Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jan 14.
Published in final edited form as: J Med Chem. 2021 Jan 4;64(1):566–585. doi: 10.1021/acs.jmedchem.0c01522

Generation of Highly Selective, Potent, and Covalent G Protein-Coupled Receptor Kinase 5 Inhibitors

Rachel A Rowlands a,*, Qiuyan Chen c,*, Renee A Bouley b,, Larisa V Avramova c, John J G Tesmer c,**, Andrew D White a,**
PMCID: PMC7909074  NIHMSID: NIHMS1669076  PMID: 33393767

Abstract

The ability of G protein-coupled receptor (GPCR) kinases (GRKs) to regulate the desensitization of GPCRs has made GRK2 and GRK5 attractive targets for treating diseases such as heart failure and cancer. Previously, our work showed that Cys474, a GRK5 subfamily-specific residue located on a flexible loop adjacent to the active site, can be used as a covalent handle to achieve selective inhibition of GRK5 over GRK2 subfamily members. However, the potency of the most selective inhibitors remained modest. Herein, we describe a successful campaign to adapt an indolinone scaffold with covalent warheads, resulting in a series of 2-haloacetyl containing compounds that react quickly and exhibit three orders of magnitude selectivity for GRK5 over GRK2 and low nanomolar potency. They however retain a similar selectivity profile across the kinome as the core scaffold, which was based on Sunitinib.

Keywords: covalent inhibitor, kinase inhibitor, GPCR, GPCR kinase, cardiac hypertrophy, cancer, selectivity

Graphical Abstract

graphic file with name nihms-1669076-f0001.jpg

INTRODUCTION

G protein-coupled receptor (GPCR) kinases (GRKs) selectively recognize and phosphorylate activated GPCRs, leading to their desensitization and internalization, a process critical for maintaining cellular homeostasis. The seven human GRKs (GRK1–7), are classified via structural and sequence similarity into three subfamilies: GRK1 (GRK1 and 7), GRK2 (GRK2 and 3), and GRK4 (GRK4, 5, and 6).1 GRK1 and 7 are found primarily in the retina and GRK4 in the testes, whereas GRK2, 3, 5, and 6 are more ubiquitously expressed. Of these kinases, GRK2 and GRK5 are the two isoforms with the highest concentration in cardiovascular tissue. Because they are overexpressed in the diseased heart and their inhibition or ablation has been shown to prevent heart failure and hypertrophic cardiomyopathy, they have become important therapeutic targets.2, 3 GRK2 and GRK5 are also potential targets for treatment of cancer4, 5 and other pathophysiological conditions1. GRK5 is further unique among GRKs because it undergoes Ca2+-calmodulin-dependent nuclear localization, where it phosphorylates histone deacetylase 5 (HDAC5) induces an increase in transcription of associated genes6. In studies where GRK5 was knocked down, cardiomyocytes were protected from hypertrophic cardiomyopathy7. The influence of GRK5 in progressive heart failure and hypertrophic cardiomyopathy however remains unclear, in part because GRK2 can also mediate hypertrophic responses8 and there are few compounds known to have clear GRK5 selectivity that could be used to test mechanistic hypotheses in physiologically relevant cells or animals.

RESULTS AND DISCUSSION

Previously, we developed a set of GRK5/6-selective pyrrolopyrimidine inhibitors using a covalent strategy9. This effort established that Cys474, located on a loop known as the active site tether (AST) that packs over the ATP binding site in AGC kinases, can serve as a covalent handle to achieve GRK5 selectivity. However, the affinities of the best compounds were low μM at best. Because the intrinsic affinity of the compound plays an important role in dictating the concentration of the protein-inhibitor covalent complex, which must persist long enough for a covalent reaction to occur10, the discovery of a more intrinsically potent scaffold for GRK5 inhibition was prioritized. To this end, we considered a set of known GRK5 modulators derived from an indolinone scaffold present in the FDA approved receptor tyrosine kinase (RTK) inhibitor Sunitinib (1) (Figure 1), a compound that targets both GRK5 and multiple RTKs including the platelet-derived growth factor receptor and vascular endothelial growth factor receptor4, 11. We focused on Ullrich-57 (5a), which was reported to have low nanomolar activity against GRK5 (GRK5 IC50 = < 0.1 μM), although its selectivity was not reported12. We independently synthesized 5a, and also tested its parent compound, Sunitinib and showed that 5a has orders of magnitude more potency against GRK5 than our previous pyrrolopyrimidine scaffold, and that both exhibit 1–2 orders of magnitude selectivity for GRK5 over GRK2 (Figure 1, Table 1). Modeling the 5a complex with GRK5 was performed by docking in the program MOE. The highest scoring pose predicted that 5a would bind in the active site of GRK5 in a fashion that would allow the formation of three hydrogen bonds with the hinge, which would in turn project its diethylamine moiety towards the AST loop (Figure 2A). We hypothesized that replacing the diethylamine arm of the scaffold with thiol reactive warheads could allow for covalent attachment to Cys474 and generation of even more selective and potent covalent GRK5 inhibitors. Thus, the goal for our first series of compounds was to identify a covalent warhead that affords the highest incorporation and the most selectivity for GRK5 over GRK2. As a control, potency against the canonical AGC kinase protein kinase A (PKA) was also evaluated, however all but one of our compounds had negligible effects against this enzyme.

Figure 1.

Figure 1.

Lead compounds 1 and 5a with design strategy.

Table 1:

IC50 Values (μM ± SD) and Reactivity of Indolinone Compounds

graphic file with name nihms-1669076-t0002.jpg
Compound R1 GRK5 GRK2 GRK2/GRK5 PKA Adduct by MS*
Sunitinib 1 graphic file with name nihms-1669076-t0003.jpg 0.83 ± 0.7 (3) 130 ± 200 (3) 150 ND NA
CCG
271421
5a graphic file with name nihms-1669076-t0004.jpg 0.015 ± 0.02 (7) 1.1 ± 0.7 (4) 74 >250 (2) NA
CCG
273262
5b graphic file with name nihms-1669076-t0005.jpg 1.30 ± 0.1 (3) 44 ± 18 (3) 32 ND NA
CCG
271423
5c graphic file with name nihms-1669076-t0006.jpg 0.021 ± 0.01 (7) 44 ± 40* (6) 2100 ND >30 min
CCG
271424
5d graphic file with name nihms-1669076-t0007.jpg 0.048 ± 0.008 (3) 22 ± 10 (3) 460 >250 (2) >30 min
CCG
271441
5e graphic file with name nihms-1669076-t0008.jpg 0.091 ± 0.04 (3) 130 ± 50 (3) 1400 >250 (2) >30 min
CCG
271442
5f graphic file with name nihms-1669076-t0009.jpg 1.94 ± 0.05 (2) 630 ± 200 (2) 330 ND >30 min
CCG
273183
7a NO2 0.73 ± 0.5 (3) 7.2 ± 3 (3) 10. ND ND
CCG
273180
9a graphic file with name nihms-1669076-t0010.jpg 2.5 ± 0.8 (3) 150 ± 30 (3) 61 ND >3 hr
CCG
273181
9b graphic file with name nihms-1669076-t0011.jpg 0.81 ± 0.7 (4) 87 ± 30 (3) 110 ND >3 hr
CCG
273182
9c graphic file with name nihms-1669076-t0012.jpg 0.74 ± 0.6 (5) 280 ± 110 (3) 370 ND 100% 3 hr
CCG
273220
9e graphic file with name nihms-1669076-t0013.jpg 0.22 ± 0.04 (3) 350 ± 100 (2) 1500 >250 (2) 90% 30 min
CCG
273221
9f graphic file with name nihms-1669076-t0014.jpg 0.36 ± 0.2 (3) 17 ± 10 (2) 47 ND >30 min
CCG
273463
9g graphic file with name nihms-1669076-t0015.jpg 0.0086 ± 0.003 (7) 12 ± 20 (3) 1400 >250 (2) 70% 30 min
CCG
273464
9h graphic file with name nihms-1669076-t0016.jpg 0.08 ± 0.03 (3) 6.7 ± 5 (3) 83 >250 (2) >30 min
CCG
273240
10a graphic file with name nihms-1669076-t0017.jpg 0.28 ± 0.1 (3) 120 ± 80 (2) 430 >250 (2) >30 min
CCG
273462
10b graphic file with name nihms-1669076-t0018.jpg 0.74 ± 0.2 (3) >250 (5) >340 ND >30 min
CCG
215022
- 0.28 ± 0.1 (6) ND - >250 (2) NA
paroxetine - ND 0.78 ± 0.3 (3) - 850 ± 400 (4) NA

All data were fit to a log([inhibitor]) versus response model with variable slope and automatic outlier rejection in GraphPad Prism. Curves that had R squared values less than 0.8 after fitting were omitted. ND, not determined. NA, not applicable Values in parentheses indicate the number of independent experimental curves. CCG215022 and paroxetine are positive controls for GRK5 and GRK2, respectively.

*

Incubation time needed to observe adduct formation with GRK5 by intact mass MS.

5a and 5b are the (R) and (S) enantiomers of Ullrich-57, respectively.

Figure 2.

Figure 2.

Docking models of lead compound 5a and most potent derivative 9j, and their comparison with the GRK5·CCG215022 complex. (A) 5a, with pink carbons, red oxygens, and blue nitrogens docked using the program MOE into the structure of GRK5 (grey, PDB ID 4WNK), with the addition of the AST loop from GRK6 (purple, PDB ID 3NYN) because this element was disordered in the 4WNK structure and contains the target Cys474 residue. The diamine moiety extends towards the AST loop where Cys474 is located. Three hydrogen bonds, shown as dashed lines, are formed with the hinge of the kinase domain. None of the compounds in the 5a-f series were able to form adducts despite some that demonstrated high potency. (B) 9j (yellow carbons) wherein the amide functionality in the covalent warhead was flipped relative to 5a, and therefore forms one less hydrogen bond with the hinge. Being less constrained, we hypothesize that this modification allows the warhead to leave the hinge region along a different vector that leads to less steric collisions with the AST and to closer proximity with Cys474. Consequently, compounds in the 9a series show adduct formation. Bromine is colored green. (C) Comparison of the fluorophenyl groups of 9j and CCG215022 (from 4WNK). The fluorine atom is colored cyan. Both ligands are modeled to form a hydrogen bond with Asp329, an invariant catalytic residue in protein kinases. However, it is unclear whether this region of the scaffold is docked correctly, because the conformation of the GRK5 kinase domain, and in particular its P loop and AST, is not known. The SAR in Table 2 is best explained with the fluorophenyl packing under the P loop as it does in the CCG215022 complex.

Synthesis for this series began with an amide coupling to give common intermediate 3 (Scheme 1). In a convergent line of synthesis, a secondary amide coupling with the starting material 6, gave intermediates 4a-e. Combining the two lines of synthesis, a Knoevenagel condensation yielded compounds 5a-f, wherein 5a and 5b represent the (R) and (S) enantiomers of Ullrich-57. We found that the (S)-enantiomer 5b was over 1000-fold less potent (Table 1), indicating that the (R)-enantiomer (5a) places the methyl and benzyl pendants in a more ideal position. The structural rationale for this based on the dock remains elusive because of unknowns about the overall conformation of the GRK5 kinase domain (see Figure 2). All other compounds were therefore synthesized with the same stereo-configuration as 5a.

Scheme 1.

Scheme 1.

Convergent route to 5a-f. 5a and 5b are enantiomers derived from 3 and 3a, respectively

Initial diversification tested a series of alkenyl or alkynyl amines as covalent modifiers. The propargyl analogue, 5c, demonstrated similar potency to 5a (GRK5 IC50 = 21 nM) but exhibited over a magnitude higher selectivity over GRK2 (2100-fold). We speculate this high level of selectivity is due to a potential clash of the propargyl warhead with the GRK2 AST and/or large lobe given the predicted vector for the alkynyl and alkenyl groups (Figure 2A). For all other amide linked compounds with an alkenyl or alkynyl warhead (5d-f), the selectivity for GRK5 over GRK2 remained between 330–1400 fold, but in each case the IC50 for GRK5 was higher than that of 5c (Table 1). Linker length also contributed to GRK5 affinity, as demonstrated by comparison of 5c with 5e, and 5d with 5f, wherein the latter compounds have homologated alkenyl and alkynyl warheads and somewhat less potency. Thus, a single methylene linker to the reactive center seems optimal for maintaining GRK5 activity among these trial compounds.

By reversing the warhead amide, the covalent warheads used in our previous work9 would become synthetically accessible. To accomplish this, a Knoevenagel with 3,5-dimethyl-4-nitro-1-pyrrolocarb-aldehyde yielded common intermediate, 7a (Scheme 2). 7a was itself tested for inhibitory activity because it possesses the ability to release nitric oxide, a known vasodilator, rendering it a possible dual mechanism compound13. The nitro group was however poorly tolerated (GRK5 IC50 = 730 nM) and only 10-fold selective for GRK5. A zinc-catalyzed reduction of the nitro group of 7a to the free amine 8a allowed for rapid derivatization through amide coupling to yield final compounds 9a-h (Scheme 2). 9a, which features a 2-butynyl acid warhead that reacted with Cys474 in our prior study9 exhibited low μM potency (Table 1). The commonly used acrylamide and vinyl sulfonamide variants, 9b and 9c, respectively, showed high nanomolar activity against GRK5 and retained >100 fold selectivity over GRK2. The 2-chloroacetylamido containing compound 9e showed moderate potency (GRK5 IC50 = 220 nM) but 1500-fold selectivity over GRK2. Because 5a had low-nanomolar potency, we introduced a similar reactive appendage, dimethylaminobutenoic acid, in 9f. This compound showed an increased IC50 (360 nM, thus 24-fold higher than 5a), but only 50-fold selectivity over GRK2. The 2-bromoacetylamido compound, 9g was found to have a greater potency against GRK5 (IC50 = 8.6 nM) than its chloro analog, 9e while retaining a high level of selectivity against GRK2 (1400-fold). Thus, we concluded that the bromo group of 9g must be better filling a lipophilic pocket than the chloro group, but it is not possible to model this because the structure of the AST loop is uncertain in our GRK5/6 overlay model (Figure 2). It was also observed that 9g took longer to fully engage Cys474 within the 30 min incubation time frame (Figure 3). 9h was also potent against GRK5 (IC50 = 80 nM) whereas compounds 10a-b exhibited only moderate potency.

Scheme 2.

Scheme 2.

General synthetic route to 9a-h and 10a-b.

Figure 3.

Figure 3.

Intact protein MS for compounds 9e, 9g, and 9j. (A) GRK5 (blue) and GRK5+9e (black) incubated for 30 min. 9e demonstrated full labelling of GRK5. (B) GRK5 and GRK5+9g (pink) incubated for 30 min. 9g only labeled 50% of GRK5. (C) GRK5 and GRK5+9j (red) incubated for 30 min. 9j fully labeled GRK5 in this timeframe. (D) GRK5-C474S mutant (purple) and GRK5-C474S+9e (teal) incubated for 30 min. 9e did not label GRK5-C474S, indicating that 9e and related compounds are engaging Cys474.

We evaluated adduct formation in this series at 30 min and 3 hours by intact mass spectrometry (MS) (Table 1, Figure 3, Figure S1). Only a few compounds exhibited signal for GRK5 at the 3 hour timepoint. 5c, which exhibited similar potency to the parent compound 5a and over 2000-fold selectivity against GRK2 was, surprisingly, not covalent under our conditions. Neither were compounds 5d-f. 9a and 9b were likewise unreactive at the 30 minute timepoint. The vinyl sulfonamide 9c was able to react after a 3 hour incubation (Figure S1) despite its moderate affinity (740 nM). The chloroacetyl 9e however had nearly complete covalent engagement by 30 min (Figure 3A). 9e was unreactive against GRK5-C474S, consistent with Cys474 being the covalent handle (Figure 3D). Its bromoacetyl analog 9g also rapidly reacted with GRK5 (Figure 3B), but 9h, 10a and 10b were unreactive at 30 minutes. Based on the sum of the data, we rationalize that compounds in the 5c-f series were unreactive because of constraints placed on the warhead by increased hydrogen bond interactions with the GRK5 hinge relative to 9c, 9e and 9g where the attaching amide was flipped, affording a different vector for the attached warhead and less conformational constraints (Figure 2 A,B).

At this point, 9e and 9g provided the most reactivity combined with the most selectivity for GRK5 over GRK2 (Table 1). Therefore, we initialized structure-activity relationships (SAR) around indolinone scaffolds bearing 2-haloacetylamido warheads (Schemes 34, Table 2). Benzyl and pyridyl pendants in the R1 position (Table 2) were explored first. Overall, in terms of potency GRK5 seemed to have a strong preference for smaller electron withdrawing substituents in the para-position of the benzyl groups: F (9j) > Cl (9o) > H (9e) > CH3 (9n). The MOE docked models do not explain this preference, but such behavior would be expected if the benzyl pendant packs instead under the P loop of the active site. In fact, most known GRK inhibitors, including CCG215022, tend to have benzyl groups that pack in this site (Figure 2C)14. The enhancement in potency of 3-Me (9i) is large compared to 9e, suggesting that a 3-Me, 4-F analog would be even more potent. However, a lack of appropriate chemical precursors prevented us from studying such combinations. We note that we took advantage of the meta position of the fluorobenzyl group of paroxetine to make a highly successful series of GRK2 selective inhibitors15. The position of the nitrogen in pyridyl pendants also appeared to make a small difference. In 9k, the potency was 3.5-fold higher than that of 9l with an ortho-nitrogen, suggesting once again that the para-nitrogen in 9k fulfills an electronic deficiency. 9j, with a 4-fluoro substituent, had 55-fold more potency (IC50 = 4 nM) relative to the parent compound 9e. It was also able to rapidly form a covalent interaction with Cys474 within 30 min (Figure 3C), 9j was thus the most intriguing lead of the second series (Table 2, Figure 2C). When the 4-fluoro substituent was maintained, and a 2-bromoacetylamido warhead was used, the resulting compound, 9p, showed slightly lower potency (IC50 = 15 nM) than 9j or the des-fluoro compound 9g (Table 1). This is different than expected from comparison of 9g and 9e (Table 1), where the bromo substitution rendered much higher potency. The structural explanation for this is unclear.

