rats | reduction in bone formation and more cartilage repair in the growth plate injury site in rats | by decreasing bone-related genes (osterix, osteocalcin) and increasing cartilage-related genes (Sox9, collagen-2a, collagen-10a1) | 64 |
bacterial infection-induced inflammation, liver injury, sepsis | (1) 2.5 mg/kg in DMSO, ip, once 30 min before LPS/D-GalN challenge in mice | (1) inhibited LPS/D-GalN-induced acute liver injury in mice | (1) by inhibition of MAPKs activation to reduce TNF-α production in liver | 65, 66 | ||
(2) 2.3 mg/kg in 7.6% DMSO in PBS, ip, 1 and 4 h before or 2 h after the antibiotic killed the GBS challenge | (2) protected d-GalN-sensitized mice from shock-induced death caused by antibiotic-killed GBS | (2) by inhibiting MAPKs and NF-kB and expression of proinflammatory cytokines/chemokines | ||||
II | CID755673 | pancreatitis | 15 or 20 mg/kg in DMSO, ip, once 60 min before or after cerulein treatment in rats | reduction in necrosis, inflammatory responses, and severity of pancreatitis and similar effects in alcoholic pancreatitis | by inhibiting NF-κB-mediated inflammatory responses, trypsinogen activation, and several cell death mediators (ATP, Bcl-2, RIPK1) | 67-69 |
diabetes | 10 mg/mL in 50% DMSO, osmotic pump, 2 weeks in mice | accelerated onset of diabetes by increasing fasting glucose in BTBRob/ob mice and decreased insulin secretion | by exacerbating stress-induced nascent granule degradation (SINGD) and inhibiting insulin secretion | 70 | ||
1 or 10 mg/kg, ip, once (acute) or once daily for 16 days (chronic) in mice | enhanced heart function and reduced heart weight in type 2 diabetes db/db mice | by improving both systolic and diastolic cardiac function with unknown mechanisms | 71 | |||
myocardial infarction | 10 mg/kg in saline, iv, every other day for 4 weeks in rats | inhibited astragaloside IV-induced angiogenesis of myocardial tissue in rats with myocardial infarction | by blocking the PKD1-HDAC5-VEGF pathway | 72 | ||
learning and memory | 182 nmol, infusion into the bilateral hippocampal CAI areas, before DHPG (a group I mGluRs) agonist in rats | prevented DHPG-induced learning and memory impairments in rats | possibly by affecting NMDAR internalization and activity | 73 | ||
III | NPKDi (13c) | cardiac hypertrophy | (1) 5, 15, 50 mg/kg in vehicle (0.5% methylcellulose and 0.5% Tween-80), po, once daily for 14 days in a DSS rat model | reduced high salt-induced cardiac hypertrophy at the 50 mg/kg po dose, but no effect in pressure overload-induced cardiac hypertrophy at the 15 mg/kg sc dose | by blocking the phosphorylation and nuclear export of the MEF2 repressor HDAC in cardiac myocytes | 35 |
(2) 15 mg/kg in acidified captisol, sc, once daily for 12 days in a TAB rat model | ||||||
BPKDi (12a) | cardiac hypertrophy | 0.5, 1.5, 5 mg/kg in acidified captisol, sc, once daily for 14 days in rat models | did not affect cardiac hypertrophy at 5 mg/kg sc dose in high salt- and pressure overload-induced cardiac hypertrophy rat models | by inhibiting phosphorylation and nuclear export of HDAC4/5 in cardiac myocytes in vitro | 61 | |
CRT0066101 | pancreatic cancer | 80 mg/kg in 5% dextrose, po, once daily for 21 days in nude mice | inhibited PANC-1 tumor xenograft growth in a subcutaneous and an orthotopic pancreatic cancer models in nude mice | by blocking NF-κB activation and NF-κB-mediated tumor cell proliferation and survival | 36 | |
80 mg/kg in 5% dextrose, po, every other day for 14 days before sacrifice at day 24 in immunocompetent MD4 mice | suppressed the growth of orthotopic KPC4662 pancreatic xenograft tumors in immunocompetent mice | by inhibiting cancer-cell autonomous tumor-promoting function of PKD in PDAC xenografts | 74 | |||
colon cancer | 40, 80, 120 mg/kg in 5% dextrose, po, once daily for 21 days in mice | inhibited HCT116 colorectal tumor xenograft growth in nude mice | by inducing G2-M arrest and apoptosis via inhibiting Akt, ERK, and NF-κB in colorectal cancer cells | 75 | ||
breast cancer (ER− and TNBC) | 80 mg/kg in 5% dextrose, po, every other day for 8 weeks in NOD scid mice | inhibited primary tumor growth, local invasion, and metastasis to distant organs in an orthotopic mouse tumor xenograft model of ER− breast cancer | by blocking the upregulated PKD3-mediated tumor cell proliferation, survival, and invasion in ER− cancer cells | 76 | ||
65 mg/kg, po, 5 times a week for 4 weeks in nude mice | inhibited subcutaneous MCF-7-ADR tumor xenograft growth in nude mice | by suppressing breast cancer stemness through GSK3/μ-catenin signaling | 77 | |||
50 or 80 mg/kg in 5% dextrose, po, once daily for 3 or 6 weeks in nude mice | inhibited TNBC tumor xenograft growth in nude mice | by causing G1 arrest and regulating phosphorylation of MYC, MAPK1/3, AKT, YAP, and CDC2 and by targeting a PKD3/CLU pathway in TNBC | 78, 79 | |||
bladder cancer | 120 mg/kg in 5% dextrose, po, 3 days per week for 25 days in nude mice | inhibited bladder tumor xenograft growth in nude mice | by inducing cell cycle arrest via reducing CDK1/cyclin B complex activity | 80 | ||
liver metastasis | 80 mg/kg in 5% dextrose, po, once daily for 28 days in nude mice | reduced lung metastasis of subcutaneously implanted hepatocellular carcinoma cells in nude mice | by inhibiting TNFα-induced EMT and HCC metastasis | 81 | ||
tumor immune response | 50 mg/kg in PBS, ip, once at day 7 before sacrifice at day 21 alone or in combination with PD-1 in C57BL/6 mice | blocked the anticancer effect of ant-PD-1 therapy | by antagonizing T-cell reactivation by anti-PD-1 therapy via inhibiting Akt | 82 | ||
Obesity and diabetes | 10 mg/kg in water, po, daily for 13 weeks in C57BL/6 mice | decreased intestinal fat absorption, reduced high-fat diet-induced obesity and diabetes and improved gut microbiota profile in mice | by inhibiting triglycerides absorption in the intestine by regulating APOA4 | 55 | ||
anxiety in myocardial infarction (MI) | 80 mg/kg, po, once daily for 14 days in rats | attenuated anxiety-like behaviors in MI rats | by blocking platelets granule release, thereby reducing serum IGF1 in MI rats | 83 | ||
pancreatitis | 20 mg/kg in DMSO, ip, once 60 min before or after cerulein treatment in rats | reduction in inflammatory responses, cell necrotic death and the severity of disease in alcoholic pancreatitis | by inhibiting NF-κB, trypsinogen activation, and several cell death mediators (ATP, Bcl-2, RIPK1) | 69 | ||
VII | SD-208 | prostate cancer | 60 mg/kg in 1% methylcellulose, po, twice daily for 21 days in nude mice | abrogated the growth of PC3 subcutaneous tumor xenografts in nude mice | by inducing G2/M arrest through induction of p21 and increased p-Cdc2 and downregulation of Cdc25c | 12 |
The list contains PKD inhibitors that have been examined in vivo, including their dosages, routes, and schedules of administration, animal species used, in vivo efficacy, and underlying mechanisms in various disease models.
