Samson A.
Souza
a,
Abby
Held
b,
Wenjie J.
Lu
c,
Brendan
Drouhard
a,
Bryant
Avila
a,
Raul
Leyva-Montes
a,
Michelle
Hu
c,
Bill R.
Miller
3rd
b and
Ho Leung
Ng
*a
aDepartment of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA. E-mail: hng@ksu.edu
bDepartment of Chemistry, Truman State University, Kirksville, MO, USA
cDepartment of Chemistry, University of Hawai‘i at Mānoa, Honolulu, HI, USA
First published on 19th March 2021
Aromatase (CYP19) catalyzes the last biosynthetic step of estrogens in mammals and is a primary drug target for hormone-related breast cancer. However, treatment with aromatase inhibitors is often associated with adverse effects and drug resistance. In this study, we used virtual screening targeting a predicted cytochrome P450 reductase binding site on aromatase to discover four novel non-steroidal aromatase inhibitors. The inhibitors have potencies comparable to the noncompetitive tamoxifen metabolite, endoxifen. Our two most potent inhibitors, AR11 and AR13, exhibit both mixed-type and competitive-type inhibition. The cytochrome P450 reductase-CYP19 coupling interface likely acts as a transient binding site. Our modeling shows that our inhibitors bind better at different sites near the catalytic site. Our results predict the location of multiple ligand binding sites on aromatase. The combination of modeling and experimental results supports the important role of the reductase binding interface as a low affinity, promiscuous ligand binding site. Our new inhibitors may be useful as alternative chemical scaffolds that may show different adverse effects profiles than current clinically used aromatase inhibitors.
AIs in clinical use include the non-steroidal AIs (NSAIs), anastrozole and letrozole, and the steroidal AI, exemestane, which interacts covalently at the active site.4,5 The most potent NSAI, letrozole, binds a distinct site exhibiting both noncompetitive and mixed-mode type Michaelis inhibition.6 This type of behavior is also observed for the noncompetitive AI endoxifen (Ki = 4 μM), a potent metabolite of tamoxifen, a clinically-used estrogen receptor α (ERα) antagonist.7 Metabolic N-demethylation of endoxifen produces a competitive AI, norendoxifen (Ki = 35 nM).8 This demonstrates two important points. First, minor chemical modifications of inhibitors (letrozole to anastrozole, and endoxifen to norendoxifen) can change the inhibitory mechanism in unexpected ways. Second, CYP19 can be allosterically modulated with high potency. The mechanism of mixed mode inhibition for CYP19 and other cytochrome P450 (CYP450) enzymes is unclear.
Steroidal AIs interact at the active site, also known as the distal heme site, with high efficacy. However, steroidal analogs frequently exert similar adverse effects. CYP19 inhibition at an alternative allosteric site provides opportunities for the discovery of novel NSAIs chemically distinct from current therapeutics and with different toxicity profiles.
In this work, we investigate the binding of small molecule inhibitors to the CYP19 heme distal site, the heme proximal site (the predicted site of CPR binding), and the substrate access channel through a combination of experimental and computational approaches. These sites have been proposed by other investigators as potential therapeutic targets.9,10 We first predict and model the interfacial contacts of cytochrome P450 reductase (CPR) with CYP19. We performed virtual screening against a library of over a million compounds to identify potential inhibitors. We use molecular dynamics simulations to model how they interact at the CPR–CYP19 interface. We then provide experimental enzyme inhibition data for four new NSAIs we discovered, AR11, AR13, AR19, and AR20. In addition, we use optical absorption spectra to characterize the effects of inhibition on the heme chemical environment. We characterize the inhibition modes of AR11, AR13, AR19, and AR20, and their likeliest binding sites closer to the catalytic site. The cytochrome P450 reductase binding site is likely a transient, low-affinity binding site for multiple ligands.