Scheme 3.

Scheme 3.

General synthetic route to 9i-p.

Scheme 4.

Scheme 4.

General synthetic route to 9q-t.

Table 2:

IC50 Values (μM ± SD) and Reactivity of Indolinone Variants with a Haloacetylamido Warhead

graphic file with name nihms-1669076-t0019.jpg
Compound R1 R2 GRK5 GRK2 GRK2/GRK5§ PKA Adduct by MS*
CCG
273261
9i 3-MeBn (R)-CH3 0.029 ± 0.03 (5) 11 ± 6 (3) 360 >250 (6) >30 min
CCG
273441
9j 4-FBn (R)-CH3 0.0038 ± 0.001 (7) 4.8 ± 3 (3) 1300 >250 (6) 90% 30 min
CCG
273442
9k 4-Py (R)-CH3 0.13 ± 0.05 (3) 1.7 ± 2 (3) 13 ND >30 min
CCG
273443
91 2-Py (R)-CH3 0.45 ± 0.1 (3) 9.6 ± 5 (3) 21 ND >30 min
CCG
273444
9m 3-ClBn (R)-CH3 0.14 ± 0.04 (3) 19 ± 10 (3) 140 >250 (6) >30 min
CCG
273445
9n 4-MeBn (R)-CH3 0.78 ± 03 (3) 2.1 ± 1 (5) 3 ND >30 min
CCG
273583
9o 4-ClBn (R)-CH3 0.11 ± 0.05 (3) 0.70 ± 0.2 (3) 7 ND 50% 8 hr
CCG
359090
9p 4-FBn (R)-CH3 0.015 ± 0.007 (3) 3.6 ± 2 (3) 230 ND ND
CCG
273561
9q Bn H 0.13 ± 0.09 (4) 13 ± 3 (3) 99 >250 (6) 50% 8 hr
CCG
273562
9r Bn Gem-(CH3)2 12 ± 5 (3) 190 ± 70 (2) 15 ND 50% 8 hr
CCG
273564
9s Bn (R)-iPr 0.087 ± 0.02 (3) 23 ± 20 (4) 260 >250 (6) 90% 8 hr
CCG
273563
9t Bn graphic file with name nihms-1669076-t0020.jpg 0.095 ± 0.03 (3) 15 ± 7 (3) 160 69±10 (6) >8 hr

Data were fit to a log([inhibitor]) versus response model with variable slope and automatic outlier rejection in GraphPad Prism. Curves with R2 values <0.8 after fitting were omitted. ND, not determined. Numbers in parentheses indicate the number of independent experimental curves.

*

Incubation time needed to observe adduct formation with GRK5 by intact mass MS.

Same as 9j but with a 2-bromoacteylamido warhead.

IC50 estimate for this compound for GRK5 and GRK2 are likely high due to its poor solubility in our assay system.

Given the profound effects of chirality on activity exhibited by compounds 5a and 5b, we also expanded SAR around the benzylic position of the scaffold (R2 in Table 2). When the stereocenter was removed (9q), there was a small increase in potency (IC50 = 130 nM) relative to the parent compound 9e (less than 2-fold). However, there were also similar increases in potency for 9s (GRK5 IC50 = 87 nM) and 9t (GRK5 IC50 = 95 nM). The geminal dimethyl of 9r however had greatly reduced potency (IC50 = 12 μM), but the poor solubility of this compound under our assay conditions may have artifactually increased IC50 measurements. Therefore, we concluded that the benzylic position (R2 in Table 2) is fairly insensitive to modification, at least when the groups concerned are the size of isopropyl or smaller. Accordingly, when 9q, 9r, and 9s were incubated for 8 hours, covalent interaction with GRK5 was observed as in 9e (Figure S2). It is not clear why 9t did not also react.

Finally, although the indolinone scaffold offers high potency and selectivity for GRK5 over GRK2, there were a few anticipated drawbacks in terms of its pharmacokinetic properties. First, 9a-t are less soluble due to the number of aromatic rings and their rigid, linear conformation. The pyridyl pendants of 9k and 9l were a first attempt to address this issue. 9l was found to improve solubility 3.5 fold over 9e to 125 μg/ml (Table S1), which is still ~3-fold less than reported for Sunitinib (350 μg/ml). Metabolic liability was also a concern because of the high lipophilicity of our compounds and thus their potential metabolism by CYP3A4. Pyridyl pendants are also known to limit metabolic liability relative to benzyl rings, as are fluorine containing pendants, as in 9j (Figure S4). We indeed found that 9j indeed had a longer half-life (HLM t1/2 =18.9 min) compared to the des-fluoro compound 9e (HLM t1/2 = 13.3 min). However, there was an opposite trend in MLMs. Limited commercial availability of fluorine substituted benzylic amines from the chiral pool however limited our ability to explore the additional ortho and meta-substitution patterns.

Ullrich-57 (5a) is already a potent (15 nM) and moderately selective GRK5 inhibitor relative to GRK2 (74-fold). Our overarching goal here was to ascertain whether potency and selectivity could be improved by covalent modification of a residue unique to the GRK5 subfamily of kinases. Interestingly, although 9e, 9g, and 9i-t all had 2-haloacetylamido warheads, only a few were able to label Cys474 within a 30 minute time period. The absence of reactivity could be explained by the higher IC50 exhibited by many of these analogs. Those compounds with lower affinity for GRK5, for example 9c, will bind in the active site of GRK5 more transiently than 9e, resulting in a longer incubation period being needed to detect covalent engagement (Figure S1). 9e, 9g, and 9j were not only the most efficacious at modifying GRK5 (Figure 3), but also they spanned a range of IC50 values (220–4 nM, respectively), and exhibited selectivity for GRK5 over GRK2 by over three orders of magnitude, 20 fold-more than exhibited by the noncovalent compound 5a. We therefore performed a detailed kinetic analysis on these compounds to gauge the impact of their reactivity on measured potency and selectivity. We first showed that all three compounds exhibited 3–5 fold less potency against GRK5-C474S (Table 3), whereas our control compound CCG215022 was essentially unchanged. We then determined KI and kinact for these compounds (Figure 4, Table 3) which showed that 9e, 9g, and 9j fully reacted with GRK5 before the first time point (hence kinact>1 min−1) and that KI scaled with the observed IC50 values reported for these compounds (Tables 12). This shared reactivity is consistent with the fact that they all contain haloacetyl warheads. The results further suggest that a direct comparison of the SAR among compounds in Table 2 along with 9e and 9g from Table 1 is reasonable. It also implies that in this situation, where the reactive cysteine in on a flexible loop near the ligand binding site, KI dominates the IC50 measurements. More reactive warheads like 2-haloacetyls may also be required to take advantage of a more transiently associated cysteine side chain given the anticipated conformational variability of the AST loop.

Table 3:

Impact and Kinetics of Covalent Modification at GRK5-Cys474

Compound GRK5 IC50 (μM) GRK5-C474S IC50 (μM) GRK5-C474S/GRK5 KI (μM) kinact (min−1)
CCG
273220
9e 0.22 ± 0.04 (3) 0.60 ± 0.2 (5) 3 1.0 ± 0.3 (3) >1.0 ± 0.05 (3)
CCG
273463
9g 0.0086 ± 0.003 (7) 0.044 ± 0.02 (5) 5 0.11 ± 0.03 (3) >1.0 ± 0.03 (3)
CCG
273441
9j 0.0038 ± 0.001 (7) 0.019 ± 0.007 (5) 5 0.019 ± 0.01 (3) >0.9 ± 0.02 (3)
CCG
215022
0.28 ± 0.01 (6) 0.22 ± 0.05 (5) 0.8 n/a n/a

Errors correspond to standard deviation, with number of replicates given in parentheses. n/a, not applicable.

Figure 4.

Figure 4.

Kinetic analysis for (A, B) 9e, (C, D) 9g, and (E, F) 9j. Extracted values for KI and kinact from fitting the plots on the right are given in Table 4. Data points represent three measurements with error bars indicating standard deviation. Because the time courses (left panels) were not linear, only the 1 minute time point was used to estimate kobs.

Having confirmed that 5c and 9g were potent and highly GRK5 selective relative to GRK2, we explored their kinome wide selectivity. When tested at 1 μM (~1000-fold higher than their IC50 values), 5c inhibits GRK5 at 92%, and GRK6 at 94%, whereas 9g inhibits GRK5 at 93%, and GRK6 at 97%. GRK3, a very close homolog of GRK2, was inhibited at only 21%. Both compounds had many off-target effects across the kinome (Figure 5 and Figure S3), which is not surprising because the indolinone series is derived from Sunitinib, which can similarly inhibit the activity of many RTKs. We confirmed that selectivity improved at lower concentrations by testing 9g at 100 and 10 nM (Figure 4). 9g however continued to inhibit a small number of tyrosine kinases and Ca2+/calmodulin-dependent protein kinases at the lowest dose, at which GRK5 was 50% inhibited, consistent with the IC50 value measured in our assays (Table 1).

Figure 5.

Figure 5.

Kinome-wide selectivity panel for 9g at concentrations of (a) 1 μM, (b) 0.1 μM, and (c) 0.01 μM. Locations of GRK5 and GRK6 are denoted by arrows. A clear dose response is evident, with fewer kinases inhibited at the lowest concentration of 9g. Illustration reproduced courtesy of Cell Signaling Technology, Inc (www.cellsignal.com).

CONCLUSIONS

In summary, we have developed a potent series of covalent inhibitors based on the indolinone scaffold that show higher potency and three orders of magnitude selectivity for GRK5 over the GRK2 subfamily, improving on the potency and selectivity of the parent compound Ullrich-57 (4-fold and 74-fold, respectively). Our MS and kinetic results further suggested that their ability to covalently engage a cysteine unique to the GRK5 subfamily, which contributes 3–5 fold to their IC50 values under our assay conditions, is responsible for these improved characteristics. The compounds thus set the stage for cell based assays that would allow one to tease apart the roles of GRK5 versus GRK2 in cellular processes linked to cardiovascular disease and cancer. They also set the stage for a future generation of GRK5 subfamily selective therapeutics that would address potential toxicity issues associated with the use of strongly reactive 2-haloacetyls, further improve selectivity versus other subfamilies of protein kinases and their moderate solubility and metabolic stability. Such would facilitate their transition into in vivo trials. While this paper was under review, a new GRK5 inhibitor, KR-39038, with 20 nM IC50 was reported, although experiments addressing its selectivity versus GRK2 and other kinases and its toxicology were not provided16. This compound showed mild positive effects in a rodent model of hypertrophy but exhibited low bioavailability and a short half-life (<1 hour). These results reinforce the potential utility of covalent GRK5 inhibitors that could long-lasting effects even if the circulating compound is rapidly cleared, which would in turn mitigate off-target toxicity10.

EXPERIMENTAL SECTION

General Chemistry.

All reagents from commercial sources were used without further purification unless otherwise noted. 1H-NMR spectra were taken in DMSO-d6, MeOD or CDCl3 at room temperature on Varian MR 400 MHz, Varian Vnmrs 500 MHz, and Varian Vnmrs 700 MHz instruments. The reported chemical shifts for the 1HNMR spectra were recorded in parts per million (ppm) on the δ scale from an internal tetramethylsilane standard (0.0 ppm). Small molecule mass spectrometry data was measured using a Waters Corporation Micromass LCT or Agilent6230 Q-TOF instrument. HPLC was used to determine purity of compounds on an Agilent 1100 series with an Agilent Zorbax Eclipse Plus-C18 column. A gradient of 10–90% acetonitrile/water over 6 minutes followed by 90% acetonitrile/water for 7 minutes was used with detection at 254 nm. Purity of all compounds was > 95% as determined by HPLC. The Sunitinib scaffold is well known to photoisomerize in solution17, and thus our compounds represent a 70:30 mix of active versus inactive isomers. All figures in the paper depict the active isomer.

Intact Protein MS and Tandem MS/MS.

Intact protein MS was acquired with a Phenomenex C4 column paired with an Agilent 6545 Q-TOF LC/MS. For intact MS and Tandem MS, all samples were prepared with 20 μM GRK in assay buffer (see below), 1 mM compound, and incubated at 4 °C for 3 hr before being quenched with 1.0 μL of formic acid. In Tandem MS/MS, we chose Glu-C as the restricting enzyme to avoid small fragments with mass-to-charge ratios below the limit of detection. All samples were digested with Glu-C sequencing enzyme, procured from Sigma Aldrich (Roche Life Sciences subsidiary) and used without further purification. MS/MS experiments were run on a nano-LC (Dionex RSLC-nano) with an Orbitrap Fusion Tribrid ETD mass spectrometer. This work was conducted by the Proteomics Resource Facility at the University of Michigan.

Structural Models and Docking.

GRK5 (PDB ID 4WNK)18 and GRK6 (PDB ID 3NYN)19 were loaded into Molecular Operating Environment 2018.01 (Molecular Operating Environment (MOE), 2018.01; Chemical Computing Group ULC, 1010 Sherbrooke St. West, Suite #910, Montreal, QC, Canada, H3A 2R7, 2018) and proteins were prepared using QuickPrep function. The sequences of both proteins were aligned and used to create a super-position of the two proteins and a hybrid structure with the kinase domain of GRK5 and the AST region of GRK6 was created for docking analysis (Figure 1A). The highest scoring docked pose for each compound in Tables 12 are provided as Supporting Information.

Inhibition Assays.

For compounds 5a-9t, IC50 values for human GRK5 and bovine GRK2 were determined using a radiometric assay, described as follows. 50 nM GRK was incubated for 3–5 minutes with 500 nM porcine brain tubulin (PurSolutions) and 0.01 – 50 μM inhibitor in 20 mM HEPES pH 7.0, 2 mM MgCl2, 0.025% dodecylmaltoside (DDM), 1% DMSO, prior to initiation with the addition of 5 μM ATP supplemented with radioactive [γ-32P]-ATP (PerkinElmer Life Sciences). Reactions were quenched at 8 minutes by addition of 5 μL of 4X SDS gel loading dye to the 10 μL reactions. 12 μL samples were separated on a 4–15% Criterion TGX precast gel (Bio-Rad). For potent inhibitors with low nanomolar IC50, the inhibitor concentration was adjusted to approximately 0 – 50x [IC50] which was estimated from the first run to get more accurate measurements. Gels were dried, exposed to a storage phosphor screen overnight, and scanned using a Personal Molecular Imager (Bio-Rad). Bands corresponding to phosphorylated tubulin were quantified using ImageQuant, plotted as a function of log[inhibitor], and fit to the four-parameter log(inhibitor) vs. response model in GraphPad Prism 7.03 to determine the IC50, and mean and standard deviation values. Outliner was eliminated automatically at 1% Q value. Experiments were performed at least three times.

PKA inhibition assays were performed with the ADP-Glo system (Promega Corporation) according to the manufactory’s instruction. 500 nM of PKA was incubated with 1 μg of CREBtide (KRREILSRRPSYR) (Genscript Corporation) substrate, 50 μM ATP and inhibitor for 30 minutes in 20 mM HEPES pH 7.0, 2 mM MgCl2, 0.025% dodecylmaltoside (DDM), 4% DMSO. The concentration range of each inhibitor varies depending on its solubility at 4% DMSO with the highest concentration from 100 μM to 500 μM. After the initial reaction, ADP-Glo reagent was added to the reaction and allowed to incubate for an additional 40 minutes. Lastly, the kinase detection reagent was added and allowed to incubate for 30 minutes, and the luminescence was measured with a FlexStation 3 Multi-mode Microplate Reader (Molecular Devices). All data was analyzed in the same way as GRK inhibition assay. Experiments were performed three times in duplicate.

Standard control compounds are run during each assay to assess consistency across time, experimenters, and subtle changes in assay conditions that are sometimes required to keep compounds soluble and dispersed (e.g. through addition of DDM or 3% DMSO). Paroxetine were used as controls for GRK220 and PKA, and CCG215022 for GRK518.

Covalent inhibition kinetic analysis.

For the covalent inhibitors 9e, 9g, and 9i, KI and kinact were determined using a radiometric assay as follows. The reactions contained 50 nM GRK5 and 5 μM porcine brain tubulin (PurSolutions) in 20 mM HEPES pH 7.0, 2 mM MgCl2, 0.025% DDM, 1% DMSO. Inhibitors at different concentrations (0.02–1 μM for 9j and 9g, 0.3–20 μM for 9e) were incubated with the reaction mix for 30 seconds, prior to initiation with the addition of 5 μM ATP supplemented with radioactive [γ-32P]-ATP (PerkinElmer Life Sciences). Reactions were quenched at 1, 2 and 5 minutes by addition of 10 μL of 4X SDS gel loading dye to the 10 μL reactions. 12 μL samples were separated on a 4–15% Criterion TGX precast gel (Bio-Rad). Gels were dried, exposed to a storage phosphor screen overnight, and scanned using a Personal Molecular Imager (Bio-Rad). Bands corresponding to phosphorylated tubulin were quantified using ImageQuant, background corrected, normalized to the intensity level of phosphorylation tubulin in the absence of any inhibitor, and plotted as a function of time. Because the inactivation process became nonlinear after the first time point, the observed inactivation rate (kobs) was estimated using the one-minute time point. The kobs values were re-plotted against inhibitor concentration and fitted to the equation, kobs = kinact [I]/(KI+[I]), to obtain kinact and KI in GraphPad Prism 7.03. Experiments were performed three times.