Short-Term Application of PKD Inhibitors In Vivo.
The diseases evaluated in this category are primarily inflammatory diseases, including pancreatitis and bacterial infection-induced inflammatory conditions. Under these acute conditions, PKD inhibitors were mostly administered once before or after the initiation of the pathological conditions, and the animals were evaluated within a few hours to several days. Besides the studies discussed below, a short-term application of PKD inhibitors has also been also reported in a study on bone repair using Gö697664 and on learning and memory functions using CID755673,73 as shown in Table 2.
Inflammatory Conditions Caused by Bacterial Infection.
The PKC/PKD inhibitor Gö6976 was evaluated in two studies involving bacterial infection-induced inflammation, liver injury, and septic shock. It was found that one-time intraperitoneal (ip) injection of Gö6976 at 2.5 mg/kg 30 min before the LPS/d-galactosamine (d-GalN) challenge that mimics bacterial infection blocked liver injury caused by LPS/D-GalN. Gö6976 acts by inhibiting MAPKs and subsequent TNF-α production in mouse liver.65 In a similar study in d-GalN-sensitized mice, one-time ip injection of 2.3 mg/kg Gö6976 before or after GBS challenge significantly protected mice from shock-induced death by inhibiting MAPKs and NF-kB and decreasing expression of the proinflammatory cytokines/chemokines.66 Since Gö6976 is a dual inhibitor of PKC and PKD, it is possible that its anti-inflammatory effects are due to the inhibition of both kinases.
Inflammatory Injuries in Pancreatitis.
Acute pancreatitis is an inflammatory condition of the pancreas that causes pancreatic acinar cell necrosis and in its severe form could lead to single or multiple organ damage and death. In three separate reports, the protective effects of two PKD inhibitors, CID755673 and CRT0066101, were demonstrated in a rat model of cerulein-induced pancreatitis. A single ip injection of CID755673 at 15 mg/kg before or after cerulein treatment significantly reduced inflammatory responses, necrosis, and severity of pancreatitis.67,68 Both CID755673 and CRT0066101 given once at 20 mg/kg ip before or after cerulein treatment also effectively attenuated inflammation, necrotic cell death, and disease severity in an alcohol-aggravated pancreatitis.69 Mechanistically, these inhibitors mainly act through inhibiting NF-κB-mediated inflammatory responses67,68 and by modulating mitochondrial ATP production, Bcl-2 family proteins, and RIP1 kinase, as well as trypsinogen activation.69 In these studies, the anti-inflammatory and protective effects of CID755673 are compelling in both alcoholic and nonalcoholic pancreatitis, indicating a broader applicability. Since CID755673 is less potent and selective compared to CRT0066101 whose effect was only apparent for alcoholic pancreatitis, it is possible that the beneficial effects of CID755673 may involve other off-target kinases. Nonetheless, the anti-inflammatory activities of CID755673 in vivo remain attractive for future drug development.
Long-Term Application of PKD Inhibitors In Vivo.
PKD inhibitors have been evaluated in several preclinical disease models that required repeated application over an extended period of several weeks to months. Below, we provide a review on the three main diseases in this category, i.e., cardiac hypertrophy, metabolic disorders (obesity and diabetes), and cancer. PKD inhibitors in chemotypes II (CID755673), III (NPKDi, BPKDi, CRT0066101), and VII (SD208) have been evaluated in this setting. Besides these diseases, two studies reported the use of PKD inhibitors in myocardial infarction (CID755673) and anxiety associated with myocardial infarction (CRT0066101), details of which are listed in Table 2.
Cardiac Hypertrophy.
Studies using genetically modified mouse models have firmly established PKD1 as a therapeutic target for stress-induced cardiac hypertrophy.52,53 This has spurred significant interest in developing PKD inhibitors as potential therapeutic agents for cardiac hypertrophy. Novartis led this effort by contributing three highly potent and selective PKD inhibitors (NPKDi, BPKDi, and 3,5-diarylisoxazole 4). Two of these (NPKDi, BPKDi) have been evaluated in vivo in two distinct rat models of cardiac hypertrophy (high salt and pressure overload). The NPKDi 13c, despite being less selective, significantly reduced both PKD autophosphorylation and high salt-induced cardiac hypertrophy given orally at 50 mg/kg, but it had no effect in pressure overload-induced cardiac hypertrophy when administered subcutaneously at 15 mg/kg. However, BPKDi 12a, a more selective analog of NPKDi, was not effective at inhibiting PKD autophosphorylation and cardiac hypertrophy when administered subcutaneously at 5 mg/kg. It appears that blood pressure increase was one of the dose-limiting factors for BPKDi. Unfortunately, no further development in this area has been reported since 2010.
Diabetes and Obesity.
Although growing evidence has identified PKD as a critical metabolic regulator in multiple organs,2,54 PKD inhibitors have not been examined extensively in metabolic diseases. PKD modulates insulin secretion from pancreatic β cells, and its dysregulation results in hyper-insulinemia and systemic insulin resistance that underlie metabolic disorders.2,54 In Type 2 diabetes, metabolic stress causes elevated stress-induced nascent insulin granule degradation (SINGD), a process negatively regulated by PKD. It was found that inhibition of PKD by CID755673 applied by osmotic pump for 2 weeks accelerated diabetes by increasing SINGD and reducing insulin secretion.70 On the other hand, CID755673 given ip once daily for 16 days at 1 or 10 mg/kg led to enhanced cardiac function in diabetic db/db mice,7 implying that inhibition of PKD may improve heart functions in obese and diabetic patients. The efficacy of CID755673 in chronic conditions that require long-term treatment is encouraging and somewhat surprising given the poor PK profile of its analog kb-NB142-70. Also, off-target effects of CID755673 could not be ruled out entirely. Another inhibitor that has been evaluated in metabolic disorders is CRT0066101. In a mouse model of high-fat-diet-induced obesity and diabetes, CRT0066101 given orally at 10 mg/kg daily for 13 weeks resulted in reduced intestinal fat absorption, decreased obesity and associated diabetes, and an improved gut microbiota profile in mice via inhibiting intestinal triglycerides absorption by regulating apolipoprotein A4 (APOA4).55 Despite these promising early findings, more studies are needed to confirm the therapeutic potential of PKD inhibitors in metabolic disorders.
