Next Article in Journal
NMR Spectroscopy for Protein Higher Order Structure Similarity Assessment in Formulated Drug Products
Previous Article in Journal
Propionic Anhydride Modification of Cellulosic Kenaf Fibre Enhancement with Bionanocarbon in Nanobiocomposites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug Discovery Targeting Focal Adhesion Kinase (FAK) as a Promising Cancer Therapy

1
Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Institute of Drug Discovery & Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
2
School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
3
Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
4
State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(14), 4250; https://doi.org/10.3390/molecules26144250
Submission received: 6 June 2021 / Revised: 30 June 2021 / Accepted: 7 July 2021 / Published: 13 July 2021
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
FAK is a nonreceptor intracellular tyrosine kinase which plays an important biological function. Many studies have found that FAK is overexpressed in many human cancer cell lines, which promotes tumor cell growth by controlling cell adhesion, migration, proliferation, and survival. Therefore, targeting FAK is considered to be a promising cancer therapy with small molecules. Many FAK inhibitors have been reported as anticancer agents with various mechanisms. Currently, six FAK inhibitors, including GSK-2256098 (Phase I), VS-6063 (Phase II), CEP-37440 (Phase I), VS-6062 (Phase I), VS-4718 (Phase I), and BI-853520 (Phase I) are undergoing clinical trials in different phases. Up to now, there have been many novel FAK inhibitors with anticancer activity reported by different research groups. In addition, FAK degraders have been successfully developed through “proteolysis targeting chimera” (PROTAC) technology, opening up a new way for FAK-targeted therapy. In this paper, the structure and biological function of FAK are reviewed, and we summarize the design, chemical types, and activity of FAK inhibitors according to the development of FAK drugs, which provided the reference for the discovery of new anticancer agents.

1. Introduction

Receptor tyrosine kinases (RTK) and non-receptor tyrosine kinases play vital roles in extracellular signals and process of cancer cell migration, proliferation, and survival, which usually are overexpressed in many human cancer cells [1,2]. Therefore, targeting RTKs and non-receptor tyrosine kinases has been considered to be a promising therapy for the treatment of human cancers [3].
FAK is one of cytoplasmic non-receptor protein tyrosine kinase, which is a member of the focal adhesion complex family [4,5]. FAK is a 125 kDa protein that is recruited in the early stages of cell adhesion and phosphorylated to mediate the formation of local adhesion [6,7,8,9]. FAK gene exists on chromosome 8q24 and encodes 1052 amino acid residues and its protein structure consists of three parts: N-terminal region, kinase region, and C-terminal region [9].
FAK was first discovered in 1992, which is related to integrin signal transduction [4]. FAK is considered to be a key regulator of growth factor receptor and integrin-mediated signaling, which controls the basic processes of human normal or cancer cells through its kinase activity and scaffold function [10,11]. FAK overexpression is detected in various tumor types, including prostate cancer, breast cancer, lung cancer, ovarian cancer, and neck cancer, and FAK is also associated with the poor prognosis of cancer patients [12,13]. Besides, it is related to unregulated proteins in cancer, providing a perfect environment for FAK-specific treatment [14]. FAK regulates tumorigenic and metastatic potential through a highly coordinated signal network, thereby promoting the occurrence of malignant tumors. These signal networks coordinate a series of cellular processes, such as cell proliferation, survival, invasion, migration, angiogenesis, and the regulation of cancer stem cell activity [5,15,16]. Different FAK antisense oligonucleotides [17], dominant-negative FAK C-terminal domain, FAK-CD or FRNK or FAK-siRNA [18,19] can lead to decreased cell survival, growth inhibition, or apoptosis. In recent years, FAK has been considered as a new potential tumor-treatment target [20].
So far, some FAK inhibitors have been identified and proved to be effective inhibitors that inhibit the tumor growth and metastasis in several preclinical and clinical models [21,22,23,24]. TAE-226 is a typical FAK inhibitor (IC50 = 5.5 nM), which has strong antiproliferative and anticancer potency against several types of cancers in vitro and in vivo [21]. GSK2256098 is a highly potent, ATP-competitive, and reversible FAK inhibitor [25], which is currently underway in clinical trials as a monotherapy or in combination with other drugs for patients with brain tumors or other cancers [26]. VS-6063 is a highly effective second-generation FAK inhibitor. Phase II clinical trials of VS-6063 in patients with KRAS mutant non-small cell lung cancer have been completed [27]. VS-6062 has now completed the first phase of research. VS-6062 not only exhibits significant antiproliferation potency of cancer cells but also plays a new role in changing tumor microenvironment [28]. VS-4718 significantly affects cell viability by inhibiting FAK activity and locking Y397 phosphorylation [23,24]. Another novel FAK inhibitor, CEP-37440, is currently in clinical trials for the treatment of cancers [22] (Table 1). The proteolytic targeting chimera (PROTAC) that can induce FAK degradation by using FAK kinase inhibitors as a “warhead” also has been developed [29]. A large number of studies have provided convincing evidence that FAK inhibitors could be used as chemical-targeted drugs to induce growth inhibition and apoptosis of cancer cells [30,31,32]. This article briefly introduces the structure and biological functions of FAK. Aiming at the development of FAK drugs, it focuses on the design, chemical types, and activities of FAK drugs to provide references for the development of new antitumor drugs.