Thermodynamic Solubility and Microsomal Stability.

Thermodynamic solubility for compounds was determined by Analiza Inc. (Cleveland OH, analiza.com) using a miniaturized shake-flask solubility assay. Microsomal stability determined by the Pharmacokinetics and Mass Spectrometry Core at the University of Michigan. Compounds 9c, 9e and 9j were dissolved in DMSO (1 mM), and then further diluted to 100 μM with 0.1 M phosphate buffer (with 3.3 mM MgCl2). Microsomes (20 mg/mL) in 0.1 M phosphate buffer (with 3.3 mM MgCl) were dosed with 20 μL of NADPH (4 mg in 240 μL of 0.1 M phosphate buffer) and incubated at 37 °C for 3 minutes. Microsomes were then dosed with 4 μL of 100 μM of 9c, 9e and 9j respectively. At the following time points, 0, 5, 10, 15, 30, 45 and 60 minutes, the reaction solutions were then stopped with cold acetonitrile containing 25 nM CE302 as an internal standard. The incubation solution was centrifuged at 3500 rpm for 10 minutes to precipitate protein. The supernatant was used for LC/MS/MS analysis.

Chemical Synthesis and Validation.

(R)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (3).

To a round bottom flask was added 297.7 mg (1.69 mmol) of 2-oxoindoline-5-carboxylic acid, dissolved in 7 mL of dry DMF. To this dark red solution were added 0.244 mL (1.88 mmol) of (S)-1-phenylethan-1-amine, 0.300 mL (1.69 mmol) of DIPEA and 746.1 mg (1.95 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 401.9 mg, 84% Molecular Formula: C17H16N2O2 ESI-MS calc: 280.12 ESI-MS found: 281.1283 [M+1] HPLC: 5.198 1H NMR (400 MHz, DMSO-d6) δ 10.62 (s, 1H), 8.63 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 7.0 Hz, 2H), 7.39 – 7.35 (m, 2H), 7.31 (dd, J = 8.4, 6.8 Hz, 2H), 7.23 – 7.18 (m, 1H), 6.88 – 6.83 (m, 1H), 5.15 (p, J = 7.2 Hz, 1H), 3.53 (s, 2H), 1.46 (d, J = 7.1 Hz, 3H). 13C NMR (100 MHz, DMSO) δ 177.17, 165.76, 146.85, 145.65, 129.29, 128.67, 126.52, 126.06, 108.90, 48.85, 36.09, 22.84.

(S)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (3a).

To a round bottom flask was added 201.8 mg (1.13 mmol) of 2-oxoindoline-5-carboxylic acid dissolved in 7 mL of dry DMF. To this dark red solution were added 0.160 mL (1.24 mmol) of (S)-1-phenylethan-1-amine, 0.200 mL (1.13 mmol) of DIPEA and 492.4 mg (1.30 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 280 mg, 84% Molecular Formula: C17H16N2O2 ESI-MS calc: 280.12 ESI-MS found: 281.0903 [M+1] HPLC: 5.249 1H NMR (500 MHz, DMSO-d6) δ 10.67 (s, 1H), 8.66 (d, J = 8.0 Hz, 1H), 7.78 (d, J = 7.2 Hz, 2H), 7.38 (d, J = 8.4 Hz, 2H), 7.31 (t, J = 7.6 Hz, 2H), 7.20 (t, J = 7.3 Hz, 1H), 6.86 (d, J = 8.2 Hz, 1H), 5.15 (p, J = 7.2 Hz, 1H), 3.53 (s, 2H), 1.46 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 176.83, 165.50, 146.49, 145.22, 128.27, 127.77, 127.54, 126.60, 126.14, 125.64, 123.69, 108.62, 48.52, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 35.70, 22.40.

(R)-2-oxo-N-(1-(m-tolyl)ethyl)indoline-5-carboxamide (3b).

To a round bottom flask was added 236.1 mg (1.33 mmol) of 2-oxoindoline-5-carboxylic acid, dissolved in 7 mL of dry DMF. To this dark red solution were added 0.200 mL (1.47 mmol) of (R)-1-(m-tolyl)ethan-1-amine, 0.300 mL (1.73 mmol) of DIPEA and 510.2 mg (1.33 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 125.6 mg, 31% Molecular Formula: C18H18N2O2 ESI-MS calc: 294.14 ESI-MS found: 295.0167 [M+1] HPLC: 5.395 1H NMR (700 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.58 (d, J = 8.1 Hz, 1H), 7.77 (d, J = 8.1 Hz, 2H), 7.18 (p, J = 7.6 Hz, 4H), 7.02 (d, J = 7.3 Hz, 1H), 6.85 (d, J = 7.9 Hz, 1H), 5.11 (p, J = 7.3 Hz, 1H), 2.28 (s, 3H), 1.44 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 176.62, 165.16, 146.32, 145.07, 137.11, 128.06, 127.65, 127.48, 127.10, 126.66, 125.53, 123.51, 123.09, 108.36, 48.29, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 38.22, 35.57, 22.34, 21.11.

(R)-N-(1-(4-fluorophenyl)ethyl)-2-oxoindoline-5-carboxamide (3c).

To a round bottom flask was added 246.6 mg (1.41 mmol) of 2-oxoindoline-5-carboxylic acid, dissolved in 7 mL of dry DMF. To this dark red solution were added 0.200 mL (1.55 mmol) of (R)-1-(4-fluorophenyl)ethan-1-amine, 0.25 mL (1.41 mmol) of DIPEA and 667.1 mg (1.61 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 315 mg, 71% Molecular Formula: C17H15FN2O2 ESI-MS calc: 298.11 ESI-MS found: 299.1216 [M+1] HPLC: 5.414 1H NMR (500 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.62 (d, J = 8.0 Hz, 1H), 7.76 (d, J = 7.5 Hz, 2H), 7.43 – 7.38 (m, 2H), 7.16 – 7.08 (m, 2H), 6.87 – 6.82 (m, 1H), 5.14 (p, J = 7.2 Hz, 1H), 3.53 (s, 2H), 1.45 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 176.84, 165.47, 160.34, 146.46, 141.34, 128.06, 128.01, 127.77, 127.49, 125.69, 123.63, 114.99, 114.87, 108.55, 47.90, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 38.33, 35.67, 22.35.

(R)-2-oxo-N-(1-(pyridin-4-yl)ethyl)indoline-5-carboxamide (3d).

To a round bottom flask was added 248.1 mg (1.41 mmol) of 2-oxoindoline-5-carboxylic acid dissolved in 7 mL of dry DMF. To this dark red solution were added 0.200 mL (1.55 mmol) of (R)-1-(pyridin-4-yl)ethan-1-amine, 0.25 mL (1.41 mmol) of DIPEA and 652.4 mg (1.61 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 200 mg, 48% Molecular Formula: C16H15N3O2 ESI-MS calc: 281.12 ESI-MS found: 282.1236 [M+1] HPLC: 2.378 1H NMR (700 MHz, DMSO-d6) δ 10.64 (s, 1H), 8.74 (dd, J = 7.6, 2.1 Hz, 1H), 8.55 – 8.52 (m, 2H), 7.79 (dd, J = 7.5, 2.2 Hz, 2H), 7.45 – 7.42 (m, 2H), 7.08 (d, J = 2.3 Hz, 1H), 7.01 (s, 1H), 6.89 – 6.85 (m, 1H), 5.14 (td, J = 7.3, 2.2 Hz, 1H), 3.55 (s, 2H), 3.46 – 3.41 (m, 3H), 1.47 (dd, J = 7.2, 2.3 Hz, 3H), 1.07 – 1.03 (m, 3H). 13C NMR (176 MHz, DMSO) δ 176.63, 165.65, 155.16, 148.63, 146.56, 127.76, 127.03, 125.62, 123.57, 121.58, 108.43, 56.00, 47.92, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 35.56, 21.45, 18.54.

(R)-2-oxo-N-(1-(pyridin-2-yl)ethyl)indoline-5-carboxamide (3e).

To a round bottom flask was added 251.1 mg (1.41 mmol) of 2-oxoindoline-5-carboxylic acid dissolved in 7 mL of dry DMF. To this dark red solution were added 0.200 mL (1.55 mmol) of (R)-1-(pyridin-2-yl)ethan-1-amine, 0.25 mL (1.41 mmol) of DIPEA and 625.1 mg (1.62 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 165 mg, 39% Molecular Formula: C16H15N3O2 ESI-MS calc: 281.12 ESI-MS found: 282.1236 [M+1] HPLC: 2.111 1H NMR (700 MHz, DMSO-d6) δ 10.62 (d, J = 5.2 Hz, 1H), 8.63 (t, J = 6.6 Hz, 1H), 8.51 (t, J = 5.3 Hz, 1H), 7.80 (d, J = 5.6 Hz, 2H), 7.74 (q, J = 7.0 Hz, 1H), 7.38 (t, J = 6.8 Hz, 1H), 7.24 (q, J = 6.0 Hz, 1H), 6.86 (t, J = 6.7 Hz, 1H), 5.17 (p, J = 7.2 Hz, 1H), 3.54 (d, J = 5.2 Hz, 3H), 1.49 (t, J = 6.4 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 177.27, 166.16, 163.50, 149.12, 137.26, 128.18, 127.79, 126.11, 124.08, 122.46, 120.59, 108.98, 50.76, 40.22, 40.10, 39.98, 39.86, 39.74, 39.62, 39.50, 36.07, 21.45.

(R)-N-(1-(3-chlorophenyl)ethyl)-2-oxoindoline-5-carboxamide (3f).

To a round bottom flask was added 251.1 mg (1.41 mmol) of 2-oxoindoline-5-carboxylic acid dissolved in 7 mL of dry DMF. To this dark red solution were added 0.200 mL (1.55 mmol) of (R)-1-(3-chlorophenyl)ethan-1-amine, 0.25 mL (1.41 mmol) of DIPEA and 717.7 mg (1.91 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 423.3 mg, 91% Molecular Formula: C17H15ClN2O2 ESI-MS calc: 314.08 ESI-MS found: 315.0438 [M+1] HPLC: 5.730 1H NMR (700 MHz, DMSO-d6) δ 10.64 – 10.57 (m, 1H), 8.65 (dd, J = 7.5, 2.2 Hz, 1H), 7.78 – 7.74 (m, 2H), 7.41 (t, J = 2.1 Hz, 1H), 7.33 (dt, J = 6.7, 1.8 Hz, 2H), 7.26 (dp, J = 6.4, 2.2 Hz, 1H), 6.85 (dd, J = 8.1, 2.5 Hz, 1H), 5.15 – 5.06 (m, 1H), 3.53 (s, 2H), 1.45 – 1.42 (m, 3H). 13C NMR (176 MHz, DMSO) δ 176.91, 165.61, 148.09, 146.73, 133.15, 130.39, 127.97, 126.74, 126.14, 125.88, 125.13, 123.78, 108.70, 56.27, 48.40, 40.12, 40.00, 39.88, 39.76, 39.64, 39.52, 39.40, 35.83, 22.43, 18.80.

(R)-2-oxo-N-(1-(p-tolyl)ethyl)indoline-5-carboxamide (3g).

To a round bottom flask was added 244.2 mg (1.41 mmol) of 2-oxoindoline-5-carboxylic acid dissolved in 7 mL of dry DMF. To this dark red solution were added 0.220 mL (1.55 mmol) of (R)-1-(3-chlorophenyl)ethan-1-amine, 0.25 mL (1.41 mmol) of DIPEA and 622.1 mg (1.62 mmol) of HATU. The resultant dark red solution was allowed to stir at room temperature for 12 hours, and then added 200 mL of sat. Na2CO3 and extracted with EtOAc (3 × 100 mL). The combined organic layer was washed with brine (2 × 50 mL) and then dried over MgSO4. Purified by column chromatography (0–15% DCM/MeOH) to give the desired product as a strawberry pink solid. Yield: 300.8 mg, 69% Molecular Formula: C18H18N2O2 ESI-MS calc: 294.14 ESI-MS found: 317 [M+Na] HPLC: 5.556 1H NMR (700 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.57 (d, J = 8.1 Hz, 1H), 7.77 (d, J = 8.1 Hz, 2H), 7.26 (d, J = 7.8 Hz, 2H), 7.11 (d, J = 7.8 Hz, 2H), 6.85 (d, J = 8.0 Hz, 1H), 5.12 (p, J = 7.3 Hz, 1H), 3.53 (s, 2H), 2.26 (s, 3H), 1.44 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 176.63, 165.17, 146.30, 142.10, 135.44, 128.66, 127.64, 127.54, 125.93, 125.51, 123.51, 108.36, 48.03, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 35.57, 22.29, 20.59.

(R)-N-(1-(4-chlorophenyl)ethyl)-2-oxoindoline-5-carboxamide (3h).

Prepared using the protocol described for 3. Yields a strawberry pink solid, 471.0 mg, quantitative yield. Molecular Formula: C17H15ClN2O2 ESI-MS calc: 314.08 ESI-MS found: 337.0717 [M+Na] HPLC: 5.682 1H NMR (500 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.64 (d, J = 7.9 Hz, 1H), 7.76 (d, J = 7.9 Hz, 2H), 7.42 – 7.34 (m, 4H), 6.85 (d, J = 8.1 Hz, 1H), 5.13 (p, J = 7.2 Hz, 1H), 3.53 (s, 2H), 1.45 (d, J = 7.0 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 176.61, 165.29, 146.40, 144.17, 130.98, 128.09, 127.92, 127.67, 127.28, 125.55, 123.49, 108.37, 47.87, 40.00, 39.83, 39.67, 39.50, 39.33, 39.17, 39.00, 35.55, 22.09.

N-benzyl-2-oxoindoline-5-carboxamide (3i).

Prepared with protocol described for 3. Yields a strawberry pink solid, 174.5 mg, 56% Molecular Formula: C16H14N2O2 ESI-MS calc: 266.11 ESI-MS found: 267.2180 [M+1] HPLC: 4.822 1H NMR (700 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.87 (t, J = 6.1 Hz, 1H), 7.79 – 7.75 (m, 2H), 7.34 – 7.28 (m, 5H), 7.23 (dt, J = 7.2, 4.3 Hz, 2H), 6.86 (d, J = 8.0 Hz, 1H), 4.46 (d, J = 5.9 Hz, 2H), 3.53 (s, 2H). 13C NMR (176 MHz, DMSO) δ 176.60, 165.95, 146.42, 139.88, 128.65, 128.19, 128.12, 127.70, 127.59, 127.47, 127.29, 127.11, 126.61, 125.66, 123.45, 108.44, 47.80, 45.00, 42.52, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 38.22, 35.57.

2-oxo-N-(2-phenylpropan-2-yl)indoline-5-carboxamide (3j).

Prepared using the protocol described for 3. Yields a strawberry pink solid, 210.8 mg, 48%. Molecular Formula: C18H18N2O2 ESI-MS calc: 294.14 ESI-MS found: 295.1456 [M+1], 317.1275 [M+Na] HPLC: 5.199 1H NMR (700 MHz, DMSO-d6) δ 10.60 (s, 1H), 8.23 (s, 1H), 7.74 (s, 1H), 7.74 – 7.71 (m, 1H), 7.35 (d, J = 7.8 Hz, 2H), 7.26 (t, J = 7.7 Hz, 2H), 7.15 (t, J = 7.2 Hz, 1H), 6.84 (d, J = 8.1 Hz, 1H), 3.53 (s, 2H), 1.65 (s, 6H). 13C NMR (176 MHz, DMSO) δ 176.64, 165.50, 148.19, 146.15, 128.51, 128.35, 127.82, 127.67, 125.59, 125.37, 124.81, 124.61, 123.65, 108.26, 55.22, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 35.59, 29.66.

(R)-N-(2-methyl-1-phenylpropyl)-2-oxoindoline-5-carboxamide (3k).

Prepared using the protocol described for 3. Yields a strawberry pink solid, 389.9 mg, quantitative yield. Molecular Formula: C19H20N2O2 ESI-MS calc: 308.15 ESI-MS found: 309.2172 HPLC: 5.793 1H NMR (700 MHz, DMSO-d6) δ 10.44 (s, 1H), 8.37 (d, J = 8.9 Hz, 1H), 7.58 (dd, J = 10.7, 2.9 Hz, 2H), 7.22 (d, J = 7.6 Hz, 2H), 7.12 (t, J = 7.5 Hz, 2H), 7.03 (t, J = 7.3 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 4.48 (t, J = 9.2 Hz, 1H), 3.36 (s, 2H), 1.06 (d, J = 6.4 Hz, 2H), 0.83 (d, J = 6.5 Hz, 3H), 0.53 (d, J = 6.7 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 176.69, 165.65, 164.62, 162.32, 146.32, 143.33, 128.02, 127.75, 127.70, 127.41, 126.61, 125.54, 123.53, 108.43, 59.92, 56.08, 41.67, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 38.23, 35.78, 35.62, 32.55, 30.77, 20.09, 19.90, 18.56.

2-oxo-N-(3-phenyloxetan-3-yl)indoline-5-carboxamide (3l).