Cancer.
CRT0066101, developed by Cancer Research Technology (CRT), is the most intensively studied PKD inhibitor in preclinical mouse models of cancer. In 2010, a joint report by the Guha group from MD Anderson and CRT identified CRT0066101 as an anticancer agent for pancreatic cancer.36 It was found to suppress the growth of subcutaneous and orthotopic PANC-1 pancreatic tumor xenografts in vivo when given orally at its maximum tolerated dose of 80 mg/kg/day for 3 weeks. No apparent signs of acute toxicity were observed during the treatment period as reflected by a stable body weight. The compound was found to have a half-life of 60 min and ~100% bioavailability. The optimal therapeutic concentration of 8 μmol/L was reached at 6 h after oral administration.36 This favorable PK/PD profile set the stage for the subsequent evaluation of this inhibitor in multiple cancer models by different research groups. As detailed in Table 2, the anticancer efficacy of CRT0066101 has been demonstrated in colon cancer,75 breast cancer including estrogen receptor negative (ER−),76 triple-negative breast cancer (TNBC),77 and bladder cancer.80 Notably, it was also effective at blocking lung metastasis of subcutaneously implanted hepatocellular carcinoma cells in nude mice,80 In these studies, CRT0066101 was given orally at a dose ranging from 40 to 120 mg/kg with 80 mg/kg/day being the most frequently used dosing regimen. Mechanistically, CRT0066101 inhibits tumor cell proliferation and survival by inducing G2/M arrest via inhibiting CDK1/cyclin B complex activity and other key pro-proliferative and pro-survival proteins, including NF-κB, ERK, and Akt.36,75,80 CRT0066101 has also been shown to induce G1 arrest, possibly through regulating MYC, MAPK1/3, AKT, YAP, and CDC2.77 In addition to CRT0066101, our group showed that SD-208, a dual inhibitor of PKD and TGFβR1, was also effective at inhibiting the growth of subcutaneous prostate cancer tumor xenografts in mice. This compound similarly induced G2/M arrest by modulating the activity of the CDK1/cyclin B complex.12 It is interesting to note that a recent study demonstrated a concerning effect of CRT0066101 in antagonizing the anti-PD-1 immunotherapy by blocking AKT reactivation in T cells.55 However, another study showed that inhibition of PKD could abrogate immunosuppressive properties of B cells and thereby promote effector T-cell function.74 In the later study, CRT0066101 suppressed the growth of orthotopic pancreatic xenograft tumors in immunocompetent mice, demonstrating its viability as an anticancer agent despite its potential complications in the immune system. Overall, these studies have established CRT0066101 as an orally effective anticancer agent in multiple solid tumor models. However, the potentially dose-limiting solubility and long-term toxicity profile of CRT0066101 would need to be further elaborated and, if needed, improved by structural modifications.
PKD Inhibitors in Combination Therapy.
One area that deserves significant future attention is the use of PKD inhibitors in combination therapy. Previous studies have mainly focused on their use in combination therapy for cancer, and the outcomes have been mixed. PKD inhibitors have been shown to synergize with several chemotherapeutics, including paclitaxel56 and cisplatin,57 as well as targeted agents such as the multikinase inhibitor regorafenib.75 Specifically, inhibition of PKD by CRT0066101 significantly enhanced the cytotoxic effect of regorafenib in multiple colon cancer cell lines by synergistically inducing apoptosis and downregulating ERK, AKT, and NF-κB signaling.75 The combination of CRT0066101 and paclitaxel synergistically reduced oncosphere and colony formation in TNBC cells and, most importantly, reduced tumor recurrence in vivo in an orthotopic breast cancer mouse model.56 Another PKD inhibitor, CID2011756,37 enhanced the sensitivity of cancer cells to cisplatin or carboplatin. This PKD inhibitor increased proteasome-mediated degradation of ATPase copper-transporting α/β (ATP7A/B) by blocking phosphorylation.57 Since the selectivity of CID2011756 is unknown, it remains to be determined if and how much inhibition of PKD1–3 accounts for its synergistic effects. It should be noted that the combined inhibition of PKD is not always desirable, and there is evidence that activating it could also be beneficial, such as the case of suramin (a PKD activator) and a DNA methyltransferase inhibitor combination for breast cancer treatment, where a role of PKD1 in tumor suppression is implicated.20 In addition, it has been shown that CRT0066101, by inhibiting Akt activation and T-cell function, accelerated tumor growth and reversed the therapeutic effect of anti-PD-1 therapy in a syngeneic mouse tumor models.82 Overall, further in-depth studies are needed in this area to fully realize the utility of PKD inhibitors in combination therapy.
Perspectives and Future Directions.
After the first reports on the discovery and biochemical characterization of PKD, considerable enthusiasm for this new target drove a 15 year period of rapid growth, culminating in a vigorous preclinical development of a significant number of structurally distinct, PKD-selective chemotypes. However, these early lead structures still suffer from a lack of selectivity between the three PKD subtypes and decreased potency in cells and in vivo and did not thoroughly address potential on-target and off-target toxicity. Initial clinical entry strategies also diverged between cardiac diseases and cancer; PKD was found to regulate myofilament function84 and influence blood pressure downstream of angiotensin II,85,86 which complicates the design of human trials of small molecule pan-PKD modulators. While mechanistic and cell-based studies on PKD inhibitors are continuing, major therapeutic breakthroughs have yet to be realized. Different PKD isoforms have seemingly opposing functions in cancer; accordingly, future clinical studies in this therapeutic field will likely require isoform-selective inhibitors. In consideration of the very high level of structural homology between PKD1, PKD2, and PKD3, particularly in the vicinity of the active site, it would be very attractive to identify allosteric inhibition strategies in order to evaluate the therapeutic profile of isoform-selective agents. In particular, for noncancer-related applications, high subtype selectivity might be a key requirement in a clinical setting. Other current gaps in the field include the scarcity of in vivo toxicity profiles of inhibitor chemotypes as well as the lack of unique and reliable biomarkers that could be used to track PKD response in patients. A side-by-side, comprehensive evaluation of different classes of PKD inhibitors would support these efforts. Furthermore, alternative classes of PKD modulators (nucleotide based, peptide based, etc.) would also provide helpful new tool compounds. Finally, as with other experimental therapeutics, drug combinations are a particularly attractive opportunity for clinical studies. In light of the large body of published research on PKD, including its medicinal chemistry and wealth of biological functions (Figure 6), as well as the broad portfolio of modern targeting strategies and structural biology tools, another growth period in PKD-driven pharmaceutical research and development is likely to emerge in the near future.