Enzyme activity was measured by monitoring the conversion of exogenous substrate 7-methoxy-4-(trifluoromethyl)coumarin (MFC) to its fluorescent product 7-hydroxy-4-(trifluoromethyl)coumarin (HFC). A CYP19/MFC high-throughput screening kit (Corning) was used to measure inhibition of aromatase activity for compounds AR11 and AR13. Briefly, 2× NADPH regeneration system (16.25 μM NADP+, 825 μM MgCl2, 825 μM glucose-6-phosphate, 0.4 U mL−1 G6P dehydrogenase) was prewarmed with inhibitors (1:2 serial dilutions) at 37 °C for 10 minutes in black 96-well plates. Reactions were initiated with prewarmed 2× enzyme–substrate mix (15 nM P450 microsomes enriched with oxidoreductase, 50 μM MFC) and incubated at 37 °C for 30 minutes. The 200 μL reaction mixtures were terminated with 75 μL 0.5 M Tris-base (80% ACN). A FluoDia T70 plate reader measured HFC product formation with excitation/emission filters of 400/530 nm. Reactions were performed in duplicate then repeated twice more on separate days. Assays were performed in-parallel with ketoconazole and endoxifen since the inhibitory activities are documented at the experimental conditions we detail here.7,40
The reaction conditions were repeated for measuring the inhibitory activity of compounds AR19 and AR20. Modifications are highlighted here. Supersomes containing CYP19 + CPR (Corning), NADPH regeneration system (Corning), MFC (Chemodex), and ketoconazole (Selleck Chemicals) were used for the reaction mixture. Temperature-controlled incubations of the 96-well plate were carried out with a dry-plate. A Tecan fluorescent plate reader measured the fluorescent product in the circle-read mode at the optimum gain with excitation/emission filters of 405/535 nm. Reactions were measured in duplicate.
Reaction blanks were used for data corrections and these results were normalized to the fluorescence response in the absence of inhibitor. Analyses were performed in GraphPad Prism 9 software, and data were fit to a 4-parameter (variable slope) logistic model on semilog axes.
Purified CYP19 was diluted with 100 mM potassium phosphate buffer (pH 7.4) to 50 μL to a final concentration of 2–3 μM P450. Inhibitors were titrated such that the endpoint would not exceed 3% ACN. Absorption scans were read with a single-beam Agilent 8453 UV-Vis spectrophotometer after resuspension and a 10 minutes incubation period at 25 °C. Buffer A components, endoxifen, AR11, AR13, AR19, and AR20 did not contribute to hyperchromic shifts in the Soret peak region.
5 × 4 Lineweaver–Burk and Dixon plots were used to diagnose AR11 and AR13 inhibitory modes. Independent and dependent variables were precalculated and entered into GraphPad Prism 9 software for linear regression analyses. Nonlinear regression curves were fit to mixed- or competitive-type Michaelis functions to return kinetic constants defined by R. A. Copeland.41
Only two of the sites were predicted by the protein–protein interface algorithm, CPORT,16 to actively participate in protein–protein interactions (sites 2 and 4 in Fig. 1). They correspond to the N-terminal region (αA′ and β1–2) and the proximal heme site. The likeliest CPR binding surface was selected based on three criteria. Firstly, the proximity of the N5 atom of FMN to the iron center of the heme group should be within a distance that is physiologically sound.44 Secondly, the orientation of the N-terminus of the reductase and CYP19 should be positioned in the same direction since they are truncations of transmembrane segments tethered to the cytoplasmic face of the endoplasmic reticulum.45 Lastly, only the highest-scoring clusters from docking aromatase with CPR with Haddock15 were assessed. Only site 4, corresponding to the proximal heme region, fulfilled all three criteria.