Prepared using the protocol described for 3. Yields a strawberry pink solid, 62.1 mg, 31% Molecular Formula: C18H16N2O3 ESI-MS calc: 308.12 ESI-MS found: 309.1235 HPLC: 4.601 1H NMR (700 MHz, DMSO-d6) δ 10.49 (d, J = 4.6 Hz, 1H), 9.16 (d, J = 4.6 Hz, 1H), 7.65 – 7.60 (m, 2H), 7.38 – 7.34 (m, 2H), 7.22 – 7.18 (m, 2H), 7.10 (tdd, J = 7.3, 4.9, 2.2 Hz, 1H), 6.74 – 6.70 (m, 1H), 4.82 (dd, J = 6.7, 4.8 Hz, 2H), 4.59 (dd, J = 6.7, 4.8 Hz, 2H), 3.38 (d, J = 4.7 Hz, 2H). 13C NMR (176 MHz, DMSO) δ 176.66, 165.18, 146.78, 143.10, 129.97, 128.28, 128.17, 127.72, 126.91, 126.72, 126.06, 125.80, 125.41, 124.81, 123.53, 120.11, 108.72, 108.56, 81.89, 58.27, 53.49, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 38.23, 35.58, 35.52, 30.66, 14.07.

N-(2-(diethylamino)ethyl)-5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxamide (4a).

To a round bottom flask were added 200.9 mg (1.20 mmol) of 5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxylic acid, 365.8 mg (1.79 mmol) of EDCI, 277.7 mg (1.79 mmol) of HOBt, 7 mL of dry DMF, 0.200 mL (1.44 mmol) of N1,N1-diethylethane-1,2-diamine and 0.34 mL (2.39 mmol) of TEA. The dark red solution was allowed to stir at room temperature for 12 hours before being quenched with water and extracted with DCM (3 × 30 mL). The combined organic layer was washed with brine (2 × 20 mL) and then washed with 10% citric acid (3 × 50 mL), drawing the desired product into the water layer. The aqueous layer was basified with Na2CO3, bringing the pH up to 10, and then extracted with DCM (4 × 50 mL). The combined organic layers were dried over MgSO4 and then evaporated onto silica gel and purified by column chromatography (5–15% MeOH/DCM). The solvent was removed under pressure to give a yellow solid. Result: light yellow solid, 90 mg, 27% Molecular Formula: C14H23N3O2, ESI-MS Calc: 265.18 ESI-MS found: 266.1749 HPLC: 2.681 1H NMR (400 MHz, DMSO-d6) δ 11.85 (s, 1H), 9.54 (s, 1H), 7.95 (s, 2H), 2.34 (d, J = 20.8 Hz, 7H), 0.99 (s, 6H).

5-formyl-2,4-dimethyl-N-(prop-2-yn-1-yl)-1H-pyrrole-3-carboxamide (4b).

To a round bottom flask were added 199.4 mg (1.20 mmol) of 5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxylic acid, 688.0 mg (1.79 mmol) of HATU, 6 mL of dry DMF, 0.100 mL (1.44 mmol) of propargylamine and 0.34 mL (2.39 mmol) of TEA. The dark red solution was allowed to stir at room temperature for 12 hours before being quenched with water and extracted with DCM (3 × 30 mL). Took the combined organic layers and washed with brine (2 × 20 mL) and then washed with 10% citric acid (3 × 50 mL), taking the desired product into the aqueous layer. The aqueous layer was basified with Na2CO3, bringing the pH up to 10, and then extracted with DCM (4 × 50 mL). The combined organic layer was dried over MgSO4 and then removed solvent to give the final product as a light orange oil. Result: light orange oil, 61 mg, 25% Molecular Formula: C11H12N2O2 ESI-MS calc: 204.09 ESI-MS found: 205.0873 [M+1], 242.1167 [M+K] HPLC: 3.625 1H NMR (700 MHz, DMSO-d6) δ 11.87 (s, 1H), 9.55 (s, 1H), 7.95 (s, 16H), 3.97 (ddd, J = 12.7, 5.7, 2.5 Hz, 3H), 3.08 (t, J = 2.4 Hz, 1H), 2.36 (s, 3H), 2.31 (s, 3H). 13C NMR (176 MHz, DMSO) δ 177.82, 164.77, 139.11, 138.51, 128.26, 119.20, 89.92, 82.16, 72.95, 55.38, 40.24, 38.71, 28.42, 12.90, 10.00.

N-allyl-5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxamide (4c).

To a round bottom flask were added 204.4 mg (1.20 mmol) of 5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxylic acid, 350.3 mg of EDCI (1.79 mmol), 275.3 mg (1.79 mmol) of HOBt, 6 mL of dry DMF, 0.110 mL (1.44 mmol) of allylamine and 0.34 mL (2.39 mmol) of TEA. The dark red solution was allowed to stir at room temperature for 12 hours before being quenched with water and extracted with DCM (3 × 30 mL). Took combined organic layers and washed with LiCl 3 × 20 mL) and then washed with 10% Citric acid (3 × 50 mL), drawing the desired product into the aqueous layer. The aqueous layer was basified with Na2CO3, bringing the pH up to 8, and then extracted with DCM (4 × 50 mL) The combined organic layer was dried over MgSO4 and then removed solvent to give a yellow solid. Result: light yellow solid, 64 mg, 25% Molecular Formula: C11H14N2O2 ESI-MS calc: 206.11 ESI-MS: 246.1621 [M+ MeCN] HPLC: 3.885 1H NMR (500 MHz, DMSO-d6) δ 11.38 (s, 1H), 9.54 (s, 1H),8.17 (s, 1H), 6.01 (tt, J = 10.8, 5.4 Hz, 1H), 5.87 (ddt, J = 16.4, 10.6, 5.3 Hz, 2H), 5.18 (t, J = 15.7 Hz, 3H), 5.08 (dd, J = 16.5, 10.2 Hz, 3H), 4.14 (d, J = 5.5 Hz, 2H), 3.82 (t, J = 5.7 Hz, 3H), 2.31 (s, 5H), 2.23 (s, 3H). 13C NMR (126 MHz, DMSO) δ 161.78, 150.29, 135.45, 135.21, 115.01, 114.35, 114.20, 40.54, 40.50, 39.50, 39.33, 39.17, 39.00, 38.83, 38.67, 38.50, 35.26, 30.25, 11.95, 9.27.

N-(but-3-yn-1-yl)-5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxamide (4d).

To a round bottom flask were added 199.6 mg (1.20 mmol) of 5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxylic acid, 684.3 mg (1.79 mmol) of HATU, 6 mL of dry DMF, 0.100 mL (1.44 mmol) of 1-amino-3-butyne and 0.30 mL (2.39 mmol) of DIPEA. The dark red solution was allowed to stir at room temperature for 12 hours before being quenched with water and extracted with DCM (3 × 30 mL). The combined organic layers were washed with LiCl (2 × 20 mL) and then washed with 10% citric acid (3 × 50 mL), drawing the desired product into the water layer. The aqueous layer was basified with Na2CO3, bringing the pH up to 10, and then extracted with DCM (4 × 50 mL). The combined organic layer was dried over MgSO4 and then removed solvent under pressure to give the final product as a yellow solid. Result: light yellow solid, 99.6 mg, 38.1% Molecular Formula: C12H14N2O2 ESI-MS calc: 218.11 ESI-MS found: 219.1129 HPLC: 3.997 1H NMR (700 MHz, DMSO-d6) δ 11.83 (s, 1H), 2.83 (s, 1H), 2.69 (d, J = 1.0 Hz, 1H), 2.60 (d, J = 2.4 Hz, 3H), 2.37 (s, 4H), 2.32 (s, 3H). 13C NMR (176 MHz, DMSO) δ 178.94, 164.54, 160.23, 152.56, 147.04, 146.58, 126.55, 115.31, 88.48, 82.43, 72.02, 55.77, 37.85, 18.85, 13.97, 10.55.

N-(but-3-en-1-yl)-5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxamide (4e).

To a round bottom flask were added 197.8 mg (1.20 mmol) of 5-formyl-2,4-dimethyl-1H-pyrrole-3-carboxylic acid, 685.8 mg (1.79 mmol) of HATU, 6 mL of dry DMF, 107.1 mg (1.44 mmol) of but-3-en-1-amine and 0.45 mL (2.40 mmol) of DIPEA. The dark red solution was allowed to stir at room temperature for 12 hours before being quenched with water and extracted with DCM (3 × 30 mL). The combined organic layers were washed with brine (2 × 20 mL) and then washed with 10% citric acid (3 × 50 mL), drawing the desired material in to the aqueous layer. The aqueous layer was basified with Na2CO3, bringing the pH up to 10, and then extracted with DCM (4 × 50 mL). The combined organic layer was dried over MgSO4. Result: light orange oil, 106.1 mg, 37.2% Molecular Formula: C12H16N2O2 ESI-MS calc: 220.12 MS: 221.1304 HPLC: 5.554 1H NMR (500 MHz, DMSO-d6) δ 9.77 (s, 1H), 8.84 (d, J = 4.5 Hz, 1H), 8.73 (d, J = 8.4 Hz, 1H), 7.95 (s, 4H), 7.66 (dd, J = 8.7, 4.6 Hz, 1H), 5.12 – 4.99 (m, 1H), 2.59 (s, 7H). 13C NMR (126 MHz, DMSO) δ 179.37, 165.02, 162.73, 152.99, 146.27, 140.66, 134.88, 130.23, 122.09, 36.21, 31.20, 14.39, 10.98.

(R,Z)-3-((4-((2-(diethylamino)ethyl)carbamoyl)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (5a).

To a dried sealed tube were added 97.9 mg (0.371 mmol) of (3), 107.0 mg (0.357 mmol) of (4a) all of which were dissolved in abs. EtOH (3.5 mL). To this solution were added 2 drops of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as an orange solid. Yield: orange solid, 38.3 mg, 22% Molecular Formula: C31H37N5O3 ESI-MS calc: 527.29 ESI-MS found: 528.2955 HPLC: 5.621 1HNMR (500 MHz, DMSO-d6) δ 13.59 (s, 1H), 11.13 (s, 1H), 8.59 (d, J = 8.1 Hz, 1H), 8.25 (s, 1H), 7.71 (d, J = 6.8 Hz, 2H), 7.44 (d, J = 6.0 Hz, 1H), 7.41 (d, J = 7.9 Hz, 2H), 7.33 (t, J = 7.7 Hz, 2H), 7.22 (t, J = 7.7 Hz, 1H), 6.93 (d, J = 8.3 Hz, 1H), 5.19 (t, J = 7.5 Hz, 1H), 3.28 (d, J = 7.0 Hz, 2H), 2.44 (d, J = 6.7 Hz, 6H), 1.51 (d, J = 7.1 Hz, 3H), 0.97 (t, J = 7.1 Hz, 7H). 13CNMR (176 MHz, DMSO) δ 169.90, 165.95, 164.71, 145.11, 140.64, 136.48, 129.93, 128.30, 127.78, 126.64, 126.37, 126.18, 125.86, 125.28, 124.09, 120.76, 117.69, 114.37, 108.94, 51.72, 48.55, 46.59, 45.44, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 37.06, 22.35, 13.40, 11.92, 10.79.

(S,Z)-3-((4-((2-(diethylamino)ethyl)carbamoyl)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (5b).

To a dried sealed tube were added 91.2 mg (0.325 mmol) of (3a), 79.9 mg (0.299 mmol) of (4a) all of which were dissolved in abs. EtOH (2.5 mL). To this solution were added 2 drops of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as an orange solid. Yield: orange solid, 38.3 mg, 22% Molecular Formula: C31H37N5O3 ESI-MS calc: 527.29 ESI MS found: 528.2043 HPLC: 5.753 1H NMR (500 MHz, DMSO-d6) δ 13.59 (s, 1H), 11.16 (s, 1H), 8.67 (s, 1H), 8.32 (d, J = 5.5 Hz, 1H), 7.75 – 7.68 (m, 2H), 7.48 (s, 1H), 7.42 (d, J = 7.7 Hz, 3H), 7.32 (t, J = 7.6 Hz, 3H), 7.22 (t, J = 7.3 Hz, 1H), 6.94 (d, J = 8.1 Hz, 1H), 5.19 (p, J = 7.2 Hz, 1H), 2.45 (d, J = 3.4 Hz, 8H), 2.37 (s, 1H), 2.32 (s, 1H), 2.08 (s, 1H), 1.51 (d, J = 7.1 Hz, 4H), 0.99 (t, J = 7.5 Hz, 8H). 13C NMR (126 MHz, DMSO) δ 170.28, 147.82, 141.01, 130.32, 128.66, 127.23, 126.98, 126.61, 126.26, 125.66, 124.60, 118.30, 106.84, 102.36, 48.92, 47.02, 22.80, 17.46, 13.83, 11.24, 9.18.

(R,Z)-3-((3,5-dimethyl-4-(prop-2-yn-1-ylcarbamoyl)-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (5c).

To a dried sealed tube were added 75.1 mg (0.27 mmol) of (3), 50 mg (0.28 mmol) of (4b) all of which were dissolved in abs. EtOH (1.8 mL). To this solution were added 0.05 mL of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and filtered off an orange solid. The solid was rinsed with cold EtOH to give the final product. Yield: bright orange solid, 58.6 mg, 46% Molecular Formula: C28H26N4O3 ESI-MS calc: 466.20 ESI-MS found: 467.2037 HPLC: 6.360 1H NMR (700 MHz, DMSO-d6) δ 13.61 (s, 1H), 11.15 (s, 1H), 8.61 (d, J = 8.0 Hz, 1H), 8.26 (s, 1H), 8.06 (t, J = 5.5 Hz, 1H), 7.71 (d, J = 9.0 Hz, 2H), 7.41 (d, J = 7.8 Hz, 2H), 7.33 (q, J = 5.8, 3.9 Hz, 2H), 7.23 (t, J = 7.2 Hz, 1H), 6.94 (d, J = 8.2 Hz, 1H), 5.20 (t, J = 7.7 Hz, 1H), 4.02 (d, J = 5.5 Hz, 2H), 3.14 – 3.08 (m, 1H), 2.44 (d, J = 9.2 Hz, 7H), 1.51 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 170.30, 166.26, 145.52, 141.06, 136.92, 130.46, 128.67, 128.29, 127.00, 126.82, 126.58, 126.28, 124.49, 120.45, 118.23, 115.03, 109.28, 82.17, 73.04, 48.87, 28.49, 22.77, 13.76, 11.13.

(R,Z)-3-((4-(allylcarbamoyl)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (5d).

To a dried sealed tube were added 53.6 mg (0.27 mmol) of (3), 50 mg (0.28 mmol) of (4c) all of which were dissolved in abs. EtOH (3 mL). To this solution were added 2 drops (0.05 mL) of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, the reaction was cooled to room temperature and then an orange solid was filtered off. The solid was washed with cold EtOH to give the final product. Result: orange solid, 79.5 mg, 63% Molecular Formula: C28H28N4O3 ESI-MS calc: 468.22 ESI-MS found: 469.2224 HPLC: 6.450 1H NMR (700 MHz, DMSO-d6) δ 13.60 (s, 1H), 11.14 (s, 1H), 8.60 (d, J = 8.0 Hz, 1H), 8.25 (d, J = 1.6 Hz, 1H), 7.83 (t, J = 5.8 Hz, 1H), 7.73 – 7.68 (m, 2H), 7.41 (d, J = 7.6 Hz, 2H), 7.33 (t, J = 7.6 Hz, 2H), 7.22 (t, J = 7.3 Hz, 1H), 6.94 (d, J = 8.1 Hz, 1H), 5.90 (ddt, J = 15.7, 10.3, 5.2 Hz, 1H), 5.22 – 5.18 (m, 2H), 5.10 (dd, J = 10.3, 1.8 Hz, 1H), 3.87 (t, J = 5.6 Hz, 2H), 2.44 (d, J = 7.8 Hz, 6H), 1.51 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 170.50, 145.29, 135.97, 130.84, 129.65, 128.44, 128.02, 126.34, 115.20, 108.42, 103.80, 91.16, 59.93, 22.53, 13.55, 10.93.

(R,Z)-3-((4-(but-3-yn-1-ylcarbamoyl)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (5e).

To a dried sealed tube were added 99.9 mg (0.357 mmol) of (3), 82 mg of (4d) all of which were dissolved in abs. EtOH (3.5 mL). To this solution were added 2 drops (0.05 mL) of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, the solution was cooled to room temperature and then an orange solid was filtered off. The solid was washed with cold EtOH to give the final product. Yield: orange solid, 19 mg, 10% Molecular Formula: C29H28N4O3 ESI-MS calc: 480.22 ESI-MS found: 481.2226 HPLC: 6.444 1H NMR (700 MHz, DMSO-d6) δ 13.60 (s, 1H), 11.15 (s, 1H), 8.62 (d, J = 8.0 Hz, 1H), 8.25 (s, 1H), 7.79 (t, J = 5.8 Hz, 1H), 7.71 (d, J = 6.3 Hz, 2H), 7.41 (d, J = 7.6 Hz, 2H), 7.33 (t, J = 7.6 Hz, 2H), 7.23 (t, J = 7.3 Hz, 1H), 6.94 (d, J = 8.2 Hz, 1H), 5.19 (p, J = 7.3 Hz, 1H), 2.86 (t, J = 2.5 Hz, 1H), 2.45 (d, J = 12.0 Hz, 6H), 2.42 (td, J = 7.1, 2.4 Hz, 2H), 1.51 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.83, 165.81, 164.71, 145.08, 140.56, 136.34, 130.00, 128.23, 127.80, 126.55, 126.12, 125.79, 125.18, 124.08, 120.61, 117.76, 82.50, 72.12, 48.42, 37.95, 22.33, 13.33, 10.70.

(R,Z)-3-((4-(but-3-en-1-ylcarbamoyl)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (5f).