Figure 6.
PKD as a potential therapeutic target in human diseases, illustrating key signaling targets of PKD and diverse PKD-regulated biological processes in four major types of human diseases.2 Created with BioRender.com.
ACKNOWLEDGMENTS
The authors gratefully acknowledge financial support from the Academy of Finland PROFI6 program (P.W.), the National Institutes of Health 5R01CA229431 (Q.J.W.), and the American Heart Association 19TPA34850096 (Q.J.W.). We also thank Dr. Sen Liu (Hubei University of Technology, Hubei, China) for performing the docking studies (Figure 5).
ABBREVIATIONS USED
- APOA4
apolipoprotein A4
- BTBRob/ob
black and tan brachyuric obese/obese mouse
- CLU
clusterin
- DBU
1,8-diazabicyclo[5.4.0]undec-7-ene
- DDQ
2,3-dichloro-5,6-dicyano-1,4-benzoquinone
- DHP
dihydropyran
- DHPG
(S)-3,5-dihydroxyphenylglycine
- d-GalN
d-galactosamine
- DSS
Dahl salt sensitive
- EDCI
1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
- ESC
embryonic stem cell
- GBS
group B streptococci
- FP
fluorescence polarization
- ip
intraperitoneal injection
- mGluRs
metabotropic glutamate receptors
- MI
myocardial infarction
- PDAC
pancreatic ductal adenocarcinoma
- PKD
protein kinase D
- PKD1
protein kinase D1
- PKD2
protein kinase D2
- PKD3
protein kinase D3
- po
oral
- PyBOP
benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate
- n.d.
not determined
- sc
subcutaneous
- SINGD
stress-induced nascent granule degradation
- TAB
thoracic aortic banded
- TFAA
trifluoroacetic anhydride
- TNBC
triple-negative breast cancer
- TPSA
topological polar surface area
Biographies
Qiming Jane Wang received her B.S. degree in Microbiology (1989) from Xiamen University in China. Thereafter she moved to the United States and obtained her Ph.D. degree in Biochemistry (1995) from Creighton University. After completing her postdoctoral training with Dr. Peter M. Blumberg at the National Cancer Institute, she joined the faculty of the University of Pittsburgh School of Medicine and is currently a Professor at the Department of Pharmacology and Chemical Biology at Pitt. Her current research mainly focuses on oncogenic protein kinases in the diacylglycerol signaling network and small molecule drug discovery and development targeting these kinases.
Peter Wipf received his Ph.D. degree in 1987 from the University of Zurich under the guidance of Professor Heinz Heimgartner. After 2 years as a Swiss NSF postdoctoral fellow with Professor Robert E. Ireland at the University of Virginia, he joined the University of Pittsburgh in 1990 and was promoted to Distinguished University Professor in 2004. He has secondary appointments in Pharmaceutical Sciences and in Bioengineering, and he is a coleader of the Cancer Therapeutics Program at the UPMC Hillman Cancer Center. His research expertise includes medicinal chemistry, total synthesis of natural products, heterocyclic chemistry, and study of strain-activated carbo- and heterocycles. He has coauthored more than 600 manuscripts, and he is a fellow of the AAAS, the RSC, and the ACS.
Footnotes
The authors declare no competing financial interest.
Contributor Information
Qiming Jane Wang, Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States.
Peter Wipf, Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States; School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland.
REFERENCES
- (1).Valverde AM; Sinnett-Smith J; Van Lint J; Rozengurt E Molecular Cloning and Characterization of Protein Kinase D: A Target for Diacylglycerol and Phorbol Esters with a Distinctive Catalytic Domain. Proc. Natl. Acad. Sci. U.S.A 1994, 91 (18), 8572–8576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (2).Zhang X; Connelly J; Chao Y; Wang QJ Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021, 11 (3), 483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (3).Gilles P; Voets L; Van Lint J; De Borggraeve WM Developments in the Discovery and Design of Protein Kinase D Inhibitors. ChemMedChem 2021, 16 (14), 2158–2171. [DOI] [PubMed] [Google Scholar]
- (4).Gschwendt M; Dieterich S; Rennecke J; Kittstein W; Mueller H-J; Johannes F-J Inhibition of Protein Kinase Cμ by Various Inhibitors. Inhibition from Protein Kinase C Isoenzymes. FEBS Lett. 1996, 392 (2), 77–80. [DOI] [PubMed] [Google Scholar]
- (5).George KM; Frantz M.-C. l.; Bravo-Altamirano K; LaValle CR; Tandon M; Leimgruber S; Sharlow ER; Lazo JS; Wang QJ; Wipf P Design, Synthesis, and Biological Evaluation of PKD Inhibitors. Pharmaceutics 2011, 3 (2), 186–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (6).Bravo-Altamirano K; George KM; Frantz M-C; LaValle CR; Tandon M; Leimgruber S; Sharlow ER; Lazo JS; Wang QJ; Wipf P Synthesis and Structure-Activity Relationships of Benzothie-nothiazepinone Inhibitors of Protein Kinase D. ACS Med. Chem. Lett 2011, 2 (2), 154–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (7).Guo J; Clausen DM; Beumer JH; Parise RA; Egorin MJ; Bravo-Altamirano K; Wipf P; Sharlow ER; Wang QJ; Eiseman JL In Vitro Cytotoxicity, Pharmacokinetics, Tissue Distribution, and Metabolism of Small-Molecule Protein Kinase D Inhibitors, KB-NB142–70 and KB-NB 165–09, in Mice Bearing Human Cancer Xenografts. Cancer Chemother. Pharmacol 2013, 71 (2), 331–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (8).Monovich L; Vega RB; Meredith E; Miranda K; Rao C; Capparelli M; Lemon DD; Phan D; Koch KA; Chapo JA; Hood DB; McKinsey TA A Novel Kinase Inhibitor Establishes a Predominant Role for Protein Kinase D as a Cardiac Class IIa Histone Deacetylase Kinase. FEBS Lett. 2010, 584 (3), 631–637. [DOI] [PubMed] [Google Scholar]