The probable binding mode contrasts with that of the crystal structure of B. megaterium P450BM3 fusion (PDB 1BVY). The N5 to iron distance in our structure is 14.4 Å, whereas the distance is 22.7 Å in P450BM3. This is within the 14–15 Å threshold limit for electron transfer in most physiological processes.46 The possibility of through-bond tunneling at much longer distances in P450 BM3 was previously refuted due to faster experimental kinetic rates than predicted from theoretical models.44 Physicochemical descriptions of the modeled and fusion P450–CPR complexes are compared in Table S2 (ESI†). In docking CYP19 against CPR in a closed conformation (PDB 3QE2), the closest N5 to iron distance was 34.9 Å. The negative electrostatic potential of the FMN domain interacts with the positive potential of the FAD/NADPH domains. In this conformation, FADH2 is in a closer proximity to reduce the FMN cofactor. In the proposed end-on interaction, 3FJO adopts an extended conformation to expose the buried FMN-binding interface for the reduction of CYP19 at the proximal heme site (Fig. 2 and Fig. S3, ESI†).
Fig. 2 Electrostatic potential surface map of the proposed end-on binding mode of CYP19 (PDB 4KQ8) in complex with CPR (PDB 3FJO) in an open conformation. Negatively charged potentials (red) to positive potentials (blue) are represented as a color gradient with neutral (gray) regions. |
At the CYP19–CPR interface there are 17 total polar–polar, polar–charged, and charged–charged bond pairs. The most important of these contacts involve sidechains from the CYP19 residues K108, Y424, K440, and Y441 (Fig. 3 and Table S2, ESI†). Salt bridges between the proximal heme lysines K440/K108 and the reductase acidic residues D125/E73 are projected to be major contributors to enyme-coupling.
These compounds showed IC50 values <75 μM. AR13 showed 3-fold increased potency over the control inhibitor, ketoconazole, an antifungal with IC50 = 3.08 μM. The IC50 value we report for AR19, 72 μM, was fit to a Hill coefficient of 1 with a projected 11% activity at saturation. The Sy.x value at 0.073 is suboptimal relative to the other inhibitors due to its poor solubility in buffer to interpolate a sigmoidal regression. There was no evidence of an inadequate model returned from a replicates test (Table S4, ESI†). AR11 and AR20 exhibited potencies comparable to an active tamoxifen metabolite, endoxifen. Dose–response curves and activity data are presented in Fig. 5 and Table 1. Values with 95% confidence intervals are provided in Table S4 (ESI†).
Inhibitor | IC50 (μM) | Hill slope | Fractional activity at saturation | Goodness of fit (Sy.x) | Sy.x3p/Sy.x4p |
---|---|---|---|---|---|
a 3-Parameter model reported with a Hill coefficient of 1. | |||||
Ketoconazole | 3.1 ± 0.2 | 1.12 ± 0.07 | 0.03 ± 0.01 | 0.02 | 1.059 |
Endoxifen | 30.9 ± 3.1 | 2.27 ± 0.41 | 0.02 ± 0.02 | 0.06 | 1.596 |
AR11 | 31.1 ± 5.4 | 1.54 ± 0.33 | 0.08 ± 0.02 | 0.06 | 1.103 |
AR13 | 0.8 ± 0.2 | 0.86 ± 0.15 | 0.07 ± 0.06 | 0.06 | 0.998 |
AR19a | 72.9 ± 66.6 | 1 | 0.11 ± 0.42 | 0.07 | 0.976 |
AR20 | 42.8 ± 22.2 | 0.86 ± 0.15 | 0.03 ± 0.01 | 0.02 | 0.994 |
A Hill slope of 2 for AR11 and endoxifen indicates that multiple binding sites are probable at saturation. The ratio of Sy.x values of the 3-parameter to the variable slope functions are 1.596 and 1.103, respectively. This indicates that the error of the residuals is reduced when the data is fit to a 4-parameter function where the Hill slopes are 2-fold greater.