To a dried sealed tube were added 94.8 mg (0.27 mmol) of (3), 87.2 mg (0.28 mmol) of (4e) all of which were dissolved in abs. EtOH (2.2 mL). To this solution were added 2 drops (0.07 mL) of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, solution was cooled to room temperature and then purified by column chromatography. Fractions were collected and solvent was removed under pressure to give the desired product, as an orange solid. Yield: orange solid, 103 mg, 59% Molecular Formula: C29H30N4O3 ESI-MS calc: 482.23 ESI-MS found: 483.2382 [M+1] HPLC: 6.720 1H NMR (700 MHz, DMSO-d6) δ 13.54 (s, 1H), 11.13 (s, 1H), 8.61 (d, J = 8.2 Hz, 2H), 8.27 – 8.18 (m, 2H), 7.73 – 7.71 (m, 1H), 7.68 (s, 1H), 7.42 (d, J = 8.3 Hz, 3H), 7.35 – 7.31 (m, 4H), 7.25 – 7.21 (m, 2H), 6.94 (d, J = 8.1 Hz, 1H), 5.21 (p, J = 7.3 Hz, 2H), 2.43 (d, J = 9.0 Hz, 2H), 2.29 (d, J = 11.4 Hz, 7H), 1.63 – 1.59 (m, 3H), 1.51 (d, J = 7.1 Hz, 6H). 13C NMR (176 MHz, DMSO) δ 169.79, 165.81, 165.04, 145.07, 140.57, 133.90, 128.74, 128.19, 127.78, 126.52, 126.11, 125.22, 123.96, 120.71, 117.70, 116.21, 114.09, 108.74, 48.41, 35.77, 33.74, 22.26, 12.40, 10.15.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2oxo-N-(1-phenylethyl)indoline-5-carboxamide (7a).

To a dried sealed tube were added 91.2 mg (0.892 mmol) of (3), 79.9 mg (1.39 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (2.5 mL). To this solution were added 2 drops (0.05 mL) of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, the reaction was cooled to room temperature and an orange solid was filtered off. The solid was washed with cold EtOH to give the desired product. Result: orange solid, 269.3 mg, 69% Molecular Formula: C24H22N4O4 ESI-MS calc: 430.16 ESI-MS found: 431.1715 HPLC: 7.400 1H NMR (700 MHz, DMSO-d6) δ 8.63 (d, J = 8.0 Hz, 1H), 8.36 (d, J = 1.6 Hz, 1H), 7.83 (s, 1H), 7.76 (dd, J = 8.1, 1.7 Hz, 1H), 7.41 (d, J = 7.5 Hz, 2H), 7.33 (t, J = 7.6 Hz, 2H), 7.22 (t, J = 7.3 Hz, 1H), 6.97 (d, J = 8.1 Hz, 1H), 5.19 (p, J = 7.2 Hz, 1H), 2.08 (d, J = 1.0 Hz, 1H), 1.50 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.90, 165.69, 144.98, 142.47, 136.80, 132.12, 130.48, 129.11, 128.20, 127.74, 126.10, 125.56, 123.33, 121.76, 115.38, 53.66, 22.28, 18.23, 10.50.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(m-tolyl)ethyl)indoline-5-carboxamide (7b).

To a dried sealed tube were added 200 mg (0.679 mmol) of (3b), 139.6 mg (0.815 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (6.0 mL). To this solution were added 0.05 ml of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as a lemon yellow solid. Yield: lemon yellow solid, 100 mg, 29.8 % Molecular Formula: C25H24N4O4 ESI-MS calc: 444.18 ESI-MS found: 445.1862 [M+1] HPLC: 7.732 1H NMR (700 MHz, DMSO-d6) δ 11.34 (s, 1H), 8.58 (d, J = 8.0 Hz, 1H), 8.33 (d, J = 1.6 Hz, 1H), 7.78 (s, 1H), 7.76 (dd, J = 8.2, 1.6 Hz, 1H), 7.23 – 7.18 (m, 3H), 7.03 (d, J = 6.1 Hz, 1H), 6.94 (d, J = 8.1 Hz, 1H), 5.16 (p, J = 7.2 Hz, 1H), 2.62 (s, 3H), 2.57 (s, 3H), 2.30 (s, 3H), 1.49 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.83, 165.49, 144.90, 141.26, 137.14, 136.75, 133.66, 128.34, 128.09, 127.51, 127.15, 126.74, 125.07, 124.71, 124.39, 123.59, 123.17, 119.39, 118.96, 109.09, 48.36, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 22.26, 21.11, 14.76, 11.21.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-N-(1-(4-fluorophenyl)ethyl)-2-oxoindoline-5-carboxamide (7c).

To a dried sealed tube were added 128 mg (0.429 mmol) of (3c), 101.3 mg (0.60 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (3.0 mL). To this solution were added 0.05 ml of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as an orange solid. Yield: orange solid, 96.3 mg, 50 % Molecular Formula: C24H21FN4O4 ESI-MS calc: 448.15 ESI-MS found: 449.1619 [M+1] HPLC: 7.422 1H NMR (700 MHz, DMSO-d6) δ 11.37 (s, 1H), 8.64 (d, J = 7.8 Hz, 1H), 8.35 (d, J = 1.7 Hz, 1H), 7.83 (s, 1H), 7.76 (dd, J = 8.1, 1.7 Hz, 1H), 7.47 – 7.44 (m, 2H), 7.18 – 7.14 (m, 2H), 6.97 (d, J = 8.1 Hz, 1H), 5.19 (p, J = 7.2 Hz, 1H), 2.65 (s, 3H), 2.60 (s, 3H), 1.51 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.85, 165.62, 136.79, 133.69, 128.31, 128.02, 127.97, 124.71, 123.71, 119.03, 114.88, 114.76, 50.22, 47.86, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 22.26, 14.78, 11.22.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(pyridin-4-yl)ethyl)indoline-5-carboxamide (7d).

To a dried sealed tube were added 100 mg (0.455 mmol) of (3d), 77.1 mg (0.459 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (3.0 mL). To this solution were added 0.05 ml of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as a yellow solid. Yield: yellow solid, 48.0 mg, 31.3 % Molecular Formula: C23H21N5O4 ESI-MS calc: 431.16 ESI-MS found: 432.1656 [M+1] HPLC: 5.496 1H NMR (700 MHz, DMSO-d6) δ 11.36 (s, 1H), 8.73 (d, J = 7.6 Hz, 1H), 8.52 – 8.50 (m, 2H), 8.36 (d, J = 1.6 Hz, 1H), 7.82 (s, 1H), 7.77 (dd, J = 8.1, 1.6 Hz, 1H), 7.40 (d, J = 5.2 Hz, 2H), 6.97 (d, J = 8.0 Hz, 1H), 5.15 (p, J = 7.3 Hz, 1H), 2.64 (s, 3H), 2.59 (s, 3H), 1.51 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 170.11, 166.24, 153.97, 149.80, 137.07, 135.63, 128.31, 127.80, 125.45, 124.98, 124.02, 121.55, 119.37, 109.42, 48.18, 40.12, 40.00, 39.88, 39.76, 39.64, 39.52, 39.40, 21.78, 15.04, 11.48.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2oxo-N-(1-(pyridin-2-yl)ethyl)indoline-5-carboxamide (7e).

To a dried sealed tube were added 165 mg (0.587 mmol) of (3e), 122.8 mg (0.762 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (3.0 mL). To this solution were added 0.05 ml of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as a yellow solid. Yield: yellow solid, 116 mg, 34 % Molecular Formula: C23H21N5O4 ESI-MS calc: 431.16 ESI-MS found: 432.0558 [M+1] HPLC: 5.49 1H NMR (700 MHz, DMSO-d6) δ 11.36 (d, J = 7.2 Hz, 1H), 8.65 – 8.58 (m, 1H), 8.55 – 8.47 (m, 1H), 8.39 (t, J = 7.7 Hz, 1H), 7.78 (ddd, J = 33.0, 15.8, 7.2 Hz, 3H), 7.42 (q, J = 10.5, 9.0 Hz, 1H), 7.25 (dd, J = 12.7, 6.8 Hz, 1H), 6.95 (d, J = 7.0 Hz, 1H), 5.22 (q, J = 8.8, 8.0 Hz, 1H), 2.66 – 2.60 (m, 3H), 2.59 (s, 3H), 1.56 – 1.49 (m, 3H). 13C NMR (176 MHz, DMSO) δ 169.21, 165.90, 148.79, 136.83, 128.23, 124.86, 123.81, 120.44, 119.09, 109.32, 58.91, 50.44, 40.00, 39.88, 39.76, 39.74, 39.64, 39.62, 39.52, 39.40, 39.28, 21.09, 11.38.

(R,Z)-N-(1-(3-chlorophenyl)ethyl)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxoindoline-5-carboxamide (7f).

To a dried sealed tube were added 242.2 mg (0.769 mmol) of (3f), 150.1 mg (0.892 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (6.0 mL). To this solution were added 0.05 ml of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as an orange solid. Yield: orange solid, 46 mg, 16% Molecular Formula: C24H21ClN4O4 ESI-MS calc: 464.13 ESI-MS found: 465.1318 HPLC: 7.699 1H NMR (700 MHz, DMSO-d6) δ 11.38 (s, 1H), 8.68 (d, J = 7.8 Hz, 1H), 8.36 (d, J = 1.6 Hz, 1H), 7.84 (s, 1H), 7.76 (dd, J = 8.2, 1.7 Hz, 1H), 7.46 (d, J = 2.1 Hz, 1H), 7.37 (d, J = 6.6 Hz, 2H), 7.29 (dt, J = 6.7, 2.3 Hz, 1H), 6.97 (d, J = 8.1 Hz, 1H), 5.17 (p, J = 7.2 Hz, 1H), 2.65 (s, 3H), 2.60 (s, 3H), 1.50 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 165.88, 147.85, 144.31, 138.20, 130.27, 126.64, 126.10, 125.34, 125.04, 123.98, 119.28, 109.31, 102.54, 48.41, 40.00, 39.88, 39.76, 39.64, 39.52, 39.40, 39.28, 22.27, 14.93.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(p-tolyl)ethyl)indoline-5-carboxamide (7g).

To a dried sealed tube were added 253.9 mg (0.862 mmol) of (3g), 140.9 mg (0.837 mmol) of 3,5-dimethyl-4-nitro-1H-pyrrole-2-carbaldehyde, all of which were dissolved in abs. EtOH (3.0 mL). To this solution was added 0.05 ml of piperidine and heated to reflux (95 °C) for 4 hours. Once complete, cooled to room temperature and then filtered off the product as an orange solid. Yield: orange solid, 76.1 mg, 25% Molecular Formula: C25H24N4O4 ESI-MS calc: 444.18 ESI-MS found: 445.1864 [M+1] HPLC: 7.662 1H NMR (700 MHz, DMSO-d6) δ 9.73 (s, 1H), 8.57 (d, J = 8.2 Hz, 2H), 8.35 (d, J = 1.7 Hz, 1H), 7.83 (s, 1H), 7.76 (dd, J = 8.1, 1.7 Hz, 2H), 7.30 (d, J = 7.8 Hz, 3H), 7.14 (d, J = 7.9 Hz, 3H), 6.96 (d, J = 8.0 Hz, 2H), 5.17 (p, J = 7.2 Hz, 1H), 2.55 (d, J = 8.7 Hz, 6H), 2.28 (s, 5H). 13C NMR (176 MHz, DMSO) δ 170.11, 165.77, 155.70, 142.17, 141.51, 137.04, 135.76, 135.76, 128.94, 126.27, 125.36, 119.23, 109.39, 52.91, 40.12, 40.00, 39.88, 39.76, 39.64, 39.52, 39.40, 22.49, 20.86, 15.04, 11.49, 10.19.

(Z)-N-benzyl-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxoindoline-5-carboxamide (7i).

Prepared using the protocol described for 7a. Yields an orange solid, 150 mg, 55% Molecular Formula: C23H20N4O4 ESI-MS calc: 416.15 ESI-MS found: 417.2934, HPLC: 7.158, 1H NMR (700 MHz, DMSO-d6) δ 11.44 – 11.06 (m, 1H), 8.84 (t, J = 6.1 Hz, 1H), 8.34 (s, 1H), 7.76 (d, J = 8.3 Hz, 1H), 7.69 (s, 1H), 7.34 (d, J = 5.6 Hz, 4H), 7.25 (d, J = 6.6 Hz, 1H), 6.92 (d, J = 8.2 Hz, 1H), 4.52 (d, J = 5.7 Hz, 2H). 13C NMR (176 MHz, DMSO) δ 169.75, 166.10, 141.33, 139.82, 136.69, 133.57, 128.23, 127.89, 127.37, 127.18, 126.67, 124.95, 124.69, 124.44, 123.25, 119.29, 118.57, 109.16, 42.62, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 14.73, 11.11.

(Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxo-N-(2-phenylpropan-2-yl)indoline-5-carboxamide (7j).

Prepared using the protocol described for 7a. Yields an orange solid, 76.1 mg, 32.1%. Molecular Formula: C25H24N4O4 ESI-MS calc: 444.18 ESI-MS found:445.2680 [M+1] HPLC: 7.713 1H NMR (700 MHz, DMSO-d6) δ 11.32 (s, 1H), 8.31 (d, J = 1.7 Hz, 1H), 8.25 (s, 1H), 7.80 (s, 1H), 7.72 (dd, J = 8.0, 1.7 Hz, 1H), 7.42 – 7.39 (m, 2H), 7.28 (t, J = 7.8 Hz, 2H), 7.17 (t, J = 7.3 Hz, 1H), 6.92 (d, J = 8.1 Hz, 1H), 2.61 (s, 3H), 2.57 (s, 3H), 1.70 (s, 6H). 13C NMR (176 MHz, DMSO) δ 169.84, 165.70, 148.16, 141.11, 136.71, 133.63, 129.19, 127.83, 127.58, 125.62, 125.05, 124.73, 124.69, 124.29, 123.61, 119.44, 119.02, 108.96, 55.31, 39.86, 39.74, 39.62, 39.50, 39.38, 39.26, 39.14, 29.65, 14.76, 11.21.

(R,Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-N-(2-methyl-1-phenylpropyl)-2-oxoindoline-5-carboxamide (7k).

Prepared using the protocol described for 7a. Yields an orange solid, 144.6 mg, 64.8% Molecular Formula: C26H26N4O4 ESI-MS calc: 458.20 ESI-MS found: 459.2109 HPLC: 7.896 1H NMR (700 MHz, DMSO-d6) δ 11.32 (s, 1H), 8.56 (p, J = 9.3, 8.3 Hz, 1H), 8.29 (dq, J = 13.4, 7.5 Hz, 1H), 7.88 – 7.63 (m, 2H), 7.48 – 7.28 (m, 4H), 7.22 (qd, J = 14.5, 9.4, 7.5 Hz, 1H), 6.94 (tt, J = 14.2, 7.5 Hz, 1H), 4.79 – 4.64 (m, 1H), 2.64 – 2.58 (m, 3H), 2.58 (s, 3H), 1.05 (tt, J = 13.4, 8.0 Hz, 3H), 0.75 (dp, J = 25.8, 6.7 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.69, 165.86, 143.12, 141.05, 127.89, 127.29, 127.17, 126.47, 125.00, 124.61, 124.32, 123.62, 119.24, 119.05, 108.85, 59.80, 39.74, 39.62, 39.50, 39.38, 39.26, 39.15, 39.03, 32.40, 19.97, 19.82, 14.64, 11.05.

(Z)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxo-N-(3-phenyloxetan-3-yl)indoline-5-carboxamide (7l).

Prepared using the protocol described for 7a. Yields an orange solid, 47 mg, 42%. Molecular Formula: C25H22N4O5 ESI-MS calc: 458.16 ESI-MS found: 459.1656 HPLC: 6.923, 1H NMR (700 MHz, DMSO-d6) δ 11.39 (s, 1H), 9.36 (s, 1H), 8.40 (d, J = 2.0 Hz, 1H), 7.83 (d, J = 2.2 Hz, 1H), 7.81 – 7.77 (m, 1H), 7.59 – 7.55 (m, 2H), 7.40 (t, J = 7.8 Hz, 2H), 7.29 (t, J = 7.4 Hz, 1H), 6.99 (dd, J = 8.4, 2.0 Hz, 1H), 5.04 (d, J = 6.7 Hz, 2H), 4.81 (d, J = 6.7 Hz, 2H), 2.63 (d, J = 2.0 Hz, 3H), 2.58 (d, J = 2.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.85, 165.50, 142.99, 141.58, 136.84, 133.69, 127.65, 127.44, 126.92, 125.26, 124.89, 124.72, 124.58, 123.78, 119.24, 109.26, 81.88, 58.33, 14.77, 11.15.

(R,Z)-N-(1-(4-chlorophenyl)ethyl)-3-((3,5-dimethyl-4-nitro-1H-pyrrol-2-yl)methylene)-2-oxoindoline-5-carboxamide (7k).

Prepared using the protocol described for 7a. Yields an orange solid, 213.3 mg, 96%. Molecular Formula: C24H21ClN4O4 ESI-MS calc: 464.13 ESI-MS found: 465.1318 [M+1] HPLC: 7.795 1H NMR (500 MHz, DMSO-d6) δ 11.34 (s, 1H), 8.65 (d, J = 7.8 Hz, 1H), 8.35 – 8.30 (m, 1H), 7.79 (s, 1H), 7.75 (dd, J = 8.1, 1.7 Hz, 1H), 7.43 (d, J = 8.3 Hz, 2H), 7.39 (d, J = 8.5 Hz, 3H), 6.94 (d, J = 8.1 Hz, 1H), 5.17 (p, J = 7.2 Hz, 1H), 2.62 (s, 3H), 2.57 (s, 3H), 1.49 (d, J = 7.1 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 169.83, 165.67, 144.05, 141.32, 136.76, 133.66, 131.02, 128.18, 128.10, 128.00, 127.49, 125.11, 124.70, 124.41, 123.63, 119.35, 118.98, 109.10, 48.00, 40.00, 39.83, 39.67, 39.50, 39.34, 39.17, 39.00, 22.07, 14.76, 11.21.