- (9).Gamber GG; Meredith E; Zhu Q; Yan W; Rao C; Capparelli M; Burgis R; Enyedy I; Zhang J-H; Soldermann N; Beattie K; Rozhitskaya O; Koch KA; Pagratis N; Hosagrahara V; Vega RB; McKinsey TA; Monovich L 3,5-Diarylazoles as Novel and Selective Inhibitors of Protein Kinase D. Bioorg. Med. Chem. Lett 2011, 21 (5), 1447–1451. [DOI] [PubMed] [Google Scholar]
- (10).Tandon M; Wang L; Xu Q; Xie X; Wipf P; Wang QJ A Targeted Library Screen Reveals a New Inhibitor Scaffold for Protein Kinase D. PLoS One 2012, 7 (9), No. e44653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (11).Tandon M;Johnson J; Li Z; Xu S; Wipf P; Wang QJ New Pyrazolopyrimidine Inhibitors of Protein Kinase D as Potent Anticancer Agents for Prostate Cancer Cells. PLoS One 2013, 8 (9), No. e75601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Tandon M; Salamoun JM; Carder EJ; Farber E; Xu S; Deng F; Tang H; Wipf P; Wang QJ SD-208, a Novel Protein Kinase D Inhibitor, Blocks Prostate Cancer Cell Proliferation and Tumor Growth in Vivo by Inducing G2/M Cell Cycle Arrest. PLoS One 2015, 10 (3), No. e0119346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (13).Howard S; Berdini V; Boulstridge JA; Carr MG; Cross DM; Curry J; Devine LA; Early TR; Fazal L; Gill AL; Heathcote M; Maman S; Matthews JE; McMenamin RL; Navarro EF; O’Brien MA; O’Reilly M; Rees DC; Reule M; Tisi D; Williams G; Vinković M; Wyatt PG Fragment-Based Discovery of the Pyrazol-4-yl Urea (AT9283), a Multitargeted Kinase Inhibitor with Potent Aurora Kinase Activity. J. Med. Chem 2009, 52 (2), 379–388. [DOI] [PubMed] [Google Scholar]
- (14).Carling PJ; Ryan BJ; McGuinness W; Kataria S; Humble SW; Milde S; Duce JA; Kapadia N; Zuercher WJ; Davis JB; Di Daniel E; Wade-Martins R Multiparameter Phenotypic Screening for Endogenous TFEB and TFE3 Translocation Identifies Novel Chemical Series Modulating Lysosome Function. Autophagy 2022, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (15).Schirmer A; Kennedy J; Murli S; Reid R; Santi DV Targeted Covalent Inactivation of Protein Kinases by Resorcylic Acid Lactone Polyketides. Proc. Natl. Acad. Sci. U.S.A 2006, 103 (11), 4234–4239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Chambers GE; Sayan AE; Brown RCD The Synthesis of Biologically Active Indolocarbazole Natural Products. Nat. Prod. Rep 2021, 38 (10), 1794–1820. [DOI] [PubMed] [Google Scholar]
- (17).Kobayashi Y; Fujimoto T; Fukuyama T Stereocontrolled Total Synthesis of (+)-K252a. J. Am. Chem. Soc 1999, 121 (27), 6501–6502. [Google Scholar]
- (18).Roy S; Eastman A; Gribble GW Synthesis of N-Alkyl Substituted Bioactive Indolocarbazoles Related to Gö6976. Tetrahedron 2006, 62 (33), 7838–7845. [Google Scholar]
- (19).Gschwendt M; Kittstein W; Johannes F-J Differential Effects of Suramin on Protein Kinase C Isoenzymes. A Novel Tool for Discriminating Protein Kinase C Activities. FEBS Lett. 1998, 421 (2), 165–168. [DOI] [PubMed] [Google Scholar]
- (20).Borges S; Döppler HR; Storz P A Combination Treatment with DNA Methyltransferase Inhibitors and Suramin Decreases Invasiveness of Breast Cancer Cells. Breast Cancer Res. Treat 2014, 144 (1), 79–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Dey D; Ramakumar S; Conn GL Targeted Redesign of Suramin Analogs for Novel Antimicrobial Lead Development. J. Chem. Inf. Model 2021, 61 (9), 4442–4454. [DOI] [PubMed] [Google Scholar]
- (22).Sharlow ER; Giridhar KV; LaValle CR; Chen J; Leimgruber S; Barrett R; Bravo-Altamirano K; Wipf P; Lazo JS; Wang QJ Potent and Selective Disruption of Protein Kinase D Functionality by a Benzoxoloazepinolone. J. Biol. Chem 2008, 283 (48), 33516–33526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Zhu Z; Zhang Y; Wang X; Wang X; Ye S-D Inhibition of Protein Kinase D by CID755673 Promotes Maintenance of the Pluripotency of Embryonic Stem Cells. Development 2020, 147 (16), dev185264. [DOI] [PubMed] [Google Scholar]
- (24).Venardos K; De Jong KA; Elkamie M; Connor T; McGee SL The PKD Inhibitor CID755673 Enhances Cardiac Function in Diabetic db/db Mice. PLoS One 2015, 10 (3), No. e0120934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (25).Higa T; Krubsack AJ Oxidations by Thionyl Chloride. VI. Mechanism of the Reaction with Cinnamic Acids. J. Org. Chem 1975, 40 (21), 3037–3045. [Google Scholar]
- (26).Raynham TM; Hammonds TR; Gilliatt JH; Charles MD; Pave GA; Foxton CH; Carr JL; Mistry NS Aminoethylamino-Aryl (AEAA) Compounds as PKD Inhibitors and Their Preparation, Pharmaceutical Compositions and Use in the Treatment of PKD-Mediated Diseases. WO 2007125331, 2007. [Google Scholar]
- (27).Raynham TM; Hammonds TR; Charles MD; Pave GA; Foxton CH; Blackaby WP; Stevens AP; Ekwuru CT Pyridine Benzamides and Pyrazine Benzamides as PKD Inhibitors and Their Preparation, Pharmaceutical Compositions and Use in the Treatment of Diseases. WO 2008074997, 2008. [Google Scholar]
- (28).Yokoyama S AP2 Inhibitor. WO 2008088006, 2008. [Google Scholar]
- (29).Singh R; Li H; Zhao H; Payan DG; Kolluri R; Tso K; Ramphal J; Gu S Preparation of Cyclic Amine Substituted Pyrimidinediamines as PKC Inhibitors. WO 2009012421, 2009. [Google Scholar]