Titration of up to 100 μM of endoxifen did not induce a Soret peak shift from 395 nm in the CYP19 ASD-bound state (Fig. 7a). This indicates that ASD remains in the active site in the presence of a 50-fold molar excess of endoxifen. This behavior is expected of a noncompetitive inhibitor where the Ki is unchanged in the presence of substrate. At a 3-fold (6 μM) molar excess, AR11 prompted the appearance of a peak shoulder near 416 nm. Fig. 7b illustrates the gradual increase of the peak shoulder at 416 nm with an increase in the concentration of inhibitor. This indicates that AR11 causes the enzyme to favor a shift to the 6-coordinate low-spin state and the displacement of ASD in the active site. At 100 μM AR11, the inhibitor-bound 6-coordinate low-spin state is apparent at 416 nm (Fig. 7d). The inset in Fig. 7d illustrates that AR11 causes CYP19 to favor the low-spin state in the absence of its native substrate. The Soret peak is red shifted 2 nm to 418 nm. This is likely a symptom of greater orbital overlap between the water ligand and iron induced by a readjustment of the active site.
AR19 and AR20 share the same Soret peak trends in the presence and absence of 2 μM ASD. Both compounds induce a gradual shift to the iron low-spin state indicated by simultaneous hyperchromic and red shifting towards 420 nm. This indicates that the population of CYP19 with iron in the 6-coordinate state increases in the presence of inhibitor. The red shift from 416 nm (blue trace) to 420 nm (red trace) is apparent in Fig. 8c and d. At 100 μM AR19, 2 μM ASD introduces a faint peak shoulder at 395 nm. Although there is a drop in the absorption at 420 nm, a peak shoulder is not apparent at this concentration. Higher concentrations of AR19 are required to induce the same Soret peak effects as AR20, suggesting that AR20 is a more potent inhibitor.
Lineweaver–Burk plots at various concentrations of AR11 yielded functions that intersected the y-axis at different inhibitor concentrations (Fig. 10a). These corresponded to different apparent Vmax values, suggesting that it does not act competitively. In Fig. 10b, curves that correspond to AR13 concentrations 0–0.05 μM nearly intersect the y-axis at the same point. However, at 0.1 μM, this convergence is not apparent.
Fig. 9 Non-linear regression curves for AR11 (a) and AR13 (b) fit to single-site mixed and competitive-type Michaelis–Menten functions. |
Fig. 10 Lineweaver–Burk plots at 10 nM P450 and various concentrations of substrate MFC with inhibitors AR11 (a) and AR13 (b). |
In Dixon-type analyses, the intersection of regression curves in Fig. 11 panels A and B correspond to the Ki values for AR11 and AR13, respectively. The intersection of the curves in panels C and D correspond to a Ki′ or inhibitor dissociation constant in the presence of substrate MFC. This value infers non-mutual exclusive binding. Upon visual inspection of panel A, AR11 behaves noncompetitively at low MFC concentrations but exhibits competitive or mixed-type behavior as curves intersect above the x-axis at higher concentrations. Inclusive of an apparent Vmax decrease (Fig. 10a) and a Soret peak shift to 418 nm at a 3:1 androstenedione to inhibitor ratio, AR11 behaves as a mixed-type inhibitor. A hallmark of this inhibition type is the intersection of curves below the x axis that would be extrapolated from Fig. 11c. An Akaike's model favored a nonlinear regression fit to a mixed model over a noncompetitive model. The ratio of these probabilities was 126. As such, AR11 was fit to a mixed-type nonlinear function.
AR13 exhibits similar trends as AR11 in a reciprocal velocity by inhibitor concentration plot (Fig. 11b). On the contrary, near-parallel slopes in panel d suggests an indeterminate Ki′ value whereby MFC and AR13 are mutually exclusive. This behavior is demonstrated at substrate concentrations near or greater than the MFC Km of 28 μM. Data at 9.9 μM MFC perplexes this interpretation since at a low concentration (9.9 μM), AR13 appears to act noncompetitively. Isolated noncompetitive behavior at low substrate concentrations has been reported with the inhibitor-pesticide glyphosate while demonstrating mixed-mode inhibition at higher substrate concentrations.49 Here, the difference between a hybrid-type mechanism and a single-type mechanism is more transparent. Competitive inhibitors should not preferentially interact at a distinct site at low substrate concentrations. Rather, the inverse may hold true. Distinguishing the difference between a noncompetitive and competitive inhibitor from linear transformations50 is often unclear and has been documented in Dixon plots.51 In an Akaike's test, a competitive model was favored over a noncompetitive fit with a 138 ratio of probabilities. Additionally, competitive-type behavior was apparent at a 1:2 enzyme:inhibitor ratio. Experiments at the lowest inhibitor concentration were carried out at a comparable ratio.
