Abstract
The efficacy of quinazoline-based antiglioma agents has been attributed to their effects on microtubule dynamics.1, 2 The design, synthesis and biological evaluation of quinazolines as potent inhibitors of multiple intracellular targets, including microtubules and multiple RTKs, is described. In addition to the known ability of quinazolines 1 and 2 to cause microtubule depolymerization, they were found to be low nanomolar inhibitors of EGFR, VEGFR-2 and PDGFR-β. Low nanomolar inhibition of EGFR was observed for 1-3 and 9-10. Compounds 1 and 4 inhibited VEGFR-2 kinase with activity better than or equal to that of sunitinib. In addition, compounds 1 and 2 had similar potency to sunitinib in the CAM angiogenesis assay. Multitarget activities of compounds in the present study demonstrates that the quinazolines can affect multiple pathways and could lead to these agents having antitumor potential caused by their activity against multiple targets.
Keywords: Quinazolines, microtubule targeting agents, receptor tyrosine kinase, multi-target inhibitors, angiogenesis
Graphical Abstract

Introduction
Angiogenesis is the formation of new blood vessels from existing vasculature and is an essential process for tumor growth and metastasis.3 Proangiogenic growth factors such as VEGF, PDGF, and EGF are released by tumors under hypoxic conditions. These growth factors bind to and activate their respective RTKs: VEGFR, PDGFR-β and EGFR, leading to a cascade of events that promote angiogenesis and tumor growth, survival and metastasis.4-6 RTK inhibitors that prevent angiogenesis established a new paradigm in cancer chemotherapy.7, 8 The use of multi-RTK inhibitors in cancer chemotherapy offers an advantage in overcoming resistance mechanisms that limit single RTK inhibitors including, point mutations in the ATP binding site and upregulation of additional RTKs.9 Thus, the use of multi-RTK inhibitors has emerged as an important approach in cancer chemotherapy and is validated by the approval of several multi-RTK inhibitors such as axitinib10, 11 (VEGFR, PDGFR, c-kit), pazopanib12 (PDGFR, VEGFR, c-kit, FGFR), vandetanib13 (VEGFR, EGFR, RET kinase), sunitinib14, 15 (VEGFR, PDGFR and c-kit) and sorafenib16, 17 (VEGFR-2, PDGFR β and Raf kinase), among others. The cytostatic activity of RTK inhibitors does not kill cancer cells and thus, they need to be combined with cytotoxic chemotherapeutic agents and/or radiation to improve therapeutic outcomes.18-21
Microtubules are essential cytoskeletal components in eukaryotic cells, and these cytoskeletal components are involved in cellular processes during both interphase and mitosis.22 In interphase, microtubules are involved in intracellular trafficking of proteins and organelles. In dividing cells, they make up the mitotic spindle and are essential for the separation of daughter chromosomes.22, 23 Microtubule targeting agents (MTAs) act by interfering with crucial processes involved in cell division and cellular transport in interphase.22-24
The strategy of using a combination of drugs, each acting at a different biological target, has been successfully applied in the treatment of many diseases.25, 26 The basic premise for such an approach is to overcome alternate pathways that may be activated when only one pathway is inhibited. Similarly, combination chemotherapy with antiangiogenic and cytotoxic agents act by simultaneously inhibiting multiple modes, thereby inhibiting the growth of tumors dependent on multiple signaling pathways.18-21
The vascular normalization theory proposes that the antiangiogenic effects of RTK inhibitors causes pruning of abnormal blood vessels and transiently normalizes the structural and functional integrity of the vasculature.27 This normalization restores blood flow to the tumors, improves delivery of co-administered cytotoxic agents and thus provides an explanation, in part, of the improved efficacy observed with combination chemotherapy with antiangiogenic agents.27, 28 Multi-target directed agents are rationally designed analogs that incorporate essential pharmacophores for inhibiting two or more biological targets.29 In cancer chemotherapy, such agents may help in preventing cancer cells from developing resistance, lowering the risk of drug-drug interactions, improving patient compliance and have a lower side effect profile.
Single agents with dual microtubule targeting and antiangiogenic activities are highly desirable because these agents can exert their cytotoxic effects throughout the tumor following normalization of tumor via the antiangiogenic component.30 Clinical evaluation of the safety and efficacy of metronomically dosed cytotoxic agents in conjunction with antiangiogenic agents such as sunitinib and sorafenib has also been explored.31-33 The dose-limiting cytotoxicity of conventional combination chemotherapy could also potentially be avoided because these agents work by complementary mechanisms.
Multi-target directed ligands with dual cytotoxic and antiangiogenic activities have been shown to significantly reduce tumor growth, metastasis and angiogenesis in tumor xenograft mouse models, without any significant toxicity.34, 35 Because of their crucial role in angiogenesis, VEGFR-2, PDGFR-β and EGFR were chosen as targets in the current studies. Due to the clinical utility of cytotoxic tubulin inhibitors, such as paclitaxel, in combination with antiangiogenic agents, tubulin was chosen as the cytotoxic target.36-38
Microtubule targeting agents, particularly those binding to the colchicine site, have been shown to act on tumor vasculature and several agents were tested in clinical trials for efficacy in glioblastoma multiforme (GBM),39 but none has yet advanced past phase II evaluation. Compound 1 (verubulin, Figure 1) was reported as a potent inducer of apoptosis, binding at the colchicine site and inhibiting tubulin polymerization in cells (T47D EC50 = 2 ± 0.1 nM).2 The compound is not a substrate for multidrug resistance transporters such as P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP1). In addition, compound 1 is able to cross the BBB to attain 30-fold higher concentrations in the brain compared to the plasma.2 These pharmacokinetic parameters led to the evaluation of 1 in GBM and other cancers, but failed to achieve orphan drug status for GBM.40
Figure 1.
Lead compound 1
The quinazoline scaffold has also been widely explored as the basis for RTK inhibitory activity as exemplified by gefitinib (EGFR), erlotinib (EGFR) and vandetanib (VEGFR-2, VEGFR-3 and EGFR) (Figure 2). Structure-activity relationships with the quinazoline scaffold using N4-H analogs as EGFR inhibitors suggest that the quinazoline scaffold has bulk tolerance at the 6- and 7-positions.41, 42 However, the corresponding N4-CH3 analogs were not explored for their RTK inhibitory activities. Separately, we have reported various scaffolds such as furo[2,3-d]pyrimidines34 and pyrrolo[3,2-d]pyrimidines35, 43 with N4-CH3 that are dual acting antitubulin compounds with multi-RTK inhibitory activities. Thus, on the basis of the structural similarities between 1 and the quinazoline based RTK inhibitors (Figure 2), it was of interest to determine the RTK inhibitory properties (if any) of 1 and its antiangiogenic attributes as well.
Figure 2.

Representative quinazoline based clinically used RTK inhibitors
Compound 1 was docked in the binding site of EGFR (Figure 3) and had a docked pose similar to erlotinib. The 2-CH3 group of 1 makes hydrophobic interactions with Ala719, the N1 and N3 make hinge region binding interactions with Met769 and a water mediated H-bond with Thr766, respectively. The 4-position aniline substitution is oriented in the pocket in a manner similar to the 3-ethynyl substituted aniline in erlotinib and lies in a pocket lined by residues Lys721 and Asp831. The quinazoline scaffold of 1 is stabilized by hydrophobic interactions with Leu820, Leu768 and Met769. The docked score of 1 was −8.68 kcal/mol, close to the −9.29 kcal/mol score of erlotinib.45
Figure 3.
Docked pose of lead compound 1 (purple) superimposed on the crystallized ligand erlotinib (cyan) in EGFR (PDB: 1M17)44
The docked pose of 1 in the binding site of VEGFR-2 (Figure 4)46 shows that the quinazoline scaffold binds to the pocket where the thiobenzamide of axitinib (a potent VEGFR inhibitor) binds and interacts with the backbone of the hinge binding site residue Phe1047. In addition, the quinazoline scaffold makes a cation-pi interaction with the sidechain of Lys868. The 4-position aniline substituent of 1 interacts with Val848, Cys919, Leu1035 and Phe1047 and binds in the pocket where the indazole scaffold of axitinib binds. The docked score of 1 in VEGFR-2 was −10.71 kcal/mol as compared to axitinib, which had a docked score of −13.25 kcal/mol.
Figure 4.
Docked pose of 1 (orange) superimposed on the crystallized ligand axitinib (cyan) in VEGFR-2 (PDB: 4AG8)46
With the docked scores reported above, it was important to evaluate 1 as an RTK inhibitor. As expected, a preliminary evaluation of 1 in an in vitro assay for inhibition of EGFR, VEGFR-2 and PDGFR-β resulted in potent inhibitory activities against all three RTKs (Table 1). Compound 1 was more potent than the clinically used agent sunitinib (EGFR ~61-fold, VEGFR-2 ~2-fold and PDGFR-β ~15-fold). Compared to erlotinib, 1 was 2-fold less potent in inhibiting EGFR but −15-fold and ~2-fold more potent in inhibiting VEGFR-2 and PDGFR-β, respectively. Thus, we discovered that, in addition to tubulin inhibition, 1 was a multitargeted RTK inhibitor as well.
Table 1.
EGFR, VEGFR-2 and PDGFR-β inhibitory activity of compound 1
| Compound | EGFR kinase inhibition (nM) |
Flk-1(VEGFR-2) kinase inhibition (nM) |
PDGFR-β kinase inhibition (nM) |
|---|---|---|---|
| 1 | 2.8 ± 1.1 | 8.4 ± 2.2 | 5.6 ± 0.4 |
| Sunitinib | 172.1 ± 19.4 | 18.9 ± 2.7 | 83.1 ± 10.1 |
| Erlotinib | 1.2 ± 0.2 | 124.7 ± 18.2 | 12.2 ± 1.9 |
Design of target compounds 2-11.
The reported antitubulin activity of 1, along with its RTK inhibitory activity of 1 (Table 1), provided the impetus for us to design quinazoline based multi-targeted inhibitors with both tubulin and RTK inhibitory properties in single agents. Compounds 2-4, Series I (Figure 5), with a 2-CH3 substitution, were designed to optimize the substitution at the 4-position for activity with multiple RTKs and at the colchicine site of tubulin. The clinically used quinazoline RTK inhibitors (Figure 2) all had a 2-H substitution and hence compounds in Series II (5-7) were designed to mimic the 2-position substitution of the known RTK inhibitors and to determine its effect, if any, on microtubule polymerization activity. Compounds in Series III were expected to bind at the active sites of EGFR and VEGFR-2 by maintaining the hinge region binding interactions seen for erlotinib in EGFR (Figure 3) and axitinib in VEGFR-2 (Figure 4) while retaining an interaction at the colchicine site of tubulin. In addition to EGFR and VEGFR-2, the designed compounds 2-11 were anticipated to inhibit PDGFR-β analogous to 1.