- (30).Van Eis M; Schuler W; Von Matt A; Soldermann N; Monovich LG; Gaul C Preparation of 2,6-Naphthyridines as AGC Protein Kinase Modulators. WO 2008122614, 2008. [Google Scholar]
- (31).Dobler MR; Jewell CF Jr.; Meredith E; Monovich LG; Siska S; Von Matt A; Van Eis M; Yoon T; Gaul C; Capparelli MP Preparation of [2,6]Naphthyridines as Protein Kinase Inhibitors. WO 2008122615, 2008. [Google Scholar]
- (32).Burgis R; Capparelli MP; Dipietro L; Gamber GG; Jewell CF Jr.; Meredith E; Miranda K; Monovich LG; Rao C; Soldermann N; Yoon T; Zhu Q 2,4’-Bipyridinyl Derivatives as Protein Kinase D Inhibitors and Their Preparation, Pharmaceutical Compositions and Use in the Treatment of Heart Failure and Cancer. WO 2009150230, 2009. [Google Scholar]
- (33).McKinsey TA; Olson E; Vega RB Inhibition of Protein Kinase C-Mu (Protein Kinase D; PKD) as a Treatment for Cardiac Hypertrophy and Heart Failure. WO 2004112763, 2004. [Google Scholar]
- (34).Buehler M; Miraglia L; Monovich LG; Orth A; Tu B New Screening Method and Discovery of Protein Kinase D1 Inhibitors for the Treatment Friedreich’s Ataxia. WO 2015019286, 2015. [Google Scholar]
- (35).Meredith EL; Ardayfio O; Beattie K; Dobler MR; Enyedy I; Gaul C; Hosagrahara V; Jewell C; Koch K; Lee W; Lehmann HJ; McKinsey TA; Miranda K; Pagratis N; Pancost M; Patnaik A; Phan D; Plato C; Qian M; Rajaraman V; Rao C; Rozhitskaya O; Ruppen T; Shi J; Siska SJ; Springer C; van Eis M; Vega RB; von Matt A; Yang L; Yoon T; Zhang J-H; Zhu N; Monovich LG Identification of Orally Available Naphthyridine Protein Kinase D Inhibitors. J. Med. Chem 2010, 53 (15), 5400–5421. [DOI] [PubMed] [Google Scholar]
- (36).Harikumar KB; Kunnumakkara AB; Ochi N; Tong Z; Deorukhkar A; Sung B; Kelland L; Jamieson S; Sutherland R; Raynham T; Charles M; Bagherzadeh A; Foxton C; Boakes A; Farooq M; Maru D; Diagaradjane P; Matsuo Y; Sinnett-Smith J; Gelovani J; Krishnan S; Aggarwal BB; Rozengurt E; Ireson CR; Guha S A Novel Small-Molecule Inhibitor of Protein Kinase D Blocks Pancreatic Cancer Growth in Vitro and in Vivo. Mol. Cancer Ther 2010, 9 (5), 1136–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (37).Sharlow ER; Mustata Wilson G; Close D; Leimgruber S; Tandon M; Reed RB; Shun TY; Wang QJ; Wipf P; Lazo JS Discovery of Diverse Small Molecule Chemotypes with Cell-Based PKD1 Inhibitory Activity. PLoS One 2011, 6 (10), No. e25134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (38).Evans IM; Bagherzadeh A; Charles M; Raynham T; Ireson C; Boakes A; Kelland L; Zachary IC Characterization of the Biological Effects of a Novel Protein Kinase D Inhibitor in Endothelial Cells. Biochem. J 2010, 429 (3), 565–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (39).LogS and TPSA values were calculated with Instant JChem 22.17.1; ChemAxon; http://www.chemaxon.com; downloaded on 11/November/2022. [Google Scholar]
- (40).Guedan A; Swieboda D; Charles M; Toussaint M; Johnston SL; Asfor A; Panjwani A; Tuthill TJ; Danahay H; Raynham T; Mousnier A; Solari R Investigation of the Role of Protein Kinase D in Human Rhinovirus Replication. J. Virol 2017, 91 (9), No. e00217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (41).Feng JA; Lee P; Alaoui MH; Barrett K; Castanedo G; Godemann R; McEwan P; Wang X; Wu P; Zhang Y; Harris SF; Staben ST Structure Based Design of Potent Selective Inhibitors of Protein Kinase D1 (PKD1). ACS Med. Chem. Lett 2019, 10 (9), 1260–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (42).Trejo A; Arzeno H; Browner M; Chanda S; Cheng S; Comer DD; Dalrymple SA; Dunten P; Lafargue J; Lovejoy B; Freire-Moar J; Lim J; McIntosh J; Miller J; Papp E; Reuter D; Roberts R; Sanpablo F; Saunders J; Song K; Villasenor A; Warren SD; Welch M; Weller P; Whiteley PE; Zeng L; Goldstein DM Design and Synthesis of 4-Azaindoles as Inhibitors of p38 MAP Kinase. J. Med. Chem 2003, 46 (22), 4702–4713. [DOI] [PubMed] [Google Scholar]
- (43).Verschueren K; Cobbaut M; Demaerel J; Saadah L; Voet ARD; Van Lint J; De Borggraeve WM Discovery of a Potent Protein Kinase D Inhibitor: Insights in the Binding Mode of Pyrazolo[3,4-d]Pyrimidine Analogues. MedChemComm 2017, 8 (3), 640–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (44).Gellibert F; Fouchet MH; Nguyen VL; Wang R; Krysa G; de Gouville AC; Huet S; Dodic N Design of Novel Quinazoline Derivatives and Related Analogues as Potent and Selective ALK5 Inhibitors. Bioorg. Med. Chem. Lett 2009, 19 (8), 2277–2281. [DOI] [PubMed] [Google Scholar]
- (45).Gehringer M; Laufer SA Emerging and Re-Emerging Warheads for Targeted Covalent Inhibitors: Applications in Medicinal Chemistry and Chemical Biology. J. Med. Chem 2019, 62 (12), 5673–5724. [DOI] [PubMed] [Google Scholar]
- (46).Fukazawa H; Ikeda Y; Fukuyama M; Suzuki T; Hori H; Okuda T; Uehara Y The Resorcylic Acid Lactone Hypothemycin Selectively Inhibits the Mitogen-Activated Protein Kinase Kinase-Extracellular Signal-Regulated Kinase Pathway in Cells. Biol. Pharm. Bull 2010, 33 (2), 168–173. [DOI] [PubMed] [Google Scholar]
- (47).Zhao YS; Xu Y; Wang K; Zeng H; Wang J; Mao SC; Zhang JH Homology Modeling and Molecular Dynamics Study of C-Terminal Catalytic Domain of Human Protein Kinase D1. Asian J. Chem 2013, 25 (3), 1259–1264. [Google Scholar]
- (48).Nalini; Chadha N; Bahia MS; Kaur M; Bahadur R; Silakari O Computational Design of New Protein Kinase D 1 (Pkd1) Inhibitors: Homology-Based Active Site Prediction, Energy-Optimized Pharmacophore, Docking and Database Screening. Mol. Diversity 2018, 22 (1), 47–56. [DOI] [PubMed] [Google Scholar]