AR11 and AR13 were fit to nonlinear functions defined by mixed- and competitive-type kinetics, correspondingly. The Ki of AR13 is 42 nM, 39–57 nM for a 95% confidence interval. This is within an order of magnitude from the working enzyme concentration and may be an indicator of partial tight-binding behavior. In a Lineweaver–Burk plot, the 1/V0 values of tight-binders converge at high substrate concentrations and curvature is most recognizable at high inhibitor concentrations.50 This may provide a rationale for a distinct Vmax (y intercept) at 0.1 μM AR13 in Fig. 10b by which the data deviates from linearity.
Table 2 presents the effects of each inhibitor on these kinetic constants. The apparent Km increases in the presence of inhibitor, indicating that more substrate MFC is needed to achieve 1/2Vmax. Meanwhile, only AR11 decreased the maximum velocity, indicating that MFC saturation will not retain full enzyme activity. Therefore, both substrate and AR11 bind CYP19 at discrete sites. In contrast, the ability to achieve the Vmax with a molar excess of MFC over AR13 indicates that both compounds bind the active site. In Table 2, we report the AR11 Ki′ as a product of the means of Ki and α. Propagation of the standard error of the means yields Ki′ values between 0.6 and 1.8-fold greater. Kinetic constants with 95% confidence intervals are supplemented (Table S5, ESI†).
V max,app (pmol HFC min−1 pmol−1 P450) | K m,app (μM) | R 2 | K i (μM) | α | K i′ (μM) | |
---|---|---|---|---|---|---|
[AR11] (μM) | 12.8 ± 1.8 | 5.0 ± 2.9 | 64.6 | |||
0 | 0.73 ± 0.03 | 29.4 ± 2.8 | 0.994 | |||
5 | 0.68 | 38.0 | 0.993 | |||
10 | 0.63 | 45.4 | 0.983 | |||
20 | 0.56 | 57.6 | 0.989 | |||
40 | 0.45 | 75.0 | 0.937 | |||
[AR13] (nM) | 42 ± 2 nM | — | — | |||
0 | 0.73 ± 0.02 | 28.42 ± 1.62 | 0.995 | |||
0.0125 | — | 36.78 | 0.994 | |||
0.025 | — | 45.14 | 0.993 | |||
0.05 | — | 61.86 | 0.996 | |||
0.1 | — | 95.31 | 0.982 |
Ligand | Proximal heme site average dissociation time (ns) | Proximal heme site average binding free energy (kcal mol−1) | Substrate access average binding free energy (kcal mol−1) | Active site average binding free energy (kcal mol−1) |
---|---|---|---|---|
a Denotes a value from 1 replicate run. b Denotes a value from 2 replicate runs. NR and a dash (—) indicate unreported and unmeasured quantities, respectively. | ||||
E-end | <200 | −17 ± 4 | — | — |
Z-end | <500 | −24 ± 3 | — | — |
AR11 | >1000 | −32 ± 4 | −29 ± 8 | — |
AR13 (1R,2S) | <100 | NR | — | −36 ± 4 |
AR13 (1S,2R) | — | — | — | −36 ± 2 |
AR19 | 400b | NR | −33 ± 3 | — |
AR20 | 800a | −22 ± 0a | — | — |
Since the Soret shifts supported an iron–imidazole interaction, AR13 was docked to the catalytic site in 25 independent runs. The binding mode of each enantiomer yielding the closest iron–imidazole distance was used for MD simulations. They bound with free energies of −36 ± 4 kcal mol−1 (1R,2S) and −36 ± 2 kcal mol−1 (1S,2R) as calculated by MM-PBSA. Both enantiomers are projected to form a π–π interaction with F221 in the substrate access channel and a low energy π–π interaction between the terminal azole and the heme's porphyrin system. The Fe–N distances for each compound averaged 6 Å, indicating an indirect heme interaction.