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)-methylene)2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (8a).

To a flask were added 90.7 mg of (7a) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 226.8 mg (14 equivalents) of Zn powder and 2 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then add EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. Because the free amine is very reactive, it was moved forward without further characterization. Result: orange solid, 66.7 mg, 79.0% Molecular Formula: C24H24N4O2 ESI-MS calc: 400.19 ESI-MS found: 401.1955 HPLC: 5.177 1H NMR (700 MHz, DMSO-d6) δ 13.45 (s, 1H), 10.88 (s, 1H), 8.56 (d, J = 8.0 Hz, 1H), 8.12 (d, J = 1.6 Hz, 1H), 7.62 (dd, J = 8.1, 1.6 Hz, 1H), 7.46 (s, 1H), 7.41 (d, J = 7.6 Hz, 3H), 7.33 (t, J = 7.6 Hz, 3H), 7.22 (t, J = 7.3 Hz, 1H), 6.89 (d, J = 8.1 Hz, 1H), 5.20 (q, J = 7.4 Hz, 1H), 4.02 (d, J = 14.5 Hz, 2H), 2.26 (s, 3H), 2.17 (s, 3H), 1.51 (d, J = 7.1 Hz, 3H).

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(m-tolyl)ethyl)indoline-5-carboxamide (8b).

To a flask were added 86.4 mg of (7b) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 210.9 mg (14 equivalents) of Zn powder and 2 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then added EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. *Note: the free amine is very reactive, so it was moved forward without further characterization* Result: red solid, 86.6 mg, 94% Molecular Formula: C25H26N4O2 ESI-MS calc: 414.21 ESI-MS found: 415.2120 [M+1] HPLC: 5.627

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(4-fluorophenyl)ethyl)-2-oxoindoline-5-carboxamide (8c).

To a flask were added 96.3 mg of (7c) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 295 mg (14 equivalents) of Zn powder and 2 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then added EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. The free amine is very reactive, so it was moved forward without further characterization. Result: red solid, 96 mg, 100% Molecular Formula: C24H23FN4O2 ESI-MS calc: 418.18 ESI-MS found: 419.1938 [M+1] HPLC: 5.713

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(pyridin-4-yl)ethyl)indoline-5-carboxamide (8d).

To a flask were added 48.0 mg of (7d) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 208 mg (14 equivalents) of Zn powder and 2 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then add EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. The free amine is very reactive, so it was moved forward without further characterization. Result: red solid, 41.5 mg, 89% Molecular Formula: C23H23N5O2 ESI-MS calc: 401.19 ESI-MS found: 402.1920 [M+1] HPLC: 3.928

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(pyridin-2-yl)ethyl)indoline-5-carboxamide (8e).

To a flask were added 73.9 mg of (7e) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 289 mg (14 equivalents) of Zn powder and 2 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then added EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. The free amine is very reactive, so it was moved forward without further characterization. Result: red solid, 73.8 mg, 100% Molecular Formula: C23H23N5O2 ESI-MS calc: 401.19 ESI-MS found: 402.1450 [M+1] HPLC: 3.920

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(3-chlorophenyl)ethyl)-2-oxoindoline-5-carboxamide (8f).

To a flask were added 46.0 mg of (7f) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 170.1 mg (14 equivalents) of Zn powder and 1.4 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then add EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. The free amine is very reactive, so it was moved forward without further characterization. Result: red solid, 40 mg, 95% Molecular Formula: C24H23ClN4O2 ESI-MS calc: 434.15 ESI-MS found: 435.1569 [M+1] HPLC: 5.556

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(p-tolyl)ethyl)indoline-5-carboxamide (8g).

To a flask were added 76.1 mg of (7g) and 5 mL of 2:1 EtOH/EtOAc. To this slurry were added 314.4 mg (14 equivalents) of Zn powder and 2.0 mL (150 equivalents) of AcOH. The turbid orange solution was allowed to stir at 50 °C for 2 hours. Once complete, the reaction was cooled to room temperature and then added EtOAc before basifying with sat. Na2CO3. The basified aqueous layer was extracted with EtOAc (3 × 30 mL) and then washed with water and brine (1 × 30 mL), respectively. The organic layer was dried over MgSO4 and then solvent was removed under pressure to give the desired product as a red solid. The free amine is very reactive, so it was moved forward without further characterization. Result: red solid, 220 mg, 100% Molecular Formula: C25H26N4O2 ESI-MS calc: 414.21 ESI-MS found: 415.2119 [M+1] HPLC: 5.573

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(4-chlorophenyl)ethyl)-2-oxoindoline-5-carboxamide (8h).

Synthesized using protocol described for 8a. Yields a red solid, 36.1 mg, 48%. Molecular Formula: C24H23ClN4O2 ESI-MS calc: 434.15 ESI-MS found: 435.15612 HPLC: 5.398

(Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-benzyl-2-oxoindoline-5-carboxamide (8i).

Synthesized using protocol described for 8a. Yields a red solid, 210 mg, quantitative yield. Molecular Formula: C23H22N4O2 ESI-MS calc: 386.17 ESI-MS found: 387.1810 HPLC: 5.129.

(Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(2-phenylpropan-2-yl)indoline-5-carboxamide (8j).

Synthesized using protocol described for 8a. Yields a red solid, 307 mg, quantitative yield. Molecular Formula: C25H26N4O2 ESI-MS calc: 414.21 ESI-MS found: 415.2119 [M+1] HPLC: 5.245

(R,Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)-methylene)-N-(2-methyl-1-phenylpropyl)-2-oxoindoline-5-carboxamide (8k).

Synthesized using protocol described for 8a. Yields a red solid, 220 mg, quantitative yield. Molecular Formula: C26H28N4O2 ESI-MS calc: 428.22 ESI-MS found: 429.2201 HPLC: 5.497

(Z)-3-((4-amino-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(3-phenyloxetan-3-yl)indoline-5-carboxamide (8l).

Synthesized using protocol described for 8a. Yields a red solid, 20.6 mg, 47%. Molecular Formula: C25H24N4O3 ESI-MS calc: 428.18 ESI-MS found: 429.1494 HPLC: 4.710

(R,Z)-3-((4-(but-2-ynamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (9a).

To a flask were added 22.3 mg (0.37 mmol) of butynoic acid, 157.5 mg (0.37 mmol) of HATU, cat. DMAP and 50 mg (0.19 mmol) of 8a dissolved in 1 mL of DMF. To this bright red solution was added 0.25 mL (1.3 mmol) of DIPEA, and the solution was allowed to stir at room temperature for 2 hours. Once complete, the reaction mixture was diluted with EtOAc and washed with sat. LiCl and then dried over MgSO4. The crude material was purified by preparatory TLC with 50% acetone/hexanes. The desired band was collected, and the material was rinsed off the silica gel with acetone and then solvent was removed under pressure to give the desired product. Result: yellow solid, 17.5 mg, 20% Molecular Formula: C28H26N4O3 ESI-MS calc: 466.20 ESI-MS found 467.2071 HPLC: 6.336 1H NMR (700 MHz, DMSO-d6) δ 13.53 (s, 1H), 11.07 (s, 1H), 9.78 (s, 1H), 8.86 (d, J = 4.5 Hz, 2H), 8.68 (d, J = 8.8 Hz, 2H), 8.60 (d, J = 7.7 Hz, 2H), 8.21 (s, 1H), 7.69 (d, J = 8.9 Hz, 2H), 7.65 – 7.62 (m, 3H), 7.41 (d, J = 7.9 Hz, 5H), 7.33 (t, J = 7.3 Hz, 7H), 7.23 (d, J = 7.0 Hz, 3H), 6.93 (d, J = 8.1 Hz, 1H), 6.59 (s, 3H), 5.21 – 5.17 (m, 1H), 2.19 (s, 4H), 2.17 (s, 4H), 2.03 (s, 3H), 1.52 (s, 2H). 13C NMR (176 MHz, DMSO) δ 170.28, 167.22, 165.79, 149.50, 148.74, 145.19, 139.73, 135.08, 130.24, 128.98, 128.19, 127.41, 126.55, 125.97, 125.02, 122.58, 120.62, 117.41, 112.97, 55.89, 22.67, 12.09, 9.59, 3.60.

(R,Z)-3-((4-acrylamido-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (9b).

To a round bottom flask that contained 8a (50 mg, 0.12 mmol) were added 0.01 mL (0.12 mmol) of acrylic acid, 58.8 mg (0.12 mmol) of HATU, cat. DMAP and 0.50 mL (0.87 mmol) of TEA. All materials were dissolved in 2 mL of DMF and sonicated to give a homogenous solution. The solution was allowed to stir at 56 °C for 2 hours. Once complete, quenched with sat. LiCl and then extracted with EtOAc (3 × 30 mL). The combined organic layers were then dried over MgSO4 and the material was purified by preparatory TLC (50% Acetone/hexanes) collecting the baseline product, which was washed off the silica gel with acetone. The solvent was removed to give the final product as an orange solid, 13.9 mg, 24%. Molecular Formula: C27H26N4O3 ESI-MS calc: 454.20 ESI-MS found: 455.1632 [M+1] HPLC: 6.147 1H NMR (700 MHz, DMSO-d6) δ 13.53 (s, 1H), 11.07 (s, 1H), 9.41 (s, 1H), 8.60 (d, J = 8.1 Hz, 2H), 8.21 (d, J = 9.5 Hz, 1H), 7.69 (d, J = 8.3 Hz, 2H), 7.66 (s, 1H), 7.41 (d, J = 7.8 Hz, 5H), 7.33 (t, J = 7.5 Hz, 5H), 7.22 (t, J = 7.4 Hz, 3H), 6.93 (dd, J = 8.1, 3.6 Hz, 1H), 6.45 (dd, J = 17.1, 10.3 Hz, 1H), 6.23 – 6.19 (m, 1H), 5.72 (dd, J = 10.7, 1.9 Hz, 1H), 5.19 (t, J = 7.3 Hz, 2H), 2.21 (s, 3H), 2.20 (s, 3H), 1.50 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 180.81, 169.69, 169.00, 167.87, 165.86, 153.99, 145.07, 141.22, 140.27, 136.23, 131.78, 129.29, 128.18, 127.56, 126.86, 126.49, 126.09, 124.16, 120.41, 109.40, 108.64, 53.84, 22.28.

(R,Z)-3-((3,5-dimethyl-4-(vinylsulfonamido)-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (9c).

Step 1: To a dried flask were added 0.03 mL (0.12 mmol) of vinyl sulfonic acid, 2 drops of DMF and 2 mL of DCM. The solution was cooled to 0 °C and then 0.02 mL (0.14 mmol) of oxalyl chloride was added in one portion. The solution was warmed to room temperature and allowed to stir until complete (1 hour). Once complete, then solvent was removed, and the resultant clear oil was rinsed with DCM (3 × 15 mL) and dried under high pressure until ready for step 2. Step 2: To the flask holding the acid chloride was added 8a (50 mg, 0.12 mmol) dissolved in 3 mL of THF. To this murky orange solution was added 0.50 mL (7 equivalents) of TEA, and the solution was allowed to stir at room temperature for 5 hours. Once complete, removed solvent under pressure and brought yellow residue back up in EtOAc. The organic layer was washed with sat. Na2CO3 and then brine (1 × 30 mL) respectively. Purified by preparatory TLC plate (40% acetone/hexanes), collecting secondary spot (Rf = 0.2–0.3). The material was rinsed off silica with acetone and the solvent was removed to give a dark orange solid as the desired product. Result: orange solid,16 mg, 27% Molecular Formula: C26H26N4O4S ESI-MS calc: 490.17 ESI-MS found: 491.1249 HPLC: 6.43 1H NMR (400 MHz, DMSO-d6) δ 13.50 (s, 1H), 11.38 (s, 1H), 11.09 (s, 1H), 8.98 (s, 1H), 8.64 – 8.57 (m, 2H), 8.36 (s, 1H), 8.20 (s, 1H), 7.84 (s, 1H), 7.77 (d, J = 8.3 Hz, 1H), 7.70 (d, J = 8.2 Hz, 1H), 7.63 (s, 1H), 7.41 (d, J = 7.8 Hz, 5H), 7.33 (t, J = 7.6 Hz, 5H), 7.21 (d, J = 7.3 Hz, 2H), 6.98 (s, 1H), 6.93 (d, J = 8.1 Hz, 1H), 6.84 (dd, J = 16.5, 9.8 Hz, 1H), 5.93 (d, J = 8.0 Hz, 1H), 5.19 (t, J = 7.4 Hz, 2H), 2.29 (s, 4H), 2.26 (s, 3H), 1.50 (d, J = 7.1 Hz, 6H). 13C NMR (176 MHz, DMSO) δ 169.90, 165.56, 145.07, 141.33, 136.84, 133.74, 128.32, 128.19, 127.65, 126.53, 126.15, 125.20, 124.79, 124.46, 123.83, 119.49, 119.12, 109.21, 48.52, 45.44, 39.88, 39.76, 39.64, 39.52, 39.40, 39.28, 39.16, 22.33, 14.83, 11.29, 8.63.

(R,Z)-3-((4-(2-cyanoacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (9d).

To a round bottom flask were added 20.2 mg of 8a (0.05 mmol), 6.7 mg (0.075 mmol) of cyanoacetic acid and 24.6 mg (0.1 mmol) of DMTMM. All materials were brought up in 2 mL of DMF, and the resulting solution was allowed to stir at room temperature for 12 hours. Once complete, the reaction was quenched with sat. NaCl, and extracting with EtOAc (3 × 50 mL). The organic layer was washed with sat. Na2CO3 and brine (1 × 50 mL) respectively and then dried over MgSO4. The solvent was removed under pressure, evaporating the material onto silica gel. Purified by column chromatography 5–100% Acetone/hexanes. Flushed the column with 7 N NH3 in MeOH to yield the final product as a bright orange solid. Result: orange solid, 10 mg, 43% Molecular Formula: C27H25N5O3 ESI-MS calc: 467.20 ESI-MS found: 468.1297 HPLC: 6.100. 1H NMR (700 MHz, DMSO-d6) δ 13.51 (s, 1H), 11.08 (s, 1H), 9.52 (s, 1H), 8.60 (d, J = 8.2 Hz, 1H), 8.21 (d, J = 1.7 Hz, 1H), 7.69 (dd, J = 8.1, 1.7 Hz, 1H), 7.65 (s, 1H), 7.41 (d, J = 7.7 Hz, 2H), 7.33 (t, J = 7.6 Hz, 3H), 7.22 (t, J = 7.3 Hz, 1H), 6.93 (d, J = 8.1 Hz, 1H), 5.19 (p, J = 7.2 Hz, 1H), 3.88 (s, 2H), 2.20 (dd, J = 12.9, 10.8 Hz, 6H), 1.50 (d, J = 7.1 Hz, H).

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (9e).

To a dried round bottom flask was added 8a (60 mg, 0.15 mmol) dissolved in 3 mL of THF. The yellow solution was cooled to 0 °C and 0.01 mL (0.18 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, quenched with water and extracted with EtOAc. The solvent was removed, and the crude material was purified by prep plate (50% acetone/hexanes) collecting the major product. The desired product was rinsed off of silica gel and then the solvent was removed to give a bright yellow solid. The yellow solid was washed with DCM and sonicated to give a red solid. The red solid was then pulped in water:acetone (30:1) to give the final product. Result: red solid, 25.1 mg, 35% Molecular Formula: C26H25ClN4O3 ESI-MS calc: 476.16 ESI-MS found: 477.0999 HPLC: 6.388 1H NMR (700 MHz, DMSO-d6) δ 11.08 (s, 1H), 9.53 (s, 1H), 8.60 (d, J = 8.0 Hz, 1H), 8.22 (s, 1H), 7.69 (d, J = 8.2 Hz, 1H), 7.65 (s, 1H), 7.41 (d, J = 7.8 Hz, 2H), 7.33 (t, J = 7.5 Hz, 2H), 7.22 (t, J = 7.4 Hz, 1H), 6.93 (d, J = 8.0 Hz, 1H), 5.19 (t, J = 7.5 Hz, −1H), 2.20 (d, J = 12.1 Hz, 7H), 2.08 (s, 3H), 1.50 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.67, 168.51, 156.78, 145.12, 140.30, 131.64, 129.98, 128.13, 127.50, 126.89, 126.44, 126.11, 124.45, 122.62, 121.01, 117.42, 116.93, 112.94, 48.39, 42.71, 22.29, 11.67, 9.24.

(Z)-3-((4-((E)-4-(dimethylamino)but-2-enamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-((R)-1-phenylethyl)indoline-5-carboxamide (9f).