Figure 5.
Series I (2-4), Series II (5-7), Series III (8-11)
The docked pose of 5 in the colchicine site of tubulin (Figure 6) is very similar to that of lead compound 1. The quinazoline scaffold in 1 and 5 undergoes hydrophobic interactions with Alaβ250, Leuβ248, Leuβ252, Leuβ255, Ileβ318, Alaβ354, and Alaβ316. The N4-CH3 in 1 and 5 are oriented towards and interact with Lysβ254. The N4-phenyl group hydrophobically interacts with Lysβ352, Alaαl80 and Valα181. The docked score of 1 in the colchicine site of tubulin was −8.68 kcal/mol and that of 5 was −8.43 kcal/mol. Compounds in Series I (2-4) had docked scores of −8.17 to −9.83 kcal/mol and compounds in Series II (5-7) −8.06 to −8.67 kcal/mol, suggesting that these compounds should have similar or better activity than 1 as MTAs binding to the colchicine site.
Figure 6.
Comparison of the docked poses of 1 (cyan) and 5 (peach) in the colchicine site of tubulin (PDB ID: 4O2B).47
In EGFR, compound 5 (Figure 7) also undergoes a hinge region binding interaction with Met769 and a water mediated H-bond with Thr766, showing similar interactions at the binding site of EGFR as observed with 1 (Figure 3). Compounds in Series I docked in a manner similar to that of 1 and had docked scores of −7.38 to −8.90 kcal/mol. Compounds in Series II docked in a manner similar to that of 5 and had docked scores of −8.42 to −9.09 kcal/mol.
Figure 7.
Superimposition of the docked poses of 1 (magenta) and 5 (orange) in EGFR (PDB: 1M17).44
Compound 5 (Figure 8), when docked in the X-ray crystal structure of VEGFR-2,46 showed two low energy binding modes, which corroborate similar findings reported for a series of furo[2,3-d]pyrimidine analogs.34 In Figure 8A, 5 binds to VEGFR-2 in a binding mode similar to that seen for compound 1 (Figure 4) and has a docked score of −10.66 kcal/mol. In addition, due to the absence of bulk at the 2-position, 5 also binds to VEGFR-2 in an alternate binding mode where the N1 of the quinazoline scaffold in 5 undergoes H-bonding with Cys919 at the hinge region of VEGFR-2 and has a similar docking score of −9.32 kcal/mol. Compounds in Series I docked in VEGFR-2 in a manner similar to that of 1 and had docked scores of −10.71 to −12.15 kcal/mol. Compounds in Series III docked in a manner similar to 5 and had docked scores of −10.66 to −12.00 kcal/mol.
Figure 8.
Superimposition of the docked poses of 1 (orange) and 5 (magenta) in VEGFR-2 in two different binding modes (PDB: 4AG8).46
The 2-Cl substituted analog of 1 was reported to be an equipotent inhibitor of cellular microtubule polymerization (T47D EC50 = 2 ± 0.1 nM) as compared with 12 and hence, by varying the bulk and electronics at the 2-position, compounds 8-11 (Series III, Figure 5) were designed to assess the inhibitory effects on multiple RTKs. These compounds docked at the colchicine site of tubulin (docked scores, −8.06 to −9.40 kcal/mol), EGFR (docked scores, −7.59 to −8.46 kcal/mol) and VEGFR-2 (docked scores, −10.72 to −12.42 kcal/mol) in a mode similar to that shown for lead compound 1 and were predicted be active at the colchicine site and with EGFR and VEGFR-2.
Materials and Methods
Compounds 1-11 were synthesized by nucleophilic displacement of 4-chloroquinazolines (12a, 12b and 12c) by the appropriate anilines in isopropyl alcohol/acetonitrile and a drop of conc. HCl at room temperature for 12 h (Scheme 2).
Scheme 2.


Synthesis of target compounds 1-11 (Series I, II and III)
Yields of compounds in Series I were 58% (1, reported yield 71%),2 65% (2, reported yield 79%),48 70% (3, reported yield 41%),2 68% (4). Yields of compounds in Series II were 76% (5, reported yield 79%),1 70% (6, reported yield 69%),49 and 72% (7). Yields of compounds in Series III were 9% (8, reported yield 87%),1 25% (9, reported yield 87%).48 The synthesis of 10 was unsuccessful under the reaction conditions described above, but instead dimer 18 was formed (1H NMR indicated four methyl singlets, two for SCH3 at 2.48 and 2.49 ppm and two for NCH3 at 3.29 and 3.57 ppm). Alternatively, a SNAr reaction in acetonitrile at rt gave 10 in 22% yield. Compound 11 was synthesized over two steps. The first step involved the nucleophilic displacement of quinazoline 12c with 6-aminonaphthalen-1-ol (19) to yield 6-((2-chloroquinazolin-4-yl)amino)naphthalen-1-ol (20). N- and O-methylation of 20 using methyl iodide and sodium hydride yielded the desired compound 11 in 47% yield.
To synthesize N-methylated aniline 16 (Scheme 3), commercially available aniline 21 was formylated using paraformaldehyde in the presence of sodium methoxide in methanol. Subsequent reduction of the N-formyl group using NaBH4 yielded aniline 16 (39%).50 To synthesize the aniline 17 from 6-aminonaphthalen-1-ol (19), both N- and O-methylation, were required, and a different strategy was necessary. 6-Aminonaphthalen-1-ol (19) was first Boc protected to give the N-Boc protected intermediate 22, which was subsequently dimethylated (both N- and O-) with methyl iodide in the presence of sodium hydride51 and deprotected using trifluoroacetic acid to afford 5-methoxy-N-methylnaphthyl-2-amine (17) (55% over 3 steps).52
Scheme 3.

Synthesis of anilines 16 and 17
Evidence of conformational restriction of 1 by 1H NMR.
Comparison of the 1H NMR spectra of compound 1 with the desmethyl analog 1a2 in DMSO-d6 suggested that the N-Me group imparts conformational restriction along the C4-N bond. In 1, the 5-H proton appears at δ 6.88, however in compound 1a, the 5-H proton is more deshielded and appears at δ 8.06 (Figure 9). The shielding effect of the 5-H can be attributed to a diamagnetic anisotropic effect in 1 due to the proximity of the phenyl ring on the 5-H proton in the more favorable conformation. The same effect, however, does not occur in the less favorable conformation since the phenyl ring lies anti to the 5-H and leads to steric clashes between the N-Me and 5-H proton groups. The diamagnetic anisotropy is also not observed for the conformationally flexible analog 1a, and the 5-H proton appears more deshielded.
Figure 9.
Conformational restriction of C4-N observed for 1 due to presence of the N-Me group.
Results and Discussion
Effect on cellular microtubules and cancer cell proliferation.
Compounds 2–11 were evaluated for their ability to depolymerize microtubules in A-10 cells with a phenotypic assay to evaluate microtubule loss. An EC50, the concentration that causes 50% cellular microtubule loss, was determined. The antiproliferative effects were evaluated in MDA-MB-435 melanoma cells using the sulforhodamine B (SRB) assay, and the IC50, the concentration that causes 50% inhibition of proliferation, was determined. The activities of the compounds were compared with 1, paclitaxel and CA-4, a small molecule that binds the colchicine site of β-tubulin at its interface with α-tubulin (Table 2).
Table 2.
IC50 values for inhibition of proliferation of MDA-MB-435 cells and EC50 values for microtubule depolymerization in A-10 cells.
| Compound | IC50 ± SD in MDA- 435 Cells (nM) |
EC50 (nM) for Microtubule Depolymerization in A·10 Cells |
EC50/IC50 Ratio |
CLogP |
|---|---|---|---|---|
| 1 | 1.7 ± 0.1 | 2.1 | 1.2 | 4.2 |
| 2 | 1.1 ± 0.2 | 3 | 2.7 | 4.8 |
| 3 | 1.2 ± 0.2 | 2 | 1.7 | 4.8 |
| 4 | 2.0 ± 0.4 | 2.4 | 1.2 | 5.3 |
| 5 | 7.1 ± 0.8 | 8.2 | 1.2 | 3.7 |
| 6 | 12.4 ± 1.7 | 23.4 | 1.9 | 4.3 |
| 7 | 7.8 ± 0.8 | 12.3 | 1.6 | 4.3 |
| 8 | 0.6 ± 0.1 | 2 | 3.3 | 4.4 |
| 9 | 0.7 ± 0.2 | 2 | 2.9 | 5.1 |
| 10 | 2.7 ± 0.5 | 2.7 | 1 | 5.1 |
| 11 | 1.4 ± 0.1 | 1.9 | 1.4 | 5.6 |
| CA-4 | 4.4 ± 0.46 | 9.8 | 2.2 | |
| Paclitaxel | 4.5 ± 0.52 | - | - |
CLogP values were calculated using ChemDraw Professional 2018
In the antiproliferative assay (Table 2), the potency of the compounds followed the trend: 2-Cl (8-11) > 2-CH3 (1-4) > 2-H (5-7) substitution, suggesting that small hydrophobic groups at the 2-position could contribute to improved cellular permeability as indicated by the CLogP values (Table 2). In addition, small hydrophobic groups at the 2-position were tolerated for binding at the colchicine site. In the microtubule depolymerization assay, compounds from Series I (1-4) and Series III (8-11) were equipotent and were 4- to 12-fold more potent than Series II (5-7) compounds with 2-H substitution. For compounds 1-11, the EC50/IC50 ratios (Table 2) are close to 1.0, suggesting a cytotoxic mechanism of action that is primarily microtubule dependent.
Clinically relevant drug resistance mechanisms associated with most clinically approved microtubule targeting agents, include expression of either the β-III isotype of tubulin or Pgp, an ATP-dependent efflux pump.53 Compounds 1-11 were tested for their efficacy in parental cells and β-III and Pgp expressing cell lines (Table 3). In β-III overexpressing HeLa cells, compounds 1-11 were able to all overcome β-III mediated resistance compared with paclitaxel and were comparable to CA-4 in their activity. The resistance ratio (Rr) values (Table 3) were 0.6-1.2 versus 1.0 for CA-4 and 8.6 for paclitaxel.
Table 3.