More rigorous global redocking of AR11 located it in the substrate access channel more often than in the proximal heme site. AR11 formed π–π interactions with F221 and W224 and a hydrogen bond between the triazole and D309. This suggests that the triazole partially obstructs a space occupied by ASD since D309 is involved in a critical contact with the C3–carbonyl of androgens. Production runs yielded an average free energy of −29 ± 8 kcal mol−1. In the proximal heme site, the free energy was −32 ± 4 kcal mol−1, despite a destabilizing effect from E357. We report the putative binding modes at both the proximal heme site and substrate access channel in Fig. 12 since their binding energies have overlapping confidence intervals. We include the decomposition scores of the interactions with the greatest contributions to these energies.
Fig. 12 3-D and 2-D interaction diagrams for AR11 with decomposition scores of the projected protein–ligand interactions at the substrate access channel (a) and the proximal heme site (b). 2-D depictions were generated with PoseView software.52 Az-azole, Ph-phenyl, Ar-aromatic groups. |
AR19 and AR20 dock to the proximal heme site but dissociate after developing unfavorable interactions with E357, as demonstrated by positive decomposition scores (Fig. S4 and S5, ESI†). We also note that control inhibitors, E- and Z-endoxifen, disassociate due to positive free energy contributions from E357. In 1 out of 3 simulations, AR20 re-associates and remains in the site >1 μs with −22 kcal mol−1 average binding free energy (Fig. S6, ESI†). We do not report decomposition scores for AR20 because it only transiently remains in a single binding mode throughout the production run. AR19 formed stable interactions in the substrate access channel with F221 and D309. Further, it had a proclivity to migrate closer into the active site for the duration of the production runs (Fig. 13a). The average free energy was −33 kcal mol−1.
Fig. 13 3-D and 2-D interaction diagrams with decomposition scores of the projected protein–ligand interactions for AR19 (a) and AR20 (b). 2-D depictions were generated with PoseView software.52 AR20 binding mode represented here is for replicate 2, and >280 ns. Decomposition scores are not available for AR20. Az-azole, Ph-phenyl, Ar-aromatic sidechain. |
CYP19's ability to bind ASD and CO in the presence of AR13 demonstrates its capacity to maintain its functionality. Above all, the need for a 28-fold molar excess of ASD to induce a blue shift to 395 nm supports the inhibitor's high potency. Among all our inhibitors, AR13 was the most potent. This correlates with its predicted average binding energy being the greatest over AR11, AR19, AR20, and endoxifen. AR13 has a 40-fold lower IC50 value than that of endoxifen and interacts with CYP19 on the same order of magnitude as norendoxifen (Ki = 35 nM). A major contributor to its potency could be through stabilizing a rigid protein-monomer. Substrate- and inhibitor-bound CYP19 complexes reduce the enzyme's intrinsic flexibility.9,55 Additionally, AR13 interaction at the active site may favor disassociation of the reductase. We deduce this from a recent study whereby androstenedione reduced the reductase binding affinity by 5-fold.56 We project that AR13 may be useful as a scaffold to design new NSAIs due to its remarkably distinct structure from current third generation NSAIs.
Titration with AR11 against androstenedione induces a peak shoulder between 414–416 nm, indicating that AR11 displaces ASD while substrate access to the heme iron is maintained. The average binding free energies at the proximal heme site and substrate access channel have overlapping confidence intervals and the Hill coefficient from a dose–response curve is greater than 1.5. As such, we project that both sites presented in this work are occupied at saturating concentrations of AR11. Our MD simulations suggest that AR11 preferentially interacts at the substrate access channel since E357 is projected to be destabilizing.