To a round bottom flask were added 8a (50 mg, 0.12 mmol), 20.2 mg (0.12 mmol) of N,N-dimethylaminobutenoic acid and 36 mg (0.12 mmol) of DMTMM and 0.20 mL (0.24 mmol) of TEA. All materials were brought up in 2 mL of DMF, and the resulting solution was allowed to stir at room temperature for 12 hours. Once complete, the reaction was quenched with Sat. NaCl, and extracting with EtOAc (3 × 50 mL). The organic layer was washed with sat. Na2CO3 and brine (1 × 50 mL) respectively and then dried over MgSO4. The solvent was removed under pressure, evaporating the material onto silica gel. Purified by column chromatography 5–100% acetone/hexanes. Flushed the column with 7 N NH3 in MeOH to yield the final product as a bright yellow oil. Result: yellow oil, 13 mg, 20% Molecular Formula: C30H33N5O3 ESI-MS calc: 511.26 ESI-MS found: 512.2110 [M+1], 534.1905 [M+Na] HPLC: 5.424 1H NMR (700 MHz, DMSOd6) δ 13.56 (s, 1H), 11.06 (s, 2H), 9.29 (s, 2H), 8.59 (d, J = 7.9 Hz, 2H), 8.21 (d, J = 1.5 Hz, 2H), 7.68 (d, J = 8.1 Hz, 2H), 7.65 (s, 2H), 7.41 (d, J = 7.6 Hz, 4H), 7.33 (t, J = 7.6 Hz, 4H), 7.22 (t, J = 7.5 Hz, 2H), 6.93 (d, J = 8.1 Hz, 2H), 6.71 – 6.64 (m, 1H), 6.27 (d, J = 15.5 Hz, 1H), 5.21 – 5.17 (m, 1H), 3.05 (d, J = 5.6 Hz, 2H), 2.21 (s, 6H), 2.19 (s, 3H), 2.18 (s, 3H), 1.50 (d, J = 7.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 170.09, 166.24, 163.98, 145.47, 141.00, 140.66, 132.25, 128.68, 128.55, 127.91, 127.30, 126.87, 126.48, 126.08, 125.81, 125.74, 125.61, 124.86, 124.52, 122.19, 117.61, 112.99, 109.03, 62.41, 48.96, 25.82, 22.64, 14.44, 12.32, 9.77.

(R,Z)-3-((4-(2-bromoacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-phenylethyl)indoline-5-carboxamide (9g).

To a round bottom flask were added 83.7 mg of 8a (0.21 mmol), 60.1 mg (0.41 mmol) of bromoacetic acid and 100 mg (0.25 mmol) of DMTMM. All materials were brought up in 2 mL of DMF, and the resulting solution was allowed to stir at room temperature for 1.5 hours. Once complete, the reaction was quenched with brine and then extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na2SO4 and then solvent was removed under pressure to give a dark orange solid. The solid was then dissolved in DCM, and the resultant solution was sonicated to give a dark red precipitate. The solid was filtered off and rinsed with excess cold DCM to give the final product. Result: red solid, 42.9 mg, 39%. Molecular Formula: C26H25BrN4O3 ESI-MS calc: 520.11 ESI-MS found: 523.1164 HPLC: 6.397 1H NMR (700 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.08 (s, 1H), 9.60 (s, 1H), 8.60 (d, J = 7.9 Hz, 1H), 8.21 (d, J = 1.6 Hz, 1H), 7.69 (dd, J = 8.1, 1.7 Hz, 1H), 7.65 (s, 1H), 7.41 (d, J = 7.7 Hz, 2H), 7.33 (t, J = 7.6 Hz, 2H), 7.22 (t, J = 7.3 Hz, 1H), 6.93 (d, J = 8.1 Hz, 1H), 5.20 (p, J = 7.2 Hz, 1H), 4.03 (s, 2H), 2.20 (d, J = 10.4 Hz, 6H), 1.51 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 169.68, 165.82, 165.54, 145.05, 140.30, 131.64, 128.15, 127.59, 126.78, 126.47, 126.07, 125.76, 125.33, 124.44, 124.13, 121.01, 117.35, 112.95, 108.62, 48.34, 29.33, 22.26, 11.61, 9.14.

(Z)-3-((4-(3-chloro-2-hydroxypropanamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-((R)-1-phenylethyl)indoline-5-carboxamide (9h).

To a round bottom flask were added 80 mg of 8a (0.20 mmol), 31.1 mg (0.26 mmol) of 3-chloro-2hydroxypropionic acid and 81.8 mg (0.32 mmol) of DMTMM. All materials were brought up in 2 mL of DMF, and the resulting solution was allowed to stir at room temperature for 12 hours. Once complete, the reaction was quenched with sat. NaCl, and extracting with EtOAc (3 × 50 mL). The organic layer was washed with sat. Na2CO3 and brine (1 × 50 mL) respectively and then dried over MgSO4. The solvent was removed under pressure, evaporating the material onto silica gel. Purified by column chromatography 5–100% acetone/hexanes. Result: orange solid, 72.8 mg, 58% Molecular Formula: C27H27ClN4O4 ESI-MS calc: 506.17 ESI-MS found: 507.1809 [M+1], 540.2377 [M+H+MeOH] HPLC: 6.043 1H NMR (700 MHz, DMSO-d6) δ 13.58 – 13.42 (m, 1H), 11.07 (d, J = 4.9 Hz, 1H), 8.60 (d, J = 8.1 Hz, 1H), 8.23 – 8.19 (m, 1H), 7.95 (s, 1H), 7.69 – 7.67 (m, 1H), 7.64 (d, J = 5.9 Hz, 1H), 7.41 (d, J = 7.2 Hz, 2H), 7.33 (t, J = 7.1 Hz, 3H), 7.22 (t, J = 7.2 Hz, 1H), 6.93 (dt, J = 8.2, 2.1 Hz, 1H), 5.19 (p, J = 7.3 Hz, 1H), 3.96 (d, J = 7.9 Hz, 1H), 3.74 (s, 1H), 2.21 – 2.18 (m, 4H), 2.16 (s, 2H), 1.50 (d, J = 7.0 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 172.44, 166.31, 162.74, 145.53, 128.63, 127.95, 126.94, 126.54, 125.88, 124.64, 121.80, 117.71, 115.92, 109.07, 107.36, 55.41, 48.82, 22.73, 21.51, 9.74.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(m-tolyl)ethyl)indoline-5-carboxamide (9i).

To a dried round bottom flask was added 86.6 mg of 8b, (0.209 mmol) dissolved in 3 mL of THF. The yellow solution was cooled to 0 °C and 0.04 mL (0.30 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, quenched with water and extracted with EtOAc. The solvent was removed, and the crude material was purified by prep plate (50% acetone/hexanes) collecting the major product. The desired product was rinsed off of silica gel and then the solvent was removed to give a bright yellow solid. The yellow solid was washed with DCM and sonicated to give a red solid. The red solid was then pulped in water:acetone (30:1) to give the final product. Result: red solid, 57.2 mg, 55% Molecular Formula: C27H27ClN4O3 ESI-MS calc: 490.18 ESI-MS found: 491.1133 HPLC: 6.68 1H NMR (500 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.07 (s, 1H), 9.53 (s, 1H), 8.56 (d, J = 8.1 Hz, 1H), 8.21 (s, 1H), 7.74 – 7.62 (m, 2H), 7.24 – 7.15 (m, 4H), 7.03 (s, 1H), 6.93 (d, J = 8.0 Hz, 1H), 5.15 (s, 1H), 4.26 (s, 2H), 2.30 (s, 3H), 2.20 (d, J = 8.9 Hz, 6H), 1.49 (d, J = 7.0 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 170.18, 169.06, 166.24, 165.84, 145.51, 140.80, 137.63, 132.18, 128.59, 128.10, 127.62, 127.24, 126.26, 125.83, 124.93, 123.68, 121.47, 117.93, 113.45, 109.13, 48.80, 43.22, 22.79, 21.62, 12.15, 9.70.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(4-fluorophenyl)ethyl)-2-oxoindoline-5carboxamide (9j).

To a dried round bottom flask was added 96.0 mg of 8c, (0.23 mmol) dissolved in 4 mL of THF. The yellow solution was cooled to 0 °C and 0.02 mL (0.25 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, quenched with water and extracted with EtOAc. The solvent was removed, and the crude material was purified by prep plate (50% acetone/hexanes) collecting the major product. The desired product was rinsed off of silica gel and then the solvent was removed to give a bright yellow solid. The yellow solid was washed with DCM and sonicated to give a red solid. The red solid was then pulped in water:acetone (30:1) to give the final product. Result: red solid, 46.5 mg, 41% Molecular Formula: C26H24FClN4O3 ESI-MS calc: 494.15 ESI-MS found: 495.0480 [M+1] HPLC: 6.439 1H NMR (700 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.08 (s, 1H), 9.54 (s, 1H), 8.63 (d, J = 7.9 Hz, 1H), 8.23 (d, J = 1.7 Hz, 1H), 7.68 (dd, J = 8.1, 1.8 Hz, 1H), 7.66 (s, 1H), 7.46 – 7.43 (m, 2H), 7.15 (tt, J = 9.9, 3.2 Hz, 3H), 6.93 (d, J = 8.2 Hz, 1H), 5.19 (p, J = 7.2 Hz, 1H), 4.28 – 4.26 (m, 4H), 2.20 (d, J = 10.1 Hz, 6H), 1.50 (d, J = 6.9 Hz, 4H). 13C NMR (176 MHz, DMSO) δ 170.20, 169.07, 166.36, 165.86, 162.12, 160.75, 141.78, 141.76, 140.85, 132.22, 128.54, 128.49, 128.41, 128.02, 127.42, 126.27, 125.87, 124.96, 124.70, 121.50, 117.90, 115.38, 115.26, 113.44, 109.15, 48.33, 43.23, 41.97, 40.37, 40.25, 40.13, 40.01, 39.89, 39.77, 39.65, 36.07, 29.51, 22.78, 12.16, 9.72.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(pyridin-4-yl)ethyl)indoline-5-carboxamide (9k).

To a dried round bottom flask was added 41.5 mg of 8d, (0.103 mmol) dissolved in 2 mL of THF. The yellow solution was cooled to 0 °C and 0.01 mL (0.103 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, quenched with water and extracted with EtOAc. The solvent was removed, and the crude material was purified by prep plate (50% acetone/hexanes) collecting the major product as the desired product. Result: red solid, 6.3 mg, 13% Molecular Formula: C25H24ClN5O3 ESI-MS calc: 477.16 ESI-MS found: 478.1734 [M+H] HPLC: 4.363 1H NMR (700 MHz, DMSO-d6) δ 13.51 (s, 1H), 11.15 (s, 1H), 10.56 (s, 1H), 9.86 (s, 1H), 8.73 (s, 1H), 8.48 (s, 2H), 7.75 (s, 1H), 7.72 (d, J = 7.9 Hz, 1H), 7.34 (d, J = 7.6 Hz, 2H), 7.27 (s, 1H), 6.96 (d, J = 7.9 Hz, 1H), 5.33 (t, J = 7.0 Hz, 1H), 4.27 (s, 2H), 2.21 (s, 6H), 1.50 (d, J = 7.8 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 168.53, 166.49, 165.34, 154.01, 141.60, 140.65, 130.38, 129.30, 129.05, 127.97, 124.37, 123.04, 122.68, 121.09, 119.53, 118.36, 108.73, 54.90, 42.72, 21.66, 10.38, 8.77.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(pyridin-2-yl)ethyl)indoline-5-carboxamide (9l).

To a dried round bottom flask was added 73.8 mg of 8e, (0.18 mmol) dissolved in 4 mL of THF. The yellow solution was cooled to 0 °C and 0.04 mL (0.50 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, removed solvent under pressure and then added DCM to the dark red residue. The DCM solution was sonicated to give a dark red precipitate. The solid was filtered off and rinsed with THF and DCM to give the desired product as a dark red solid. Result: red solid, 37.1 mg, 42% Molecular Formula: C25H24ClN5O3 ESI-MS calc: 477.16 ESI-MS found: 478.1547 [M+1] HPLC: 4.743 1H NMR (700 MHz, DMSO-d6) δ 13.28 (t, J = 13.6 Hz, 1H), 11.02 (q, J = 7.5, 5.9 Hz, 1H), 9.86 (s, 1H), 8.98 (d, J = 52.4 Hz, 1H), 8.57 (d, J = 9.4 Hz, 1H), 8.42 (d, J = 9.4 Hz, 1H), 8.26 (q, J = 9.9, 9.4 Hz, 2H), 7.86 (d, J = 11.1 Hz, 1H), 7.64 (s, 2H), 7.58 (d, J = 9.2 Hz, 1H), 7.50 (dt, J = 27.4, 8.6 Hz, 1H), 6.72 (p, J = 8.1 Hz, 1H), 5.23 – 5.16 (m, 1H), 4.03 (t, J = 8.6 Hz, 1H), 2.92 (d, J = 16.9 Hz, 14H), 2.26 (d, J = 10.2 Hz, 7H), 2.17 (dt, J = 18.1, 8.8 Hz, 4H), 1.97 (q, J = 8.9 Hz, 2H), 1.48 – 1.40 (m, 3H). 13C NMR (176 MHz, DMSO) δ 169.75, 166.56, 165.36, 159.41, 145.67, 142.08, 141.13, 140.72, 131.91, 128.62, 127.17, 126.78, 126.44, 125.11, 124.85, 124.26, 123.99, 121.10, 118.54, 52.68, 42.73, 20.48, 11.23, 8.81.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(3-chlorophenyl)ethyl)-2-oxoindoline-5-carboxamide (9m).

To a dried round bottom flask was added 40 mg of 8f, (0.16 mmol) dissolved in 4 mL of THF. The yellow solution was cooled to 0 °C and 0.01 mL (0.18 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, removed solvent under pressure and then added DCM to the dark red residue. The DCM solution was sonicated to give a dark red precipitate. The solid was filtered off and rinsed with THF and DCM to give the desired product as a dark red solid. Result: red solid, 23.4 mg, 28% Molecular Formula: C26H24Cl2N4O3 ESI-MS calc: 510.12 ESI-MS found: 511.1579 [M+1] HPLC: 6.742 1H NMR (500 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.09 (s, 1H), 9.53 (s, 1H), 8.66 (d, J = 7.9 Hz, 1H), 8.21 (d, J = 1.7 Hz, 1H), 7.68 (dd, J = 8.2, 1.7 Hz, 1H), 7.66 (s, 1H), 7.47 – 7.45 (m, 1H), 7.38 – 7.36 (m, 2H), 7.29 (dq, J = 6.2, 2.2 Hz, 1H), 6.94 (d, J = 8.1 Hz, 1H), 5.17 (p, J = 7.2 Hz, 1H), 4.26 (s, 2H), 2.20 (d, J = 8.6 Hz, 6H), 1.50 (d, J = 7.1 Hz, 4H). 13C NMR (126 MHz, DMSO) δ 169.51, 165.17, 147.64, 140.23, 132.71, 131.57, 129.96, 127.20, 126.77, 126.30, 125.79, 125.55, 125.23, 124.73, 124.28, 120.82, 117.27, 112.71, 48.03, 42.55, 39.83, 39.67, 39.50, 39.33, 39.17, 39.00, 38.83, 21.96, 11.48, 9.02.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(1-(p-tolyl)ethyl)indoline-5-carboxamide (9n).

To a dried round bottom flask was added 93 mg of 8g, (0.19 mmol) dissolved in 5 mL of THF. The yellow solution was cooled to 0 °C and 0.1 mL (1.3 mmol) of chloroacetylchloride was added dropwise. The solution was allowed to stir at 0 °C for 30 minutes. Once complete by TLC, removed solvent under pressure and then added DCM to the dark red residue. The DCM solution was sonicated to give a dark red precipitate. The solid was filtered off and rinsed with THF and DCM to give the desired product as a dark red solid. Result: red solid, 73.6 mg, 77% Molecular Formula: C27H27ClN4O3 ESI-MS calc: 490.18 ESI-MS found 473.05 [M-Cl] HPLC: 6.625 1H NMR (500 MHz, DMSO-d6) δ 13.51 (s, 1H), 11.06 (s, 1H), 9.52 (s, 1H), 8.53 (d, J = 8.0 Hz, 1H), 8.20 (s, 1H), 7.71 – 7.65 (m, 1H), 7.64 (s, 1H), 7.27 (t, J = 10.7 Hz, 4H), 7.12 (d, J = 7.8 Hz, 6H), 6.92 (d, J = 8.1 Hz, 1H), 5.18 – 5.12 (m, 2H), 2.26 (d, J = 3.9 Hz, 8H), 1.48 (d, J = 7.1 Hz, 5H).

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(4-chlorophenyl)ethyl)-2-oxoindoline-5carboxamide (9o).

Synthesized using the protocol described in 8h. Yields a brown solid, 43.5 mg, 100%. Molecular Formula: C26H24Cl2N4O3 ESI-MS calc: 510.12 ESI-MS found: 511.12917 [M+1] HPLC: 6.723. 1H NMR (499 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.08 (d, J = 7.8 Hz, 1H), 9.53 (s, 1H), 8.64 (dd, J = 7.9, 5.3 Hz, 2H), 8.20 (d, J = 1.5 Hz, 1H), 7.67 (dt, J = 8.0, 2.1 Hz, 2H), 7.65 (s, 1H), 7.44 – 7.40 (m, 4H), 7.40 – 7.37 (m, 5H), 6.93 (d, J = 8.1 Hz, 1H), 5.16 (dq, J = 13.7, 7.0 Hz, 1H), 4.26 (d, J = 2.8 Hz, 2H), 2.20 (d, J = 8.4 Hz, 6H), 1.49 (d, J = 7.1 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 170.31, 169.68, 165.92, 165.35, 144.15, 140.36, 131.73, 130.99, 128.11, 128.01, 127.44, 126.92, 126.09, 125.76, 125.35, 124.44, 123.36, 122.09, 121.43, 120.99, 120.43, 117.37, 108.64, 55.81, 42.72, 22.10, 11.65, 9.21.

(R,Z)-3-((4-(2-bromoacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(1-(4-fluorophenyl)ethyl)-2-oxoindoline-5-carboxamide (9p).