Compound activity in parental, β-III and Pgp expressing cell lines
| Compound | IC50 ± SD in HeLa (nM) |
IC50 ± SD in WT βIII (nM) |
Rr Value (WT/HeLa) |
IC50 ± SD in SK-OV-3 Cells (nM) |
IC50 ± SD in SK-OV- 3-MDR-1- 6/6 Cells (nM) |
Rr Value (MDR- 6/6/SK- OV-3) |
|---|---|---|---|---|---|---|
| 1 | 2.0 ± 0.2 | 1.5 ± 0.2 | 0.8 | 2.2 ± 0.1 | 2.6 ± 0.1 | 1.2 |
| 2 | 1.2 ± 0.5 | 1.2 ± 0.4 | 1.0 | 1.5 ± 0.3 | 2.2 ± 0.6 | 1.5 |
| 3 | 1.3 ± 0.6 | 1.1 ± 0.4 | 0.8 | 1.6 ± 0.2 | 2.1 ± 1.1 | 1.3 |
| 4 | 2.5 ± 0.3 | 1.7 ± 0.2 | 0.7 | 2.9 ± 0.3 | 3.5 ± 0.4 | 1.2 |
| 5 | 8.6 ± 0.3 | 7.4 ± 0.7 | 0.9 | 10.4 ± 1.1 | 14.3 ± 0.5 | 1.4 |
| 6 | 16.8 ± 2.2 | 13.1 ± 0.4 | 0.8 | 18.1 ± 1.8 | 23.4 ± 4.5 | 1.3 |
| 7 | 8.5 ± 0.8 | 7.3 ± 0.4 | 0.6 | 13.6 ± 0.9 | 15.6 ± 1.4 | 1.1 |
| 8 | 0.6 ± 0.2 | 0.6 ± 0.1 | 1.0 | 0.8 ± 0.1 | 1.1 ± 0.3 | 1.4 |
| 9 | 0.6 ± 0.2 | 0.7 ± 0.2 | 1.2 | 0.8 ± 0.2 | 1.0 ± 0.5 | 1.3 |
| 10 | 5.0 ± 0.4 | 3.2 ± 0.2 | 0.6 | 3.3 ± 0.1 | 3.5 ± 0.6 | 1.1 |
| 11 | 2.8 ± 0.5 | 1.6 ± 0.3 | 0.6 | 2.8 ± 0.4 | 2.6 ± 0.4 | 0.9 |
| CA-4 | 3.3 ± 0.4 | 3.3 ± 0.3 | 1.0 | 5.5 ± 0.5 | 7.2 ± 1.1 | 1.3 |
| Paclitaxel | 2.8 ± 0.4 | 24.0 ± 0.2 | 8.6 | 5.0 ± 0.6 | 1200 ± 58 | 240 |
Paclitaxel is a known substrate for Pgp, and cells expressing Pgp are resistant to its effects, resulting in a 240-fold loss in activity (Table 3). Compounds 1-11 were evaluated for their ability to circumvent Pgp-mediated drug resistance in the SK-OV-3 isogenic cell line pair. As expected, the compounds showed activity similar to that of CA-4 and were essentially equipotent in the parental and Pgp-expressing cells (Rr values 0.9-1.5). In contrast, paclitaxel had a Rr value of 240, indicating its susceptibility to Pgp-mediated resistance.
Selected quinazoline derivatives, on the basis of their activities in Tables 2 and 3, were also tested for their activity as inhibitors of tubulin assembly and their ability to inhibit the binding of [3H] colchicine to tubulin. The compounds also had potencies similar to that of CA-4 for inhibition of colchicine binding in both the assays.
Evaluation of compounds 1-4, 6, 8-9 in EGFR, VEGFR-2 and PDGFR-β assays suggested that these compounds were potent inhibitors of multiple RTKs (Table 5). With respect to the structure-activity relationship, within Series I, the 4-methoxy-N-methylaniline substitution (1) was equipotent with the tetrahydroquinoline substitution (2) and 3-fold better than 3 and 4 with EGFR. For VEGFR-2, compounds 1-4 had similar inhibitory IC50 values, suggesting the VEGFR-2 binding site is more accommodating towards introduction of bulk at the 4-position of the quinazoline scaffold. With PDGFR-β, 1 and 2 has similar activity, but, introduction of a thiomethyl group in 3 results in ~ 41-fold loss of potency and ~ 5-fold loss in potency for compound 4 with PDGFR-β. Comparing the tetrahydroquinoline compounds 2, 6 and 9 across Series I-III, the 2-H substituted compound 6 resulted in ~7-10-fold loss in potency for all the RTKs and also a similar loss of activity in the cytotoxicity assay, suggesting the presence of bulk at the 2- position is important for activity in RTKs. 2-CH3 and 2-Cl substitution (2 and 9) did not affect EGFR activity but results in ~6-10-fold loss of potency for VEGFR-2 and PDGFR- β, respectively.
Table 5.
RTK inhibitory activities of quinazolines (1-4, 6, 8-9)
| Compound | EGFR kinase inhibition (nM) |
Flk-1 (VEGFR-2) kinase inhibition (nM) |
PDGFR-β kinase inhibition (nM) |
A431 Cytotoxicity (nM) |
U251 cytotoxicity (nM) |
CAM angiogenesis inhibition (μM) |
|---|---|---|---|---|---|---|
| 1 | 2.8 ± 1.1 | 8.4 ± 2.2 | 5.6 ± 0.4 | 0.9 ± 0.2 | 2.8 ± 0.6 | 2.8 ± 1.1 |
| 2 | 5.0 ± 0.4 | 9.3 ± 3.9 | 7.8 ± 0.5 | 2.5 ± 0.2 | 3.1 ± 0.3 | 3.1 ± 1.3 |
| 3 | 8.7 ± 1.6 | 14.7 ± 2.8 | 231.5 ± 29.9 | 2.9 ± 0.5 | 4.9 ± 0.9 | 9.8 ± 0.7 |
| 4 | 10.6 ± 1.8 | 16.8 ± 4.1 | 29.4 ± 5.1 | 5.3 ± 0.9 | 4.2 ± 2.0 | ND |
| 6 | 48.9 ± 6.0 | 111.1 ± 19.2 | 54 ± 14.1 | 16.3 ± 2.0 | 27.8 ± 1.8 | ND |
| 8 | 3.6 ± 0.6 | 97.5 ± 10.1 | 76.6 ± 16.2 | 1.2 ± 0.4 | 32.5 ± 4.1 | 130.0 ± 22.2 |
| 9 | 5.4 ± 0.3 | 55.2 ± 11.1 | 80.6 ± 12.1 | 1.8 ± 0.1 | 14.8 ± 1.8 | 41.4 ± 6.1 |
| Sunitinib | 172.1 ± 19.4 | 18.9 ± 2.7 | 83.1 ± 10.1 | - | - | - |
| Erlotinib | 1.2 ± 0.2 | 124.7 ± 18.2 | 12.2 ± 1.9 | - | - | 29.1 ± 1.9 |
ND-Not Determined
In addition, comparing the 2-position quinazoline substitution across Series I, II and III, for EGFR 2-CH3 and 2-Cl substitution were ~ 10-fold better than the 2-H substitution. For VEGFR-2 binding, 2-CH3 was 6-fold better than the 2-Cl substitution and ~ 12-fold better than the 2-H substitution. In contrast, for PDGFR-β, the 2-H substitution was 1.5-fold better than the 2-Cl substitution, which in turn was 7-fold less potent than the 2-CH3 substitution.
Moreover, compounds 1-3 were 3- to 10-fold more potent than erlotinib in the CAM assay for angiogenesis inhibition (Table 5). However, compounds in Series III were 1- to 4-fold less potent than erlotinib in inhibiting angiogenesis.
Conclusion
Substituted quinazolines have been explored as a scaffold for targeting RTKs as well as microtubules. Here we describe the design, synthesis and evaluation of 4-anilino quinazolines with multiple modes of action contributing to their antiproliferative activities in cancer cells. Compounds were synthesized by varying substitutions at the 2- and 4- positions of the quinazoline scaffold. For the MTA effects in general, the 2-H substituted compounds (5-7) were 7- to 10-fold less potent than the 2-CH3 (1-4) and the 2-Cl (8-9) substituted compounds. This suggests that bulk at the 2-position of the quinazoline scaffold is important for binding to the colchicine site. In the RTK inhibitory assays, the 2-CH3 and 2-Cl substituted compounds were more potent than the 2-H substituted compounds. For 2-substituted quinazolines (1-4), varying the aniline substitution at the 4-position was well tolerated. The 2-Cl substituted compound 8 was a potent inhibitor of EGFR but resulted in a 10-fold and 15-fold loss of potency with VEGFR-2 and PDGFR-β, respectively, when compared to the corresponding 2-CH3 compound 1. Our studies establish that the quinazoline based analogs are indeed single agents with multitargeted activities, and that they inhibit both tubulin and RTKs and have potential as anticancer agents.
Experimental Section
Chemistry.
All evaporations were carried out under vacuum with a rotary evaporator. Analytical samples were dried in vacuo in a CHEM-DRY drying apparatus over P2O5 at 50 °C. Melting points were determined either using a MEL-TEMP II melting point apparatus with FLUKE 51 K/J electronic thermometer or using an MPA100 OptiMelt automated melting point system and are uncorrected. Thin-layer chromatography (TLC) was performed on Whatman® PE SIL G/UV254 flexible silica gel plates or Sorbetch silica gel TLC plates w/UV254, and the spots were visualized under 254 and 365 nm ultraviolet illumination. Proportions of solvents used for TLC are by volume. All analytical samples were homogeneous on TLC in at least two different solvent systems. Flash chromatography was carried out on the CombiFlash® Rf 200 (Teledyne ISCO) automated flash chromatography system with prepacked RediSep® Rf normal-phase flash columns (230 to 400 mesh) of various sizes. The weight of silica gel for column chromatography was in the range of 50-100 times the weight of the crude compounds being separated. Nuclear magnetic resonance spectra for proton (1H NMR) were recorded on the Bruker Avance II 400 (400 MHz) or Bruker Avance II 500 (500 MHz) NMR systems and were analyzed using MestReC NMR data processing software. The chemical shift values (δ) are expressed in ppm (parts per million) relative to tetramethylsilane as an internal standard: s, singlet; d, doublet; dd, doublet of doublet; t, triplet; q, quartet; m, multiplet; br, broad singlet; td, triplet of doublet; dt, doublet of triplet; quin, quintet. Elemental analyses were performed by Atlantic Microlab, Inc., Norcross, GA. Element compositions are within ± 0.4% of the calculated values. Fractional moles of water or organic solvents frequently found in some analytical samples could not be prevented despite 24 to 48 hours of drying in vacuo and were confirmed where possible by their presence in the 1H NMR spectra. All solvents and chemicals were purchased from Sigma-Aldrich or Fisher Scientific and were used as received.