- (49).Seeliger D; de Groot BL Ligand Docking and Binding Site Analysis with Pymol and Autodock/Vina. J. Comput.-Aided Mol. Des 2010, 24 (5), 417–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (50).Lv D; Chen H; Feng Y; Cui B; Kang Y; Zhang P; Luo M; Chen J Small-Molecule Inhibitor Targeting Protein Kinase D: A Potential Therapeutic Strategy. Front. Oncol 2021, 11, 680221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (51).Vega RB; Harrison BC; Meadows E; Roberts CR; Papst PJ; Olson EN; McKinsey TA Protein Kinases C and D Mediate Agonist-Dependent Cardiac Hypertrophy Through Nuclear Export of Histone Deacetylase 5. Mol. Cell. Biol 2004, 24 (19), 8374–8385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (52).Fielitz J; Kim MS; Shelton JM; Qi X; Hill JA; Richardson JA; Bassel-Duby R; Olson EN Requirement of Protein Kinase D1 for Pathological Cardiac Remodeling. Proc. Natl. Acad. Sci. U.S.A 2008, 105 (8), 3059–3063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (53).Harrison BC; Kim MS; van Rooij E; Plato CF; Papst PJ; Vega RB; McAnally JA; Richardson JA; Bassel-Duby R; Olson EN; McKinsey TA Regulation of Cardiac Stress Signaling by Protein Kinase D1. Mol. Cell. Biol 2006, 26 (10), 3875–3888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (54).Kolczynska K; Loza-Valdes A; Hawro I; Sumara G Diacylglycerol-Evoked Activation of PKC and PKD Isoforms in Regulation of Glucose and Lipid Metabolism: A Review. Lipids Health Dis. 2020, 19 (1), 113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (55).Trujillo-Viera J; El-Merahbi R; Schmidt V; Karwen T; Loza-Valdes A; Strohmeyer A; Reuter S; Noh M; Wit M; Hawro I; Mocek S; Fey C; Mayer AE; Löffler MC; Wilhelmi I; Metzger M; Ishikawa E; Yamasaki S; Rau M; Geier A; Hankir M; Seyfried F; Klingenspor M; Sumara G Protein Kinase D2 Drives Chylomicron-Mediated Lipid Transport in the Intestine and Promotes Obesity. EMBO Mol. Med 2021, 13 (5), No. e13548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (56).Lieb WS; Lungu C; Tamas R; Berreth H; Rathert P; Storz P; Olayioye MA; Hausser A The GEF-H1/PKD3 Signaling Pathway Promotes the Maintenance of Triple-Negative Breast Cancer Stem Cells. Int. J. Cancer 2020, 146 (12), 3423–3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (57).Janardhanan P; Somasundaran AK; Balakrishnan AJ; Pilankatta R Sensitization of Cancer Cells Towards Cisplatin and Carboplatin by Protein Kinase D Inhibitors through Modulation of ATP7A/B (Copper Transport ATPases). Cancer Treat. Res. Commun 2022, 32, 100613. [DOI] [PubMed] [Google Scholar]
- (58).Karaman MW; Herrgard S; Treiber DK; Gallant P; Atteridge CE; Campbell BT; Chan KW; Ciceri P; Davis MI; Edeen PT; Faraoni R; Floyd M; Hunt JP; Lockhart DJ; Milanov ZV; Morrison MJ; Pallares G; Patel HK; Pritchard S; Wodicka LM; Zarrinkar PP A Quantitative Analysis of Kinase Inhibitor Selectivity. Nat. Biotechnol 2008, 26 (1), 127–132. [DOI] [PubMed] [Google Scholar]
- (59).Chen J; Lu GW; Wang QJ Protein Kinase C-Independent Effects of Protein Kinase D3 in Glucose Transport in L6Myotubes. Mol. Pharmacol 2005, 67 (1), 152–162. [DOI] [PubMed] [Google Scholar]
- (60).LaValle CR; Bravo-Altamirano K; Giridhar KV; Chen J; Sharlow E; Lazo JS; Wipf P; Wang QJ Novel Protein Kinase D Inhibitors Cause Potent Arrest in Prostate Cancer Cell Growth and Motility. BMC Chem. Biol 2010, 10, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (61).Meredith EL; Beattie K; Burgis R; Capparelli M; Chapo J; Di Pietro L; Gamber G; Enyedy I; Hood DB; Hosagrahara V; Jewell C; Koch KA; Lee W; Lemon DD; McKinsey TA; Miranda K; Pagratis N; Phan D; Plato C; Rao C; Rozhitskaya O; Soldermann N; Springer C; van Eis M; Vega RB; Yan W; Zhu Q; Monovich LG Identification of Potent and Selective Amidobipyridyl Inhibitors of Protein Kinase D. J. Med. Chem 2010, 53 (15), 5422–5438. [DOI] [PubMed] [Google Scholar]
- (62).Gilles P; Kashyap RS; Freitas MJ; Ceusters S; Van Asch K; Janssens A; De Jonghe S; Persoons L; Cobbaut M; Daelemans D; Van Lint J; Voet ARD; De Borggraeve WM Design, Synthesis and Biological Evaluation of Pyrazolo[3,4-d]Pyrimidine-Based Protein Kinase D Inhibitors. Eur. J. Med. Chem 2020, 205, 112638. [DOI] [PubMed] [Google Scholar]
- (63).Bain J; Plater L; Elliott M; Shpiro N; Hastie CJ; McLauchlan H; Klevernic I; Arthur JSC; Alessi DR; Cohen P The Selectivity of Protein Kinase Inhibitors: A Further Update. Biochem. J 2007, 408 (3), 297–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (64).Chung R; Foster BK; Xian CJ Inhibition of Protein Kinase-D Promotes Cartilage Repair at Injured Growth Plate in Rats. Injury 2013, 44 (7), 914–922. [DOI] [PubMed] [Google Scholar]
- (65).Duan GJ; Zhu J; Xu CY; Wan JY; Zhang L; Ge XD; Liu LM; Liu YS Protective Effect of Go6976, a PKD Inhibitor, on LPS/D: -GalN-Induced Acute Liver Injury in Mice. Inflamm. Res 2011, 60 (4), 357–366. [DOI] [PubMed] [Google Scholar]