Note that our control inhibitor, E/Z-endoxifen, exhibits similar trends as AR11 in this study. Both inhibitors dock to the proximal heme site and yield a Hill coefficient that suggests the presence of multiple binding sites. However, there are two differences that infer distinct inhibitory mechanisms. Firstly, MD simulations predict that E- and Z-endoxifen do not remain in the proximal heme site, which is consistent with work by Sgrignani et al.9 Secondly, E/Z-endoxifen does not cause disassociation of ASD from the active site. It likely occupies a distinct locale, whereas AR11 protrudes into a space that is occupied by ASD.
Titration with AR19 and AR20 induces a 4 nm red shift from a water-bound iron to 420 nm. Production runs revealed that AR19 was mobile in the substrate access channel and active site. It has a terminal furan capable of directly interacting with the heme iron. Additionally, it may form a water-bridged ternary complex to induce greater water–iron orbital overlap. AR20 is a 1,2,4-triazole that may also directly interact with iron or water. P450 ternary complexes have been reported with 1,2,3- and 1,2,4-triazoles.53 Alternatively, heme perturbation induced by allosteric interactions may cause such Soret peak shifts. This would correlate with MD simulations that project transient AR20-proximal heme interactions.
Altogether, we conclude that E/Z-endoxifen, AR11, and AR19 preferentially occupy the substrate access channel, inhibiting CYP19 on the same order of magnitude. Since the substrate access channel is dynamic, compounds forming weaker interactions such as AR19 may migrate towards the active site to form a direct or ternary complex with iron. E/Z-endoxifen may bind the substrate access channel with high affinity and a second site at saturating levels of inhibitor. We expect that AR11 interacts at the substrate access channel and the proximal heme site at saturating levels. In the substrate access channel, AR11 can bind deeper into the pocket such that it partially occupies space by ASD near the catalytically active D309. AR20 is projected to transiently interact at the proximal heme site.
MD simulations project that the proximal heme site is acting as a low-affinity binding site for multiple inhibitors. It is an attractive target to modulate P450 activity because it is a conserved feature among P450s and distinct from the catalytic core. Despite class 2 P450s having less than a 40% sequence identity, all of them are expected to partner with CPR at this site. CYP19 may serve as a model to study its druggability for many reasons. Firstly, a crystal structure with the pentameric PEG bound at the proximal heme site is available and PEG inhibited enzyme activity in a time-dependent manner.57 Secondly, by conformational selection, CYP19-reductase coupling affects the on and off rate of substrate at the active site.56 Thirdly, CYP19 has a well-defined cavity of 584 Å3, roughly 200 Å3 larger than the active site, that can accommodate a larger library of compounds 57. Lastly, the loop region between helices k’’ and L contains the meander region of 21 residues long. Roughly 30–40% of P450s have a loop 14 or 15 residues long-all of which are class 2 P450 enzymes. This disparity offers a niche to selectively target CYP19 as less than 5% of P450s have a meander loop as lengthy.58 It is likely that higher affinity inhibitors to the proximal heme site can be identified.
Other P450s that may be allosterically inhibited at the proximal heme site include CYP3A4 and CYP1A2. They are the major enzymes in drug metabolism and have well-documented heterotrophic effectors.59–61 It is possible that these P450s can be modulated at the proximal heme site as the cavities are large enough to accommodate ligand-binding. The CYP3A4 and CYP1A2 pockets are 1.1 and 0.7 times the size of the CYP19 proximal cavity.57 More research is needed to investigate the druggability of alternative ligand binding sites in P450s.62–65
In a broader context, nearly all exogenous compounds are subject to redox chemistry. Azoles and furan-containing xenobiotics, such as those presented here, are often P450 substrates.66 Many of these are important azole-containing drugs used to treat fungal infections.67 Profiling distinct chemical scaffolds can aid in predicting off-target P450 interactions that may lead to toxicity or adverse drug interactions.
Footnote |
† Electronic supplementary information (ESI) available. See DOI: 10.1039/d1cb00046b |
This journal is © The Royal Society of Chemistry 2021 |