N-Methyl-4-(methylthio)aniline (16)
4-(Methylthio)aniline (21) (1.13 g, 8.12 mmol) was added to a suspension of sodium methoxide (2.19 g, 40.6 mmol) in MeOH (10 mL). This mixture was poured into another suspension of paraformaldehyde (0.340 g, 11.4 mmol) in MeOH (10 mL). The reaction mixture was then stirred at room temperature. After 5 h, sodium borohydride (0.31 g, 8.1 mmol) was added, and the mixture was kept at reflux for 2 h. After the reaction was complete, the solvent was evaporated and 1 M KOH (10 mL) was added. The sample was then extracted 3 times using ether and dried over MgSO4. The combined organic fractions were evaporated, a silica gel plug was made, and the product was purified by column chromatography using 10% EtOAc in hexanes to yield 16 as a brown liquid (0.49 g, 39%). TLC Rf = 0.76 (Hex: EtOAc; 1:1); 1H NMR, DMSO-d6 (400 MHz): 2.33 (s, 3 H, SCH3), 2.64-2.66 (d, 3 H, J = 5.09, NHCH3), 5.72-5.73 (m, 1 H, exch, NH), 6.49-6.52 (d, 2 H, J = 8.58, Ar), 7.11-7.13 (d, 2 H, J = 8.57, Ar). 1H NMR agreed well with the literature54 values.
5-Methoxy-N-methylnaphthalen-2-amine (17)
Compound 17 was prepared over 3 steps. 6-Aminonaphthalen-1-ol (19) (1.25 g, 7.85 mmol) was stirred with boc anhydride (5 mL) in ethyl acetate (15 mL) at room temperature. After 12 h, boc anhydride was removed by evaporation, and the residue was dissolved in ethyl acetate and purified using flash chromatography in CHCl3/MeOH to afford 22 as a brown liquid (1.74 g, 85%). TLC Rf = 0.67 (CH3OH: CHCl3; 1:5); 1H NMR, DMSO-d6 (400 MHz): 1.51 (s, 9 H, (CH3)3), 6.71-6.73 (dd, 1 H, J1 = 7.13, J2 = 1.21, Ar), 7.17-7.25 (m, 2 H, Ar), 7.43-7.46 (dd, 1 H, J1 = 1.97, J2 = 9.14, Ar), 8.00-8.01 (d, 2 H, J = 8.87, Ar), 9.54 (s, 1 H, exch., OH), 10.00 (s, 1 H, exch., NH). Compound 22 (1.70 g, 6.79 mmol) was dissolved in DMF (30 mL) under argon. Sodium hydride (0.488 g, 20.4 mmol) was added, and the mixture was stirred at 0 °C for 15 min. Methyl iodide (2.12 g, 14.9 mmol) was added, and the reaction mixture was stirred at room temperature for 2 h. At the end of the reaction, solvent was evaporated, silica gel was added with methanol to make a plug, and the mixture was purified using flash chromatography with hexanes/ethyl acetate to afford 23 (1.36 g, 72%) as a yellow-colored liquid. TLC Rf = 0.8 (Hex: EtOAc; 1:1); 1H NMR, DMSO-d6 (400 MHz): 1.41 (s, 9 H, (CH3)3), 3.28 (s, 3 H, NCH3), 3.96 (s, 3 H, OCH3), 6.92-6.94 (m, 1 H, Ar), 7.42-7.43 (m, 2 H, Ar), 7.45-7.46 (d, 1 H, J = 2.19, Ar), 7.72-7.73 (d, 1 H, J = 2.12, Ar), 8.07-8.09 (d, 1 H, J = 9.03, Ar). Boc deprotection of 23 (1.3 g, 4.5 mmol) was carried out in trifluoroacetic acid (TFA, 15 ml) at room temperature for 2 h. At the end of the reaction, TFA was removed by evaporation, and the mixture was neutralized using an aq. solution of K2CO3. Extraction with ethyl acetate (20 mL × 3), followed by concentration and drying over Na2SO4 yielded 17 (0.810 g, 95%) as a green-colored solid. TLC Rf = 0.77 (Hex: EtOAc; 1:1); 1H NMR, DMSO-d6 (400 MHz): 2.75-2.76 (d, 3 H, NCH3), 3.89 (s, 3 H, OCH3), 5.98-5.99 (q, 1 H, NHCH3, exch.), 6.57-6.59 (d, 1 H, J = 7.43, Ar), 6.61-6.62 (d, 1 H, J = 1.87, Ar), 6.88-6.90 (dd, 1 H, J1 = 1.42, J2 = 9.05, Ar), 7.15-7.23 (m, 2 H, Ar), 7.83-7.86 (d, 1 H, J = 9.05, Ar). The compound was carried to the next step without further purification.
4-Chloro-2-methylquinazoline (12a)
To a 250 mL round-bottomed flask were added 2-methylquinazolin-4(3H)-one (13a) (1.0 g, 6.2 mmol), trimethylamine (3 mL), and toluene (30 mL). The mixture was cooled to 0 °C, and phosphorus oxychloride (2 mL) was added. The reaction mixture was stirred at room temperature for 1 h. The reaction was then heated to 95 °C for 3 h. After monitoring the reaction by TLC, the reaction mixture was cooled to room temperature and diluted with 30 mL of ethyl acetate. The solution was then washed with 10 mL of ice cold water, 10 mL of saturated NaHCO3, 10 mL of water, 5 mL of 1 N HCl, 10 mL of water, 10 mL of saturated NaHCO3 and 10 mL of saturated NaCl. The organic layer was dried over Na2SO4, filtered and concentrated to obtain 12a as a yellow solid (0.600 g, 54%). TLC Rf = 0.8 (MeOH:CHCl3; 1:5); mp, 83.9-85.3 °C (lit.55 85-86 °C); 1H NMR, DMSO-d6 (400 MHz): 2.58 (s, 3 H, CH3), 7.59-7.63 (t, 1 H, J1 = 7.58, J2 = 7.58, Ar), 7.78-7.80 (d, 1 H, J = 7.98, Ar), 7.90-7.95 (t, 1 H, J1 = 7.67, J2 = 7.67, Ar), 8.13-8.15 (d, 1 H, J = 7.65, Ar).
4-Chloroquinazoline (12b)
To a 250 mL round-bottomed flask were added quinazolin-4(3H)-one (13b) (2.00 g, 13.7 mmol), trimethylamine (3 mL), and toluene (30 mL). The mixture was cooled to 0 °C, and phosphorus oxychloride (2 mL) was added. The reaction mixture was stirred at room temperature for 1 h. The reaction was then heated to 95 °C for 3 h. After monitoring the reaction by TLC, the reaction mixture was cooled to room temperature and diluted with 30 mL of ethyl acetate. The solution was then washed with 10 mL of ice cold water, 10 mL of saturated NaHCO3, 10 mL of water, 5 mL of 1 N HCl, 10 mL of water, 10 mL of saturated NaHCO3 and 10 mL of saturated NaCl. The organic layer was dried over Na2SO4, filtered and concentrated to obtain 12b as a yellow solid (1.79 g, 80%). TLC Rf = 0.85 (MeOH:CHCl3; 1:5); mp, 144.2-146.1 °C (lit.56 145-147.5 °C); 1H NMR, DMSO-d6 (400 MHz): 7.59-7.63 (dt, 1 H, J1 = 1.09, J2 = 7.23, J3 = 8.01, Ar), 7.76-7.78 (d, 1 H, J = 8.24, Ar), 7.89-7.92 (dt, 1 H, J1 = 1.47, J2 = 7.24, J3 = 8.29, Ar), 8.15-8.17 (d, 1 H, J = 7.03, Ar), 8.58 (s, 1 H, Ar).
General method for the synthesis of compounds 1-10
To a 100 mL round-bottomed flask substituted-4-chloroquinazolines (1 equivalent), appropriate anilines (1.2 equivalents) and isopropanol (10 mL) were added and stirred at room temperature for 12 h. The compounds precipitated as HCl salts, which were filtered and dried in vacuo over P2O5 to afford the desired compounds.
N-(4-Methoxyphenyl)-N,2-dimethylquinazolin-4-amine (1)
Using the general method described above, 12a (0.150 g, 0.840 mmol) and 4-methoxy-N-methylaniline (14) (0.138 g, 1.01 mmol) were reacted for 12 h. At the end of the reaction, solvent was evaporated, silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified by flash chromatography using chloroform/methanol to afford 1 as a brown solid (0.136 g, 58%). TLC Rf = 0.8 (MeOH:CHCl3; 1:5); mp, 99.1-101.2 °C; 1H NMR, DMSO-d6 (400 MHz): 2.66 (s, 3 H, 2-CH3), 3.60 (s, 3 H, NCH3), 3.81 (s, 3 H, OCH3), 6.88-6.90 (d, 1 H, J = 8.35, Ar), 7.04-7.06 (d, 2 H, J = 8.89, Ar), 7.14-7.18 (t, 1 H, J1 = 7.49, J2 = 7.49, Ar), 7.30-7.32 (d, 2 H, J = 8.78, Ar), 7.68-7.75 (m, 2 H, Ar). Anal. Calcd. for C17H17N3O .0.52 HCl: C, 68.46; H, 5.92; N, 14.08. Found C, 68.50; H, 6.24; N, 13.80; Cl, 4.54.
4-(6-Methoxy-3,4-dihydroquinolin-1(2H)-yl)-2-methylquinazoline (2)
Using the general method described above, 12a (0.150 g, 0.840 mmol) and 6-methoxy-1,2,3,4-tetrahydroquinoline (15) (0.164 g, 1.01 mmol) were reacted to afford 2 as a yellow precipitate (0.188 g, 65%). TLC Rf = 0.85 (MeOH:CHCl3; 1:5); mp, 255.8-257.9 °C; 1H NMR, DMSO-d6 (400 MHz): 2.1 (quin, 2 H, CH2), 2.75 (s, 3 H, 2-CH3), 2.87-2.90 (t, 2 H, CH2), 3.79 (s, 3 H, OCH3), 4.20 (t, 2 H, NCH2), 6.68-6.71 (dd, 1 H, J1 = 2.88, J2 = 8.85, Ar), 7.02-7.03 (d, 1 H, J = 2.81, Ar), 7.05-7.08 (d, 1 H, J = 8.87, Ar), 7.26-7.28 (d, 1 H, J = 8.53, Ar), 7.36-7.40 (dt, 1 H, J1 = 1.38 , J2 = 6.83, J3 = 8.28, Ar), 7.88-7.96 (m, 2 H, Ar). Anal. Calcd. for C19H19N3O. 1.07HCl: C, 66.26; H, 5.87; N, 12.20. Found C, 66.28; H, 5.75; N, 12.11; Cl, 10.49.