- (66).Upadhyay K; Park JE; Yoon TW; Halder P; Kim YI; Metcalfe V; Talati AJ; English BK; Yi AK Group B Streptococci Induce Proinflammatory Responses via a Protein Kinase D1-Dependent Pathway. J. Immunol 2017, 198 (11), 4448–4457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (67).Yuan J; Liu Y; Tan T; Guha S; Gukovsky I; Gukovskaya A; Pandol SJ Protein Kinase D Regulates Cell Death Pathways in Experimental Pancreatitis. Front. Physiol 2012, 3, 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (68).Yuan J; Tan T; Geng M; Tan G; Chheda C; Pandol SJ Novel Small Molecule Inhibitors of Protein Kinase D Suppress NF-kappaB Activation and Attenuate the Severity of Rat Cerulein Pancreatitis. Front. Physiol 2017, 8, 1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (69).Yuan J; Chheda C; Tan G; Elmadbouh O; Pandol SJ Protein Kinase D: A Therapeutic Target in Experimental Alcoholic Pancreatitis. Biochim. Biophys. Acta Mol. Basis Dis 2022, 1868 (11), 166486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (70).Pasquier A; Vivot K; Erbs E; Spiegelhalter C; Zhang Z; Aubert V; Liu Z; Senkara M; Maillard E; Pinget M; Kerr-Conte J; Pattou F; Marciniak G; Ganzhorn A; Ronchi P; Schieber NL; Schwab Y; Saftig P; Goginashvili A; Ricci R Lysosomal Degradation of Newly Formed Insulin Granules Contributes to B Cell Failure in Diabetes. Nat. Commun 2019, 10 (1), 3312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (71).Venardos K; De Jong KA; Elkamie M; Connor T; McGee SL The PKD Inhibitor CID755673 Enhances Cardiac Function in Diabetic db/db Mice. PLoS One 2015, 10 (3), No. e0120934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (72).Yang L; Liu N; Zhao W; Li X; Han L; Zhang Z; Wang Y; Mao B Angiogenic Function of Astragaloside IV in Rats with Myocardial Infarction Occurs via the PKD1-HDAC5-VEGF Pathway. Exp. Ther. Med 2019, 17 (4), 2511–2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (73).Wang W; Duclot F; Groveman BR; Carrier N; Qiao H; Fang X-Q; Wang H; Xin W; Jiang X-H; Salter MW; Ding X-S; Kabbaj M; Yu X-M Hippocampal Protein Kinase D1 Is Necessary for DHPG-Induced Learning and Memory Impairments in Rats. PLoS One 2018. 13 (4), No. e0195095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (74).Michaud D; Mirlekar B; Steward C; Bishop G; Pylayeva-Gupta YB Cell Receptor Signaling and Protein Kinase D2 Support Regulatory B Cell Function in Pancreatic Cancer. Front. Immun 2022, 12, 745873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (75).Wei N; Chu E; Wipf P; Schmitz JC Protein Kinase D as a Potential Chemotherapeutic Target for Colorectal Cancer. Mol. Cancer Ther 2014, 13 (5), 1130–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (76).Borges S; Perez EA; Thompson EA; Radisky DC; Geiger XJ; Storz P Effective Targeting of Estrogen Receptor Negative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101. Mol. Cancer Ther 2015, 14 (6), 1306–1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (77).Kim DY; Park EY; Chang E; Kang H-G; Koo Y; Lee EJ; Ko JY; Kong HK; Chun K-H; Park JH A Novel miR-34a Target, Protein Kinase D1, Stimulates Cancer Stemness and Drug Resistance through GSK3/β-Catenin Signaling in Breast Cancer. Oncotarget 2016, 7 (12), 14791–14802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (78).Liu Y; Wang Y; Yu S; Zhou Y; Ma X; Su Q; An L; Wang F; Shi A; Zhang J; Chen L The Role and Mechanism of CRT0066101 as an Effective Drug for Treatment of Triple-Negative Breast Cancer. Cell. Physiol. Biochem 2019, 52 (3), 382–396. [DOI] [PubMed] [Google Scholar]
- (79).Liu Y; Zhou Y; Ma X; Chen L Inhibition Lysosomal Degradation of Clusterin by Protein Kinase D3 Promotes Triple-Negative Breast Cancer Tumor Growth. Adv. Sci 2021, 8 (4), 2003205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (80).Li QQ; Hsu I; Sanford T; Railkar R; Balaji N; Sourbier C; Vocke C; Balaji KC; Agarwal PK Protein Kinase D Inhibitor CRT0066101 Suppresses Bladder Cancer Growth in Vitro and Xenografts via Blockade of the Cell Cycle at G2/M. Cell. Mol. Life Sci 2018, 75 (5), 939–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (81).Zhu Y; Cheng Y; Guo Y; Chen J; Chen F; Luo R; Li A Protein Kinase D2 Contributes to TNF-alpha-Induced Epithelial Mesenchymal Transition and Invasion via the PI3K/GSK-3beta/beta-Catenin Pathway in Hepatocellular Carcinoma. Oncotarget 2016, 7 (5), 5327–5341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (82).Miyamoto K; Hayabuchi H; Tokifuji Y; Ando M; Onishi N; Okamura T; Yoshimura A; Chikuma S A Protein Kinase D Inhibitor Suppresses AKT on T cells and Antagonizes Cancer Immunotherapy by Anti-PD-1. Int. Immunol 2022, dxac035. [DOI] [PubMed] [Google Scholar]
- (83).Liu Z; Wang Y; Pan S; Zhang Y; Zhu H; Liu Y; Zhu L; Zhang J Platelets-Released Insulin-Like Growth Factor 1 is Correlated with Anxiety in Myocardial Infarction. Biochem. Biophys. Res. Commun 2019, 520 (2), 441–448. [DOI] [PubMed] [Google Scholar]
- (84).Haworth RS; Cuello F; Herron TJ; Franzen G; Kentish JC; Gautel M; Avkiran M Protein Kinase D Is a Novel Mediator of Cardiac Troponin I Phosphorylation and Regulates Myofilament Function. Circ. Res 2004, 95 (11), 1091–1099. [DOI] [PubMed] [Google Scholar]
- (85).Shapiro BA; Olala L; Arun SN; Parker PM; George MV; Bollag WB Angiotensin II-Activated Protein Kinase D Mediates Acute Aldosterone Secretion. Mol. Cell. Endocrinol 2010, 317 (1), 99–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (86).Abedi H; Rozengurt E; Zachary I Rapid Activation of the Novel Serine/Threonine Protein Kinase, Protein Kinase D by Phorbol Esters, Angiotensin II and PDGF-BB in Vascular Smooth Muscle Cells. FEBS Lett. 1998, 427 (2), 209–212. [DOI] [PubMed] [Google Scholar]