This material was prepared using the protocol described for 8c. Yields a bright orange solid, 13.7 mg, 23%. HRMS: 541.1106 [M+1, Br81], HPLC: 6.449. 1H NMR (700 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.08 (s, 1H), 9.60 (s, 1H), 8.61 (d, J = 8.1 Hz, 1H), 8.20 (s, 1H), 7.68 (d, J = 8.4 Hz, 1H), 7.65 (s, 1H), 7.44 (t, J = 4.8 Hz, 2H), 7.17 – 7.13 (m, 2H), 6.93 (dd, J = 8.2, 2.5 Hz, 1H), 5.19 (q, J = 7.4 Hz, 1H), 4.26 (d, J = 2.4 Hz, 1H), 4.03 (d, J = 2.4 Hz, 1H), 2.22 – 2.18 (m, 6H), 1.51 – 1.48 (m, 3H). 13C NMR (176 MHz, DMSO) δ 170.07, 167.64, 166.24, 162.00, 155.00, 144.70, 140.72, 132.05, 129.71, 128.41, 127.92, 124.56, 122.76, 121.42, 117.77, 115.96, 115.26, 109.01, 48.19, 29.73, 22.66, 12.01, 9.54.

(Z)-N-benzyl-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxoindoline-5-carboxamide (9q).

Synthesized using protocol described for 9i. Yields an orange solid, 18.2 mg, 21%. Molecular Formula: C25H23ClN4O3 ESI-MS calc: 462.15 ESI-MS found: 463.1529 [M+1] HPLC: 6.177. 1H NMR (700 MHz, DMSO-d6) δ 13.49 (s, 1H), 11.08 (s, 1H), 9.52 (s, 1H), 8.85 (dt, J = 12.4, 6.1 Hz, 1H), 8.26 (d, J = 1.7 Hz, 1H), 7.78 (d, J = 9.4 Hz, 1H), 7.71 (dd, J = 8.2, 1.7 Hz, 1H), 7.64 (s, 1H), 7.35 – 7.32 (m, 4H), 7.24 (tt, J = 6.2, 3.0 Hz, 2H), 6.94 (d, J = 8.1 Hz, 1H), 4.51 (d, J = 6.0 Hz, 2H), 4.26 (s, 2H), 2.21 (s, 3H), 2.18 (s, 3H). 13C NMR (176 MHz, DMSO) δ 170.18, 166.91, 165.85, 140.89, 140.42, 132.22, 128.74, 128.72, 128.70, 127.98, 127.78, 127.68, 127.67, 127.62, 127.38, 127.15, 126.21, 125.93, 124.95, 124.54, 123.95, 121.48, 117.63, 113.41, 109.28, 68.98, 56.32, 43.22, 43.09, 43.03, 40.37, 40.25, 40.13, 40.01, 39.89, 39.77, 39.65, 36.08, 32.59, 30.08, 12.14, 11.67, 9.92, 9.65.

(Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(2-phenylpropan-2-yl)indoline-5-carboxamide (9r).

Synthesized using the protocol described for 9i. Yields a brown solid, 14.2 mg, 15%. Molecular Formula: C27H27ClN4O3 ESI-MS calc: 490.18 ESI-MS found: 491.1832 [M+1], 513.1656 [M+Na] HPLC: 6.63. 1H NMR (500 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.06 (s, 1H), 9.53 (s, 1H), 8.24 (s, 1H), 8.19 (d, J = 1.8 Hz, 1H), 7.68 (s, 1H), 7.64 (dd, J = 8.1, 1.7 Hz, 1H), 7.40 (dd, J = 7.9, 1.8 Hz, 3H), 7.28 (t, J = 7.8 Hz, 2H), 7.16 (dd, J = 8.3, 6.3 Hz, 1H), 6.91 (d, J = 8.1 Hz, 1H), 4.26 (s, 2H), 2.20 (d, J = 8.2 Hz, 6H), 1.69 (s, 6H). 13C NMR (126 MHz, DMSO) δ 170.12, 166.37, 165.75, 148.67, 143.56, 140.59, 132.03, 128.86, 128.24, 127.26, 126.00, 125.65, 125.11, 124.86, 124.66, 121.37, 117.90, 108.94, 55.69, 43.14, 30.13, 12.07, 9.61.

(R,Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-N-(2-methyl-1-phenylpropyl)-2-oxoindoline-5-carboxamide (9s).

Synthesized using the protocol described for 9i. Yields an orange solid, 5.5 mg, 5.8%. Molecular Formula: C28H29ClN4O3 ESI-MS calc: 504.19 ESI-MS found: 505.1997 HPLC: 6.803 1H NMR (700 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.06 (s, 1H), 9.52 (s, 1H), 8.53 (d, J = 8.9 Hz, 1H), 8.14 (d, J = 1.6 Hz, 1H), 7.67 – 7.62 (m, 2H), 7.43 – 7.40 (m, 2H), 7.32 (t, J = 7.6 Hz, 2H), 7.22 (t, J = 7.3 Hz, 1H), 6.92 (d, J = 8.1 Hz, 1H), 4.69 (t, J = 9.2 Hz, 1H), 4.26 (s, 2H), 2.20 (d, J = 10.3 Hz, 6H), 1.04 (d, J = 6.6 Hz, 4H), 0.73 (d, J = 6.7 Hz, 4H). 13C NMR (126 MHz, DMSO) δ 170.14, 166.69, 165.80, 151.43, 143.78, 140.68, 132.14, 128.47, 128.45, 127.89, 127.78, 127.38, 127.05, 125.84, 124.90, 124.74, 121.43, 118.01, 108.97, 60.35, 43.19, 32.97, 20.45, 12.12, 9.62.

(Z)-3-((4-(2-chloroacetamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-(3-phenyloxetan-3-yl)indoline-5-carboxamide (9t).

Synthesized using the protocol described for 9i. Yields a red solid, 5.0 mg, 21%. Molecular Formula: C27H25ClN4O4 ESI-MS calc: 504.16 ESI-MS found: 505.1619 HPLC: 5.332 1H NMR (700 MHz, DMSO-d6) δ 13.52 (s, 1H), 11.28 (s, 1H), 9.54 (d, J = 13.2 Hz, 1H), 8.43 (s, 1H), 7.84 (s, 2H), 7.77 (d, J = 13.4 Hz, 2H), 7.52 (t, J = 9.1 Hz, 4H), 7.39 (dt, J = 14.7, 7.8 Hz, 5H), 7.31 (dd, J = 16.7, 7.7 Hz, 3H), 7.19 (s, 1H), 7.11 (s, 1H), 7.04 (s, 2H), 4.85 (d, J = 9.0 Hz, 2H), 4.45 (t, J = 9.9 Hz, 2H), 4.27 (d, J = 3.1 Hz, 2H), 2.22 (d, J = 4.8 Hz, 6H). 13C NMR (126 MHz, DMSO) δ 170.16, 167.57, 165.90, 154.45, 143.80, 130.21, 129.17, 129.09, 129.04, 128.64, 126.55, 126.07, 125.78, 125.47, 125.31, 122.08, 121.39, 120.35, 115.60, 110.34, 78.59, 72.40, 43.21, 11.78, 9.23.

(Z)-3-((4-((E)-2-cyano-4,4-dimethylpent-2-enamido)-3,5-dimethyl-1H-pyrrol-2-yl)methylene)-2-oxo-N-((R)-1-phenylethyl)indoline-5-carboxamide (10a).

To a sealed tube were added 30 mg of 9g (0.064 mmol), 0.05 mL of pivaldehyde (0.45 mmol), and 0.10 mL of piperidine (1.0 mmol). All materials were then dissolved in 3 mL of abs. EtOH. The solution was then heated to 75 °C for 12 hours. Once complete, the reaction was cooled to room temperature and then solvent was removed under pressure, evaporating the crude material onto silica gel. The material was purified by column chromatography (50% acetone/hexanes) to give the desired product. Yields a yellow solid, 14 mg, 41%. MS: 536.1375 [M+1], 558 [M+Na] HPLC: 7.69 1H NMR (700 MHz, DMSO-d6) δ 13.54 (s, 1H), 11.09 (s, 1H), 9.57 (d, J = 11.5 Hz, 1H), 8.60 (s, 2H), 8.22 (s, 1H), 7.95 (s, 1H), 7.69 (d, J = 8.4 Hz, 1H), 7.66 (d, J = 9.8 Hz, 1H), 7.41 (d, J = 7.7 Hz, 3H), 7.33 (t, J = 7.5 Hz, 3H), 7.23 (d, J = 7.9 Hz, 2H), 6.93 (d, J = 8.4 Hz, 1H), 5.19 (s, 1H), 2.22 – 2.17 (m, 6H), 1.50 (d, J = 7.2 Hz, 4H), 1.15 (d, J = 9.5 Hz, 9H), 1.04 (d, J = 6.1 Hz, 3H). 13C NMR (176 MHz, DMSO) δ 170.08, 167.90, 166.20, 160.95, 153.74, 145.51, 141.02, 140.65, 135.87, 132.53, 128.55, 128.02, 126.86, 126.46, 124.51, 123.07, 120.43, 117.75, 115.67, 98.56, 56.19, 28.98, 25.24, 22.64, 12.00, 9.70.

(Z)-3-((3,5-dimethyl-4-(oxirane-2-carboxamido)-1H-pyrrol-2-yl)methylene)-2-oxo-N-((R)-1-phenylethyl)indoline-5-carboxamide (10b).

To a flask were added 36.4 mg of 9h, and 47.2 mg of K2CO3. All materials were brought up in 3.0 mL of acetone and 1 mL of MeCN and then the reaction was refluxed (60 °C) for 30 minutes. After 30 minutes, the reaction was heated to 70 °C for 1 h, with cat. KI to push the reaction to completion. Once complete, the reaction was quenched with water (1 × 30 mL) and then extracted with EtOAc (2 × 25 mL). The combined organic layers were dried over Na2SO4 and then purified by column chromatography (25–100% acetone/hexanes). The desired fractions were collected, and the solvent was removed in vacuo giving the product as a yellow solid. Result: yellow solid, 3.0 mg, 6% Molecular Formula: C27H26ClN4O4 ESI-MS calc: 470.20 ESI-MS found: 471.2158 [M+1] HPLC: 6.591 1H NMR (700 MHz, acetone-d6) δ 13.62 (s, 1H), 10.01 (s, 1H), 8.24 – 8.22 (m, 2H), 7.83 (d, J = 8.3 Hz, 1H), 7.75 (dt, J = 8.1, 2.2 Hz, 1H), 7.69 (s, 1H), 7.46 (d, J = 8.0 Hz, 3H), 7.33 (t, J = 7.7 Hz, 3H), 7.23 (t, J = 7.3 Hz, 1H), 6.99 (dd, J = 8.3, 5.5 Hz, 1H), 5.33 (q, J = 7.3 Hz, 2H), 3.91 (s, 2H), 3.79 (s, 2H), 2.30 (s, 2H), 2.25 (d, J = 2.7 Hz, 3H), 2.19 (d, J = 5.5 Hz, 2H), 2.14 (d, J = 5.1 Hz, 3H), 1.56 (dd, J = 7.1, 1.3 Hz, 3H). 13C NMR (126 MHz, DMSO) δ 171.84, 169.70, 167.77, 152.56, 145.07, 140.26, 132.32, 128.18, 127.58, 127.45, 126.50, 126.09, 124.43, 121.76, 120.08, 118.18, 108.61, 54.27, 54.08, 48.37, 22.28, 11.87, 9.44.

Supplementary Material

Supplementary Material

ACKNOWLEDGMENT

We thank Dr. Venky Bashar and Kevin Conlon, of the University of Michigan Proteomic Resource Facility for assistance with tandem mass spectrometry efforts.

Funding Sources

This work was supported by NIH grants HL071818, HL122416, CA254402 and the Walther Cancer Foundation (to JJGT), the American Heart Association grants 19POST34450193 (to Q.C.) and 18POST33960047 (to RAB) and a pilot grant from the University of Michigan Center for Discovery of New Medicines (to ADW).

ABBREVIATIONS

AST

active site tether

GPCR

G protein-coupled receptor

GRK

G protein-coupled receptor kinase

P-loop

phosphate-binding loop

MS

mass spectrometry

RTK

receptor tyrosine kinase

SAR

structure-activity relationship

Footnotes

Supporting Information

Supporting figures S1–3, Table S1, Molecular Strings File, and coordinates for docked ligand complexes are provided in the Supporting Information.

The Supporting Information is available free of charge on the ACS Publications website.

The authors declare no competing financial interest.

REFERENCES

  • 1.Gurevich EV; Tesmer JJ; Mushegian A; Gurevich VV, G protein-coupled receptor kinases: more than just kinases and not only for GPCRs. Pharmacol Ther 2012, 133 (1), 40–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lieu M; Koch WJ, GRK2 and GRK5 as therapeutic targets and their role in maladaptive and pathological cardiac hypertrophy. Expert Opin Ther Targets 2019, 23 (3), 201–214. [DOI] [PubMed] [Google Scholar]
  • 3.Schumacher-Bass SM; Traynham CJ; Koch WJ, G protein-coupled Receptor Kinase 2 as a Therapeutic Target for Heart Failure. Drug Discov Today Ther Strateg 2012, 9 (4), e155–e162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sommer AK; Falcenberg M; Ljepoja B; Frohlich T; Arnold GJ; Wagner E; Roidl A, Downregulation of GRK5 hampers the migration of breast cancer cells. Scientific reports 2019, 9 (1), 15548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Nogues L; Palacios-Garcia J; Reglero C; Rivas V; Neves M; Ribas C; Penela P; Mayor F Jr., G protein-coupled receptor kinases (GRKs) in tumorigenesis and cancer progression: GPCR regulators and signaling hubs. Semin Cancer Biol 2018, 48, 78–90. [DOI] [PubMed] [Google Scholar]
  • 6.Martini JS; Raake P; Vinge LE; DeGeorge BR Jr.; Chuprun JK; Harris DM; Gao E; Eckhart AD; Pitcher JA; Koch WJ, Uncovering G protein-coupled receptor kinase-5 as a histone deacetylase kinase in the nucleus of cardiomyocytes. Proc Natl Acad Sci U S A 2008, 105 (34), 12457–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gold JI; Gao E; Shang X; Premont RT; Koch WJ, Determining the absolute requirement of G protein-coupled receptor kinase 5 for pathological cardiac hypertrophy: short communication. Circ Res 2012, 111 (8), 1048–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schlegel P; Reinkober J; Meinhardt E; Tscheschner H; Gao E; Schumacher SM; Yuan A; Backs J; Most P; Wieland T; Koch WJ; Katus HA; Raake PW, G protein-coupled receptor kinase 2 promotes cardiac hypertrophy. PLoS One 2017, 12 (7), e0182110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rowlands RA; Cato MC; Waldschmidt HV; Bouley RA; Chen Q; Avramova L; Larsen SD; Tesmer JJG; White AD, Structure-Based Design of Selective, Covalent G Protein-Coupled Receptor Kinase 5 Inhibitors. ACS Med Chem Lett 2019, 10 (12), 1628–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Singh J; Petter RC; Baillie TA; Whitty A, The resurgence of covalent drugs. Nat Rev Drug Discov 2011, 10 (4), 307–17. [DOI] [PubMed] [Google Scholar]
  • 11.O’Farrell AM; Abrams TJ; Yuen HA; Ngai TJ; Louie SG; Yee KW; Wong LM; Hong W; Lee LB; Town A; Smolich BD; Manning WC; Murray LJ; Heinrich MC; Cherrington JM, SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 2003, 101 (9), 3597–605. [DOI] [PubMed] [Google Scholar]
  • 12.Ullrich A; Falcenberg M; Örfi Z; Kéri G; ÖRFI L; Horváth Z; Szokol B; Dobos J; Nemes Z Indolinone Derivatives as GRK5 Modulators. WO2015022437 A1, February 19, 2015.
  • 13.Chen K; Pittman RN; Popel AS, Nitric oxide in the vasculature: where does it come from and where does it go? A quantitative perspective. Antioxid Redox Signal 2008, 10 (7), 1185–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Homan KT; Tesmer JJ, Molecular basis for small molecule inhibition of g protein-coupled receptor kinases. ACS Chem Biol 2015, 10 (1), 246–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Waldschmidt HV; Homan KT; Cato MC; Cruz-Rodriguez O; Cannavo A; Wilson MW; Song J; Cheung JY; Koch WJ; Tesmer JJ; Larsen SD, Structure-Based Design of Highly Selective and Potent G Protein-Coupled Receptor Kinase 2 Inhibitors Based on Paroxetine. J Med Chem 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lee JH; Seo HW; Ryu JY; Lim CJ; Yi KY; Oh KS; Lee BH, KR-39038, a Novel GRK5 Inhibitor, Attenuates Cardiac Hypertrophy and Improves Cardiac Function in Heart Failure. Biomol Ther (Seoul) 2020, 28 (5), 482–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ngai MH; So CL; Sullivan MB; Ho HK; Chai CL, Photoinduced Isomerization and Hepatoxicities of Semaxanib, Sunitinib and Related 3-Substituted Indolin-2-ones. ChemMedChem 2016, 11 (1), 72–80. [DOI] [PubMed] [Google Scholar]
  • 18.Homan KT; Waldschmidt HV; Glukhova A; Cannavo A; Song J; Cheung JY; Koch WJ; Larsen SD; Tesmer JJ, Crystal Structure of G Protein-coupled Receptor Kinase 5 in Complex with a Rationally Designed Inhibitor. J Biol Chem 2015, 290 (34), 20649–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Boguth CA; Singh P; Huang CC; Tesmer JJ, Molecular basis for activation of G protein-coupled receptor kinases. EMBO J 2010, 29 (19), 3249–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Thal DM; Homan KT; Chen J; Wu EK; Hinkle PM; Huang ZM; Chuprun JK; Song J; Gao E; Cheung JY; Sklar LA; Koch WJ; Tesmer JJ, Paroxetine is a direct inhibitor of g protein-coupled receptor kinase 2 and increases myocardial contractility. ACS Chem Biol 2012, 7 (11), 1830–9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Material

RESOURCES