N,2-Dimethyl-N-(4-(methylthio)phenyl)quinazolin-4-amine (3)
Using the general method described above, 12a (0.135 g, 0.756 mmol) and N-methyl-4-(methylthio)aniline (16) (0.135 g, 0.907 mmol) were reacted to afford 3 as a white precipitate (0.175 g, 70%). TLC Rf = 0.9 (MeOH:CHCl3; 1:5); mp, 235.2-236.4 °C; 1H NMR, DMSO-d6 (400 MHz): 2.55 (s, 3 H, 2-CH3), 2.77 (s, 3 H, SCH3), 3.74 (s, 3 H, NCH3), 6.87-6.89 (d, 1 H, J = 8.39, Ar), 7.30-7.34 (dt, 1 H, J1 = 1.33, J2 = 7.0, J3 = 8.48, Ar), 7.43-7.50 (m, 4 H, Ar), 7.85-7.89 (dt, 1 H, J1 = 1.14, J2 = 7.05, J3 = 8.34, Ar), 7.93-7.95 (d, 1 H, J = 8.32, Ar). Anal. Calcd. for C17H17N3S. 1.34 HCl: C, 59.28; H, 5.37; N, 12.20; S, 9.31. Found C, 59.37; H, 5.62; N, 11.93; S, 9.03; Cl, 10.55.
N-(5-Methoxynaphthalen-2-yl)-N,2-dimethylquinazolin-4-amine (4)
Using the general method described above, 12a (0.135 g, 0.840 mmol) and 5-methoxy-N-methylnaphthalen-2-amine (17) (0.156 g, 0.831 mmol) were reacted for 12 h. At the end of the reaction, solvent was evaporated, and silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified using flash chromatography using chloroform/methanol to afford 4 as a yellow solid (0.170 g, 68%). TLC Rf = 0.9 (MeOH:CHCl3; 1:10); mp, 203.0-204.4 °C; 1H NMR, DMSO-d6 (400 MHz): 2.70 (s, 3 H, 2-CH3), 3.72 (s, 3 H, NCH3), 3.99 (s, 3 H, OCH3), 6.91-6.93 (d, 1 H, J = 8.26, Ar), 7.00-7.02 (d, 1 H, J = 7.46, Ar), 7.05-7.09 (t, 1 H, J1 = 7.70, J2 = 7.70, Ar), 7.40-7.49 (m, 3 H, Ar), 7.66-7.69 (t, 1 H, J1 = 7.59, J2 = 7.59, Ar), 7.74-7.76 (d, 1 H, J = 8.33, Ar), 7.84 (s, 1 H, Ar), 8.21-8.23 (d, 1 H, J = 8.99, Ar). Anal. Calcd. for C21H19N3O. 0.41 HCl: C, 73.22; H, 5.68; N, 12.20. Found C, 73.19; H, 5.82; N, 12.35; Cl, 2.22.
N-(4-Methoxyphenyl)-N-methylquinazolin-4-amine (5)
Using the general method described above, 12b (0.150 g, 0.911 mmol) and N-methyl-4-methoxyaniline (14) (0.150 g, 1.09 mmol) were reacted to afford 5 as a pale-yellow precipitate (0.210 g, 76%). TLC Rf = 0.8 (MeOH:CHCl3; 1:5); mp, 244.3-245.3 °C; 1H NMR, DMSO-d6 (400 MHz): 3.75 (s, 3 H, NCH3), 3.84 (s, 3 H, OCH3), 6.84-6.86 (d, 1 H, J = 8.43, Ar), 7.12-7.16 (d, 2 H, J = 8.97, Ar), 7.32-7.36 (dt, 1 H, J1 = 1.12, J2 = 6.93, J3 = 8.45, Ar), 7.46-7.50 (d, 2 H, J = 8.96, Ar), 7.87-7.91 (dt, 1 H, J1 = 1.05, J2 = 6.94, J3 = 8.25, Ar), 7.95-7.97 (d, 1 H, J = 8.15, Ar), 9.06 (s, 1 H, Ar). Anal. Calcd. for C16H15N3O. 1.10 HCl: C, 62.89; H, 5.31; N, 13.75. Found C, 62.89; H, 5.33; N, 13.75; Cl, 11.43.
4-(6-Methoxy-3,4-dihydroquinolin-1(2H)-yl)quinazoline (6)
Using the general method described above, 12b (0.150 g, 0.911 mmol) and 6-methoxy-1,2,3,4-tetrahydroquinoline (15) (0.179 g, 1.09 mmol) were reacted to afford 6 as a yellow precipitate (0.211 g, 70%). TLC Rf = 0.8 (MeOH:CHCl3; 1:5); mp, 234.2-235.3 °C; 1H NMR, DMSO-d6 (400 MHz): 2.05-2.13 (quint, 2 H, CH2), 2.87-2.91 (t, 2 H, CH2), 3.79 (s, 3 H, OCH3), 4.20-4.21 (t, 2 H, NCH2), 6.67-6.70 (dd, 1 H, J1 = 2.91, J2 = 8.85, Ar), 7.02-7.03 (d, 1 H, J = 2.82, Ar), 7.08-7.10 (d, 1 H J = 8.85, Ar), 7.33-7.35 (d, 1 H, J = 8.50, Ar), 7.42-7.46 (dt, 1 H, J1 = 1.55, J2 = 6.67, J3 = 8.32, Ar), 7.91-7.98 (m, 2 H, Ar), 9.04 (s, 1 H, Ar). Anal. Calcd. for C18H17N3O. 1.22 HCl: C, 64.38; H, 5.47; N, 12.51. Found C, 64.39; H, 5.47; N, 12.45; Cl, 10.41.
N-Methyl-N-(4-(methylthio)phenyl)quinazolin-4-amine (7)
Using the general method described above, 12b (0.150 g, 0.911 mmol) and N-methyl-4-(methylthio)aniline (16) (0.168 g, 1.09 mmol) were reacted to afford 7 as a pale yellow precipitate (0.209 g, 72%). TLC Rf = 0.8 (MeOH:CHCl3; 1:5); mp, 233.4-235.9 °C; 1H NMR, DMSO-d6 (400 MHz): 2.54 (s, 3 H, SCH3), 3.75 (s, 3 H, NCH3), 6.94-6.96 (d, 1 H, J = 8.74, Ar), 7.35-7.39 (t, 1 H, J1 = 7.69, J2 = 7.69, Ar), 7.43-7.45 (d, 2 H, J = 8.70, Ar), 7.48-7.50 (d, 2 H, J = 8.68, Ar), 7.88-7.92 (t, 1 H, J1 = 7.55, J2 = 7.55, Ar), 7.95-7.97 (d, 1 H, J = 7.68, Ar), 9.08 (s, 1 H, Ar). Anal. Calcd. for C16H15N3S. 1.13 HCl: C, 59.59; H, 5.04; N, 13.03; S, 9.94. Found C, 59.56; H, 5.13; N, 12.97; S, 10.22; Cl, 10.22.
2-Chloro-N-(4-methoxyphenyl)-N-methylquinazolin-4-amine (8)
Using the general method described above, 12c (0.500 g, 2.51 mmol) and 4-methoxy-N-methylaniline (14) (0.413 g, 3.01 mmol) were reacted for 12 h. At the end of reaction, solvent was evaporated, silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified using flash chromatography using chloroform/methanol to afford 8 as buff-colored solid (0.070 g, 9%). TLC Rf = 0.95 (MeOH:CHCl3; 1:5); mp, 146.3-148.7 °C; 1H NMR, DMSO-d6 (400 MHz): 3.52 (s, 3 H, NCH3), 3.81 (s, 3 H, OCH3), 6.86-6.88 (d, 1 H, Ar), 7.03-7.06 (d, 2 H, J = 8.96, Ar), 7.11-7.16 (m, 1 H, Ar), 7.32-7.35 (d, 2 H, J = 8.95, Ar), 7.66-7.67 (m, 2 H, Ar). Anal. Calcd. for C16H14C1N3O: C, 64.11; H, 4.71; N, 14.02; Cl, 11.83. Found C, 64.08; H, 4.77; N, 13.93; Cl, 11.97.
2-Chloro-4-(6-methoxy-3,4-dihydroquinolin-1(2H)-yl)quinazoline (9)
Using the general method described above, 12c (0.300 g, 1.51 mmol) and 6-methoxy-1,2,3,4-tetrahydroquinoline (15) (0.295 g, 1.81 mmol) were reacted for 12 h. At the end of the reaction, solvent was evaporated, silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified by flash chromatography using chloroform/methanol to afford 9 as a yellow solid (0.121 g, 25%). TLC Rf = 0.97 (MeOH:CHCl3; 1:5); mp, 135.9-137.0 °C (lit.48 136-138 °C); 1H NMR, DMSO-d6 (400 MHz): 2.13-2.18 (m, 2 H, CH2), 2.88-2.91 (t, 2 H, CH2), 3.85 (s, 3 H, OCH3), 4.11-4.16 (m, 2 H, CH2), 6.59-6.62 (dd, 1 H, J1 = 2.89, J2 = 8.81, 7´-Ar), 6.75-6.77 (d, 1 H, J = 8.85, 8´-Ar), 6.857-6.863 (d, 1 H, J = 2.86, 5´-Ar), 7.16-7.19 (m, 1 H, J1 = 1.26, J2 = 6.96, J3 = 8.43, Ar), 7.31-7.33 (dd, 1 H, J1 = 0.90, J2 = 8.53, Ar), 7.68-7.71 (m, 1 H, J1 = 1.36, J2 = 6.97, J3 = 8.39, Ar), 8.05-8.07 (d, 1 H, Ar). Anal. Calcd. for C18H16ClN3O . 0.24 (CH3)2CHOH: C, 66.08; H, 5.31; N, 12.34; Cl, 10.41. Found C, 66.30; H, 5.19; N, 12.35; Cl, 10.17.
2-Chloro-N-methyl-N-(4-(methylthio)phenyl)quinazolin-4-amine (10)
Using the general procedure described above, 12c (0.300 g, 1.51 mmol) and 16 (0.23 g, 1.5 mmol) were reacted in acetonitrile for 12 h. At the end of reaction, solvent was evaporated, silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified by flash chromatography using chloroform/methanol to afford 10 (0.1 g, 22%) as a dark yellow solid. TLC Rf = 0.76 (MeOH:CHCl3; 1:5); mp, 123.6 °C, 1H NMR, CDCl3 (400 MHz): 2.55 (s, 3 H, SCH3), 3.68 (s, 3 H, NCH3), 6.97-7.02 (m, 1 H, Ar), 7.10 (s, 1 H, Ar), 7.17 (d, 2 H, J = 8.6 Hz, Ar), 7.32 (d, 2 H, J = 8.6 Hz, Ar), 7.64 (s, 1 H, Ar), 7.95 (s, 1 H, Ar). Anal. Calcd. for C16H14ClN3S: C, 60.85; H, 4.47; N, 13.31; S, 10.15; Cl, 11.22. Found: C, 60.97; H, 4.55; N, 13.19; S, 10.12; Cl, 11.07.
N2,N4-Dimethyl-N2,N4-bis(4-(methylthio)phenyl)quinazoline-2,4-diamine (18)
Compound 12c (0.200 g, 1.00 mmol) and N-methyl-4-(methylthio)aniline (16) (0.185 g, 1.21 mmol) were stirred at room temperature for 12 h. At the end of the reaction, solvent was evaporated, silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified using flash chromatography using chloroform/methanol which afforded dimer 18 as a yellow semisolid (0.195 g, 45%). TLC Rf = 0.87 (Hex:EtOAc; 1:1); 1H NMR, DMSO-d6 (400 MHz): 2.48 (s, 3 H, SCH3), 2.49 (s, 3 H, SCH3), 3.29 (s, 3 H, NCH3), 3.57 (s, 3 H, NCH3), 6.76-6.81 (m, 1 H, Ar), 6.88-6.90 (dd, 1 H, J = 0.89, J = 8.43, Ar), 7.14-7.16 (d, 2 H, J = 8.68, Ar) 7.25-7.29 (m, 4 H, Ar), 7.37-7.45 (m, 4 H, Ar).
6-((2-Chloroquinazolin-4-yl)amino)naphthalen-1-ol (20)
Using the general method described above, 12c (0.200 g, 1.00 mmol) and 6-aminonaphthalen-1-ol 19 (0.144 g, 0.904 mmol) were reacted for 12 h. At the end of the reaction, the solvent was evaporated, silica gel and methanol were added, and the methanol evaporated in vacuo to give a dried plug. The reaction mixture was purified using flash chromatography using chloroform/methanol to the afford 20 as white solid (0.070 g, 22%). TLC Rf = 0.67 (MeOH:CHCl3; 1:5); 1H NMR, DMSO-d6 (400 MHz): 6.83-6.87 (t, 1 H, Ar), 7.32-7.33 (d, 2 H, J = 4.55, Ar), 7.67-7.71 (dt, 1 H, J1 = 1.13, J2 = 6.96, J3 = 8.09, Ar), 7.74-7.76 (dd, 1 H, J1 = 0.79, J2 = 8.31, Ar), 7.81-7.84 (dd, 1 H, J1 = 2.11, J2 = 9.07, Ar), 7.90-7.94 (dt, 1 H, J1 = 1.13, J2 = 7.08, J3 = 8.35, Ar), 8.16-8.18 (d, 1 H, J = 9.02, Ar), 8.24-8.25 (d, 1 H, J = 2.03, Ar), 8.63-8.65 (dd, 1 H, J1 = 1.35, J2 = 8.66, Ar), 10.16 (s, 1 H, exch., NH), 10.40 (s, br, 1 H, exch., OH). The compound was used without further characterization.
2-Chloro-N-(5-methoxynaphthalen-2-yl)-N-methylquinazolin-4-amine (11)
Compound 20 (0.070 g, 0.22 mmol) was dissolved in anhydrous DMF (10 mL) in a three necked round-bottomed flask, and the reaction mixture was stirred at 0 °C under argon. Sodium hydride (0.016 g, 0.65 mmol) was added to the above mixture, which was stirred at 0 °C for an additional 15 min. Methyl iodide (0.068 g, 0.40 mmol) was added while maintaining the reaction at room temperature for 4 h. Methanol was added at the end of the reaction, solvent was evaporated, and the reaction mixture was then extracted with water and ethyl acetate (25 mL × 3) and dried over Na2SO4. Silica gel was added to the combined fractions, the solvent was removed by evaporation to yield a dry plug, and a column was run using hexanes/ethyl acetate. The fractions containing the product (TLC) were pooled and evaporated to obtain the desired compound 11 as an off-white solid (0.036 g, 47%). TLC Rf = 0.82 (MeOH:CHCl3; 1:5); mp, 171.9-173.8 °C; 1H NMR, DMSO-d6 (400 MHz): 3.64 (s, 3 H, NCH3), 3.99 (s, 3 H, OCH3), 6.86-6.88 (d, 1 H, J = 8.54, Ar), 7.00-7.06 (m, 2 H, Ar), 7.40-7.49 (m, 3 H, Ar), 7.63-7.71 (m, 2 H, Ar), 7.88-7.89 (d, 1 H, J = 2.02, Ar), 8.21-8.23 (d, 1 H, J = 8.92, Ar). Anal. Calcd. for C20H16ClN3O . 0.11 CH3(CH2)4CH3: C, 69.07; H, 4.93; N, 11.68; Cl, 9.86. Found C, 68.84; H, 5.06; N, 11.69; Cl, 9.64.
Molecular modeling and computational studies
Molecular modeling was performed for all analogs with the tubulin (PDB: 4O2B47), VEGFR-2 (PDB: 4AG846) and EGFR (PDB: 1M1757) crystal structures using the induced fit docking protocol of Maestro 11.9.45 The ligands were prepared using the Ligprep application of Maestro. The docking protocol was validated by re-docking the co-crystallized ligands colchicine, axitinib and erlotinib in tubulin, VEGFR-2, and EGFR, respectively, with RMSD of 0.17 Å, 0.50 Å, and 1.19 Å, respectively. The centroids around the ligands were defined as the ligand binding site. The OPLS3e force field was used, and amino acid residues within 5 Å from the docked poses were allowed to be optimized using prime refinement.
Biological studies
Effects of compounds on cellular microtubules.
A-10 cells were used to evaluate the effects of the compounds on cellular microtubules using indirect immunofluorescence techniques. Cells were treated overnight with the compounds, fixed in cold MeOH, and microtubules were visualized with a β-tubulin antibody (Sigma–Aldrich, St. Louis, MO). EC50 values were calculated as previously described58 and represent an average of at least three independent experiments.
Sulforhodamine B (SRB) assay.
The SRB assay was used, as previously described, to evaluate the antiproliferative and cytotoxic effects of the compounds against cancer cells.59 MDA-MB-435, SK-OV-3 and HeLa cells were purchased from the American Type Culture Collection (Manassas, VA). Details about the generation of the SK-OV-3-MDR1-6/6 and HeLa WT-βIII cells were previously described.59 The IC50 values represent an average of at least 3 independent experiments using triplicate points in each experiment.
Antibodies.
The PY-HRP antibody was obtained from BD Transduction Laboratories (Franklin Lakes, NJ). Antibodies against EGFR, VEGFR-2, and PDGFR-β were purchased from Cell Signaling Technology (Danvers, MA).
Phosphotyrosine ELISA.
A high-throughput phosphotyrosine ELISA was developed for evaluating the effect of compounds on RTKs.60 Cells used for these experiments have been shown to overexpress particular RTKs; specifically A431 for EGFR, U251 for VEGFR-2, and SH-SY5Y for PDGFR-β. Briefly, cells at 60–75% confluence are placed in serum-free medium for 18 h to reduce background phosphorylation. Cells were always >98% viable by trypan blue exclusion. Cells were then pretreated for 60 min with 100 nM-1.4 μM concentrations of the compound of interest followed by the addition of 100 ng/mL of purified growth factor (EGF, VEGF, or PDGFR-β) for 10 min. The reaction was stopped, and cells were permeabilized by quickly removing the media and adding ice-cold Tris-buffered saline (TBS) containing 0.05% Triton X-100, protease inhibitor cocktail, and tyrosine phosphatase inhibitor cocktail (Sigma Chemical). The TBS solution was then removed, and cells fixed to the plate for 30 min at 60 °C, with a further incubation in 70% ethanol for an additional 30 min. Cells were then exposed to a blocking solution (TBS with 1% BSA) for 1 h, washed, and then a horseradish peroxidase (HRP)-conjugated phosphotyrosine (PY) antibody was added overnight. The antibody was removed, cells were washed again in TBS, exposed to an enhanced luminol ELISA substrate (Pierce Chemical EMD, Rockford, IL), and light emission was measured using a plate reader (BioTek, Winooski VT). The known RTK-specific inhibitors (semaxanib, sunitinib, erlotinib) were used as positive controls for kinase inhibition. Data were graphed as a percent of cells receiving growth factor alone, and IC50 values were calculated from two to three separate biological replicates (n = 8 technical replicates per biological replicate) using sigmoidal dose-response relations in Prism 8.0 software (GraphPad).
Chorioallantoic membrane (CAM) assay.
The CAM assay is a standard assay for testing antiangiogenic agents. The CAM assay used in these studies was performed as described previously (Zhang, X.; Raghavan, S.; Ihnat, M.; Thorpe, J. E.; Disch, B. C.; Bastian, A.; Bailey-Downs, L. C.; Dybdal-Hargreaves, N. F.; Rohena, C. C.; Hamel, E.; Mooberry, S. L.; Gangjee, A. Bioorg Med Chem 2014, 22, 3753-72) Briefly, fertile leghorn chicken eggs (Ideal Poultry, Cameron, TX) were incubated for 10 days. The proangiogenic factors, human VEGF-165 and bFGF (100 ng each) were then added at saturation to a 6 mm microbial testing disk (BBL, Cockeysville, MD) and placed onto the CAM by breaking a small hole in the superior surface of the egg. Antiangiogenic compounds, at various doses, were added 8 h after the VEGF/bFGF at saturation to the same microbial testing disk, and the embryos were incubated for an additional 40 h. After 48 h, the CAMs were perfused with 2% paraformaldehyde containing 0.025% Triton X-100 for 20 sec, excised around the area of treatment, fixed again in 4% paraformaldehyde for 30 min, placed on Petri dishes, and a digitized image was taken using a dissecting microscope (Leica Z16-APO; Wetzlar, Germany) at 7.5X magnification. A grid was then added to the digital CAM images, and the average number of vessels within 5–7 grids counted as a measure of vascularity. Sunitinib and semaxanib were used as positive controls for antiangiogenic activity. Data were graphed as a percent of CAMs receiving bFGF/VEGF only and IC50 values calculated from two to three separate experiments with 2-5 replicates per experiment using non-linear regression dose-response relation analysis.
Quantitative tubulin studies.
Bovine brain tubulin was purified as described previously.61 Briefly, 1.0 mg/mL of tubulin (10 μM) was incubated for 15 min with 0.8 M monosodium glutamate (pH of 2 M stock solution adjusted to 6.6 with HCl), varying compound concentrations and 4% (v/v) dimethyl sulfoxide as solvent. After a 15 min preincubation, 0.4 mM GTP was added. The reaction mixtures were transferred to cuvettes held at 0 °C in a cuvette holder equipped with an electronic temperature controller in a recording spectrophotometer. After baselines were established, the temperature was jumped over about 30 s to 30 °C, and changes in turbidity were monitored for 20 min. The compound concentration that caused a 50% reduction in increase in turbidity, interpolated from the values obtained with defined compound concentrations, was defined as the IC50 value. The assay to measure inhibition of [3H]colchicine binding was described in detail previously.62 Briefly, 0.1 mg/mL (1.0 μM) tubulin was incubated, at 37 °C, with 5.0 μM [3H]colchicine and potential inhibitors at 1.0 or 5.0 μM, as indicated. Incubation was for 10 min, at which point 40-60% of the maximum colchicine that can be bound without inhibitor is reached. The [3H]colchicine was a product of Perkin-Elmer. CA-4 was a generous gift of Dr. G. R. Pettit, Arizona State University.
Scheme 1.

Synthesis of 4-chloro-2-methylquinazoline 12a and 4-chloroquinazoline 12b
Table 4.
Inhibition of [3H] colchicine binding and inhibition of tubulin assembly
| Compound | Inhibition of tubulin assembly | Inhibition of colchicine binding | |
|---|---|---|---|
| IC50 (μM ± SD) | 5 μM inhibitor | 1 μM inhibitor | |
| 3 | 0.48 ± 0.07 | 99 ± 0.3 | 91 ± 0.5 |
| 4 | 0.64 ± 0.01 | 98 ± 0.2 | 88 ± 0.2 |
| 8 | 0.47 ± 0.02 | 99 ± 0.8 | 92 ± 0.2 |
| CA-4 | 0.73 ± 0.04 | 98 ± 0.3 | 87 ± 0.8 |
Acknowledgements
This work was supported, in part, by a grant from the National Institutes of Health, National Cancer Institute (RO1 CA142868 (AG, SLM)); by the Duquesne University Adrian Van Kaam Chair in Scholarly Excellence (AG); and by an NSF equipment grant for NMR instrumentation (NMR: CHE 0614785).
Disclaimer
This research was supported in part by the Developmental Therapeutics Program in the Division of Cancer Treatment and Diagnosis of the National Cancer Institute, which includes federal funds under Contract No. HHSN261200800001E. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.
Abbreviations:
- RTKs
Receptor tyrosine kinases
- VEGF
vascular endothelial growth factor
- VEGFR-2
vascular endothelial growth factor receptor-2
- PDGFR-β
platelet-derived growth factor receptor-β
- EGFR
epidermal growth factor receptor
- CA-1
combretastatin A-1
- CA-4
combretastatin A-4
- CAM
chorioallantoic membrane
- Pgp
P-glycoprotein
- MTAs
microtubule targeting agents
Footnotes
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References:
- 1.Sirisoma N; Kasibhatla S; Pervin A; Zhang H; Jiang S; Willardsen JA; Anderson MB; Baichwal V; Mather GG; Jessing K; Hussain R; Hoang K; Pleiman CM; Tseng B; Drewe J; Cai SX, Discovery of 2-chloro-N-(4-methoxyphenyl)-N-methylquinazolin-4-amine (EP128265, MPI-0441138) as a potent inducer of apoptosis with high in vivo activity. J Med Chem 2008, 51, 4771–4779. [DOI] [PubMed] [Google Scholar]
- 2.Sirisoma N; Pervin A; Zhang H; Jiang S; Willardsen JA; Anderson MB; Mather G; Pleiman CM; Kasibhatla S; Tseng B; Drewe J; Cai SX, Discovery of N-(4-methoxyphenyl)-N,2-dimethylquinazolin-4-amine, a potent apoptosis inducer and efficacious anticancer agent with high blood brain barrier penetration. J Med Chem 2009, 52, 2341–2351. [DOI] [PubMed] [Google Scholar]
- 3.Lugano R; Ramachandran M; Dimberg A, Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci 2019, 77, 1745–1770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brand TM; Iida M; Luthar N; Starr MM; Huppert EJ; Wheeler DL, Nuclear EGFR as a molecular target in cancer. Radiother Oncol 2013, 108 (3), 370–7. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 5.Ramjiawan RR; Griffioen AW; Duda DG, Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? Angiogenesis 2017, 20 (2), 185–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jitariu AA; Raica M; Cimpean AM; Suciu SC, The role of PDGF-B/PDGFR-BETA axis in the normal development and carcinogenesis of the breast. Crit Rev Oncol Hematol 2018, 131, 46–52. [DOI] [PubMed] [Google Scholar]
- 7.Ferrara N; Adamis AP, Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov 2016, 15, 385–403. [DOI] [PubMed] [Google Scholar]
- 8.Yamaoka T; Kusumoto S; Ando K; Ohba M; Ohmori T, Receptor tyrosine kinase-targeted cancer therapy. Int J Mol Sci 2018, 19 (11), 3491–3525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Moserle L; Jimenez-Valerio G; Casanovas O, Antiangiogenic therapies: going beyond their limits. Cancer Discov 2014, 4, 31–41. [DOI] [PubMed] [Google Scholar]
- 10.Rini BI; Escudier B; Hariharan S; Roberts WG; Tarazi J; Rosbrook B; Askerova Z; DeAnnuntis LL; Motzer RJ, Long-term safety with axitinib in previously treated patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 2015, 13, 540–547. [DOI] [PubMed] [Google Scholar]
- 11.Swiecicki PL; Zhao L; Belile E; Sacco AG; Chepeha DB; Dobrosotskaya I; Spector M; Shuman A; Malloy K; Moyer J; McKean E; McLean S; Wolf GT; Eisbruch A; Prince M; Bradford C; Carey T; Worden FP, A phase II study evaluating axitinib in patients with unresectable, recurrent or metastatic head and neck cancer. Invest New Drugs 2015, 33, 1248–1256. [DOI] [PubMed] [Google Scholar]
- 12.Beaumont JL; Salsman JM; Diaz J; Deen KC; McCann L; Powles T; Hackshaw MD; Motzer RJ; Cella D, Quality-adjusted time without symptoms or toxicity analysis of pazopanib versus sunitinib in patients with renal cell carcinoma. Cancer 2016, 122, 1108–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kreisl TN; McNeill KA; Sul J; Iwamoto FM; Shih J; Fine HA, A phase I/II trial of vandetanib for patients with recurrent malignant glioma. Neuro Oncol 2012, 14, 1519–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Buchbinder EI; Sosman JA; Lawrence DP; McDermott DF; Ramaiya NH; Van den Abbeele AD; Linette GP; Giobbie-Hurder A; Hodi FS, Phase 2 study of sunitinib in patients with metastatic mucosal or acral melanoma. Cancer 2015, 121, 4007–4015. [DOI] [PubMed] [Google Scholar]
- 15.Reichardt P; Kang YK; Rutkowski P; Schuette J; Rosen LS; Seddon B; Yalcin S; Gelderblom H; Williams CC Jr.; Fumagalli E; Biasco G; Hurwitz HI; Kaiser PE; Fly K; Matczak E; Chen L; Lechuga MJ; Demetri GD, Clinical outcomes of patients with advanced gastrointestinal stromal tumors: safety and efficacy in a worldwide treatment-use trial of sunitinib. Cancer 2015, 121, 1405–1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Janjigian YY; Vakiani E; Ku GY; Herrera JM; Tang LH; Bouvier N; Viale A; Socci ND; Capanu M; Berger M; Ilson DH, Phase II trial of sorafenib in patients with chemotherapy refractory metastatic esophageal and gastroesophageal (GE) junction cancer. PLoS One 2015, 10, e0134731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Thomson DJ; Silva P; Denton K; Bonington S; Mak SK; Swindell R; Homer J; Sykes AJ; Lee LW; Yap BK; Slevin NJ, Phase II trial of sorafenib in advanced salivary adenoid cystic carcinoma of the head and neck. Head Neck 2015, 37, 182–187. [DOI] [PubMed] [Google Scholar]
- 18.Goedegebuure RSA; de Klerk LK; Bass AJ; Derks S; Thijssen V, Combining radiotherapy with anti-angiogenic therapy and immunotherapy; A therapeutic triad for cancer? Front Immunol 2018, 9, 3107–3121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yi M; Jiao D; Qin S; Chu Q; Wu K; Li A, Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol Cancer 2019, 18, 60–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.El-Kenawi AE; El-Remessy AB, Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol 2013, 170, 712–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Steinmetz MO; Prota AE, Microtubule-targeting agents: Strategies to hijack the cytoskeleton. Trends Cell Biol 2018, 28, 776–792. [DOI] [PubMed] [Google Scholar]
- 22.Akhmanova A; Steinmetz MO, Control of microtubule organization and dynamics: two ends in the limelight. Nat Rev Mol Cell Biol 2015, 16, 711–726. [DOI] [PubMed] [Google Scholar]
- 23.Field JJ; Kanakkanthara A; Miller JH, Microtubule-targeting agents are clinically successful due to both mitotic and interphase impairment of microtubule function. Bioorg Med Chem 2014, 22, 5050–5059. [DOI] [PubMed] [Google Scholar]
- 24.Kaul R; Risinger AL; Mooberry SL, Microtubule-targeting drugs: More than antimitotics. J Nat Prod 2019, 82, 680–685. [DOI] [PubMed] [Google Scholar]
- 25.Singh N; Yeh PJ, Suppressive drug combinations and their potential to combat antibiotic resistance. J Antibiot (Tokyo) 2017, 70, 1033–1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.He B; Lu C; Zheng G; He X; Wang M; Chen G; Zhang G; Lu A, Combination therapeutics in complex diseases. J Cell Mol Med 2016, 20, 2231–2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Stylianopoulos T; Munn LL; Jain RK, Reengineering the tumor vasculature: Improving drug delivery and efficacy. Trends Cancer 2018, 4, 258–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Martin JD; Seano G; Jain RK, Normalizing function of tumor vessels: Progress, opportunities, and challenges. Annu Rev Physiol 2019, 81, 505–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhou J; Jiang X; He S; Jiang H; Feng F; Liu W; Qu W; Sun H, Rational design of multitarget-directed ligands: Strategies and emerging paradigms. J Med Chem 2019, 62, 8881–8914. [DOI] [PubMed] [Google Scholar]
- 30.Jain RK, Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J Clin Oncol 2013, 31, 2205–2218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Murray A; Little SJ; Stanley P; Maraveyas A; Cawkwell L, Sorafenib enhances the in vitro anti-endothelial effects of low dose (metronomic) chemotherapy. Oncol Rep 2010, 24, 1049–1058. [DOI] [PubMed] [Google Scholar]
- 32.Naganuma Y; Choijamts B; Shirota K; Nakajima K; Ogata S; Miyamoto S; Kawarabayashi T; Emoto M, Metronomic doxifluridine chemotherapy combined with the anti-angiogenic agent TNP-470 inhibits the growth of human uterine carcinosarcoma xenografts. Cancer Sci 2011, 102, 1545–1552. [DOI] [PubMed] [Google Scholar]
- 33.Zhou F; Hu J; Shao JH; Zou SB; Shen SL; Luo ZQ, Metronomic chemotherapy in combination with antiangiogenic treatment induces mosaic vascular reduction and tumor growth inhibition in hepatocellular carcinoma xenografts. J Cancer Res Clin Oncol 2012, 138, 1879–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhang X; Raghavan S; Ihnat M; Thorpe JE; Disch BC; Bastian A; Bailey-Downs LC; Dybdal-Hargreaves NF; Rohena CC; Hamel E; Mooberry SL; Gangjee A, The design and discovery of water soluble 4-substituted-2,6-dimethylfuro[2,3-d]pyrimidines as multitargeted receptor tyrosine kinase inhibitors and microtubule targeting antitumor agents. Bioorg Med Chem 2014, 22, 3753–3772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gangjee A; Pavana RK; Ihnat MA; Thorpe JE; Disch BC; Bastian A; Bailey-Downs LC; Hamel E; Bai R, Discovery of antitubulin agents with antiangiogenic activity as single entities with multitarget chemotherapy potential. ACS Med Chem Lett 2014, 5, 480–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bello E; Taraboletti G; Colella G; Zucchetti M; Forestieri D; Licandro SA; Berndt A; Richter P; D'Incalci M; Cavalletti E; Giavazzi R; Camboni G; Damia G, The tyrosine kinase inhibitor E-3810 combined with paclitaxel inhibits the growth of advanced-stage triple-negative breast cancer xenografts. Mol Cancer Ther 2013, 12, 131–140. [DOI] [PubMed] [Google Scholar]
- 37.Ulahannan SV; Brahmer JR, Antiangiogenic agents in combination with chemotherapy in patients with advanced non-small cell lung cancer. Cancer Invest 2011, 29, 325–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Heist RS; Wang X; Hodgson L; Otterson GA; Stinchcombe TE; Gandhi L; Villalona-Calero MA; Watson P; Vokes EE; Socinski MA; Alliance for Clinical Trials in, O., CALGB 30704 (Alliance): A randomized phase II study to assess the efficacy of pemetrexed or sunitinib or pemetrexed plus sunitinib in the second-line treatment of advanced non-small-cell lung cancer. J Thorac Oncol 2014, 9, 214–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Calinescu AA; Castro MG, Microtubule targeting agents in glioma. Transl Cancer Res 2016, 5 (Suppl 1), S54–S60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=290809 (Accessed 30 September 2020).
- 41.Rewcastle GW; Denny WA; Bridges AJ; Zhou H; Cody DR; McMichael A; Fry DW, Tyrosine kinase inhibitors. 5. Synthesis and structure-activity relationships for 4-[(phenylmethyl)amino]- and 4-(phenylamino)quinazolines as potent adenosine 5'-triphosphate binding site inhibitors of the tyrosine kinase domain of the epidermal growth factor receptor. J Med Chem 1995, 38 (1), 3482–3487. [DOI] [PubMed] [Google Scholar]
- 42.Bridges AJ; Zhou H; Cody DR; Rewcastle GW; McMichael A; Showalter HD; Fry DW; Kraker AJ; Denny WA, Tyrosine kinase inhibitors. 8. An unusually steep structure-activity relationship for analogues of 4-(3-bromoanilino)-6,7-dimethoxyquinazoline (PD 153035), a potent inhibitor of the epidermal growth factor receptor. J Med Chem 1996, 39, 267–276. [DOI] [PubMed] [Google Scholar]
- 43.Pavana RK; Choudhary S; Bastian A; Ihnat MA; Bai R; Hamel E; Gangjee A, Discovery and preclinical evaluation of 7-benzyl-N-(substituted)-pyrrolo[3,2-d]pyrimidin-4-amines as single agents with microtubule targeting effects along with triple-acting angiokinase inhibition as antitumor agents. Bioorg Med Chem 2017, 25, 545–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yosaatmadja Y; Silva S; Dickson JM; Patterson AV; Smaill JB; Flanagan JU; McKeage MJ; Squire CJ, Binding mode of the breakthrough inhibitor AZD9291 to epidermal growth factor receptor revealed. J Struct Biol 2015, 192, 539–544. [DOI] [PubMed] [Google Scholar]
- 45.Schrödinger Release 2019-2: Maestro, S., LLC, New York, NY, 2019. [Google Scholar]
- 46.McTigue M; Murray BW; Chen JH; Deng YL; Solowiej J; Kania RS, Molecular conformations, interactions, and properties associated with drug efficiency and clinical performance among VEGFR TK inhibitors. Proc Natl Acad Sci U S A 2012, 109, 18281–18289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Prota AE; Danel F; Bachmann F; Bargsten K; Buey RM; Pohlmann J; Reinelt S; Lane H; Steinmetz MO, The novel microtubule-destabilizing drug BAL27862 binds to the colchicine site of tubulin with distinct effects on microtubule organization. J Mol Biol 2014, 426, 1848–1860. [DOI] [PubMed] [Google Scholar]
- 48.Wang XF; Wang SB; Ohkoshi E; Wang LT; Hamel E; Qian K; Morris-Natschke SL; Lee KH; Xie L, N-aryl-6-methoxy-1,2,3,4-tetrahydroquinolines: a novel class of antitumor agents targeting the colchicine site on tubulin. Eur J Med Chem 2013, 67, 196–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wang S-B; Wang X-F; Qin B; Ohkoshi E; Hsieh K-Y; Hamel E; Cui M-T; Zhu D-Q; Goto M; Morris-Natschke SL; Lee K-H; Xie L, Optimization of N-aryl-6-methoxy-1,2,3,4-tetrahydroquinolines as tubulin polymerization inhibitors. Bioorg Med Chem 2015, 23, 5740–5747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Teichert A; Jantos K; Harms K; Studer A, One-pot homolytic aromatic substitutions/HWE olefinations under microwave conditions for the formation of a small oxindole library. Org Lett 2004, 6, 3477–3480. [DOI] [PubMed] [Google Scholar]
- 51.Holden JK; Kang S; Hollingsworth SA; Li H; Lim N; Chen S; Huang H; Xue F; Tang W; Silverman RB; Poulos TL, Structure-based design of bacterial nitric oxide synthase inhibitors. J Med Chem 2015, 58, 994–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kamata M; Yamashita T; Imaeda T; Tanaka T; Masada S; Kamaura M; Kasai S; Hara R; Sasaki S; Takekawa S; Asami A; Kaisho T; Suzuki N; Ashina S; Ogino H; Nakano Y; Nagisa Y; Kato K; Kato K; Ishihara Y, Melanin-concentrating hormone receptor 1 antagonists. Synthesis and structure-activity relationships of novel 3-(aminomethyl)quinolines. J Med Chem 2012, 55, 2353–2366. [DOI] [PubMed] [Google Scholar]
- 53.Yang CH; Horwitz SB, Taxol: The First Microtubule Stabilizing Agent. Int J Mol Sci 2017, 18 (8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Devambatla RKV; Li W; Zaware N; Choudhary S; Hamel E; Mooberry SL; Gangjee A, Design, synthesis, and structure-activity relationships of pyrimido[4,5-b]indole-4-amines as microtubule depolymerizing agents that are effective against multidrug resistant cells. Bioorg Med Chem Lett 2017, 27, 3423–3430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Connolly DJ; Lacey PM; McCarthy M; Saunders CP; Carroll AM; Goddard R; Guiry PJ, Preparation and resolution of a modular class of axially chiral quinazoline-containing ligands and their application in asymmetric rhodium-catalyzed olefin hydroboration. J Org Chem 2004, 69, 6572–6589. [DOI] [PubMed] [Google Scholar]
- 56.Sun Z; Wang H; Wen K; Li Y; Fan E, Solvent-free or low-solvent large-scale preparation of chloropyrimidine and analogues. J Org Chem 2011, 76, 4149–4153. [DOI] [PubMed] [Google Scholar]
- 57.Stamos J; Sliwkowski MX; Eigenbrot C, Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J Biol Chem 2002, 277, 46265–46272. [DOI] [PubMed] [Google Scholar]
- 58.Gangjee A; Zhao Y; Hamel E; Westbrook C; Mooberry SL, Synthesis and biological activities of (R)- and (S)-N-(4-methoxyphenyl)-N,2,6-trimethyl-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-a minium chloride as potent cytotoxic antitubulin agents. J Med Chem 2011, 54, 6151–6155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Risinger AL; Jackson EM; Polin LA; Helms GL; LeBoeuf DA; Joe PA; Hopper-Borge E; Luduena RF; Kruh GD; Mooberry SL, The taccalonolides: microtubule stabilizers that circumvent clinically relevant taxane resistance mechanisms. Cancer Res 2008, 68, 8881–8888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Stockwell BR; Haggarty SJ; Schreiber SL, High-throughput screening of small molecules in miniaturized mammalian cell-based assays involving post-translational modifications. Chem. Biol 1999, 6, 71–83. [DOI] [PubMed] [Google Scholar]
- 61.Hamel E; Lin CM, Separation of active tubulin and microtubule-associated proteins by ultracentrifugation and isolation of a component causing the formation of microtubule bundles. Biochemistry 1984, 23, 4173–4184. [DOI] [PubMed] [Google Scholar]
- 62.Verdier-Pinard P; Lai J-Y; Yoo H-D; Yu J; Marquez B; Nagle DG; Nambu M; White JD; Falck JR; Gerwick WH; Day BW; Hamel E, Structure-activity analysis of the interaction of curacin A, the potent colchicine site antimitotic agent, with tubulin and effects of analogs on the growth of MCF-7 breast cancer cells. Mol. Pharmacol. 1998, 53, 62–76. [DOI] [PubMed] [Google Scholar]