2. FAK Structure and Function

The FAK protein molecule can be roughly divided into N-terminal FERM region, middle kinase region, C-terminal focal adhesion target (FAT), and proline-rich regions (PRR1, PRR2, and PRR3). FAK has 6 tyrosine sites that can be phosphorylated: Y397, Y407, Y576, Y577, Y861, and Y925, all of them are the key parts in FAK’s signal transduction. Y397 and Y407 sites are located at the N-terminus, Y576 and Y577 are located in the activation loop of the kinase region, and Y861 and Y925 are located at the C-terminus. Y397 is an autophosphorylation site of FAK, which provides a high-affinity-binding site for Src family proteins and plays a vital role in downstream signaling pathways [16].
There are binding sites for a variety of proteins at the N-terminus, such as signal transduction proteins, cytoskeleton proteins, and integrin β subunits [33]. The N-terminal FERM domain is composed of three closely related subdomains F1, F2, and F3, forming a three-leaf clover-like structure [34] (Figure 1). Among of them, F1 and F2 subdomains can interact with p53 and hinder the apoptosis of tumor cells; the F2 subdomain can regulate kinase-independent activities and mediate cell survival; the F3 subdomain can interact with Mdm-2 to enhance ubiquitination of p53 [33]. The kinase domain of FAK also called the catalytic domain, has a highly conserved amino acid sequence that can phosphorylate corresponding amino acid residues in proteins such as PI3K, growth factor receptor-binding protein 2 (GRB2), Src, and Cas. The focal adhesion targeting region at the C-terminus could interact with focal adhesion proteins, including paxillin [35], Talin [36], and Vinculin [37]. FAT can also bind to the F2 leaf in the FERM domain, leading to the dimerization of FAK, which is of great significance to the activation and nuclear localization of FAK [38]. PRR1 is located between the FERM region and the kinase region and has a binding site to the SH3 domain, which can bind to proteins such as Src. PRR2 and PRR3 near the C-terminus can mediate the interaction between FAK and protein molecules containing SH3 domains.
FAK shows kinase-dependent enzyme function and a kinase-independent scaffold function, which play important roles in regulating the survival, proliferation, migration, and angiogenesis of cancer cells. FAK is overexpressed in many human cancers, including glioma, breast, colon, prostate, and other cancers [10,16,39]. Inhibition of FAK has been shown to inhibit tumor progression and metastasis.

3. The Discovery of FAK Inhibitors and Their Therapeutic Significance

Because of its important roles in human cancer cells, targeting FAK inhibitors is emerging as a promising target for cancer therapy with small molecules. Currently, six FAK inhibitors are undergoing clinical and many FAK inhibitors have been reported with strong inhibitory activity of FAK and significant antiproliferation potency against cancer cells (Table 1). In this section, we summarized the progress for the development of FAK inhibitors.

3.1. FAK Inhibitors in Clinical Trials

GSK2256098 is a highly effective and selective FAK inhibitor (IC50 = 0.4 nM). GSK2256098 significantly inhibited FAK-Y397 phosphorylation in A549, OVCAR8, and U87MG cells (IC50 = 12, 15, and 8.5 nM, respectively). GSK2256098 inhibited FAK Y397 phosphorylation and then suppressed FAK-related Akt/ERK activation, which led to the decrease of cell viability, anchorage-independent growth, and motility in a dose-dependent manner in pancreatic ductal adenocarcinoma cells [40]. The results of the phase I trial of GSK2256098 in sixty-two patients with advanced solid tumors suggested GSK2256098 had an acceptable safety profile and showed clinical activity in patients with mesothelioma. A phase Ib trial study indicated that GSK2256098 combined MEK inhibitor trametinib in the treatment of patients with advanced solid tumors could increase the exposure of trametinib and has an acceptable safety profile [26].
VS-6063 is another FAK and Pyk2 dual inhibitor with more effective activity against FAK and Pyk2 than that of VS-6062. VS-6063 inhibited FAK-Y397 phosphorylation in a time and dose-dependent manner in ovarian cancer cells. The combination of VS-6063 with paclitaxel could significantly exhibit the antiproliferation effects and increased apoptosis in ovarian cancer cells [24]. Notably, VS-6063 could overcome YB-1-mediated paclitaxel resistance by an AKT-dependent pathway. VS-6063 showed an acceptable safety profile in forty-six patients with advanced solid tumors according to the results of phase I trial [27]. The phase II trial, multicenter cohorts, open-label, single-arm study of VS-6063 treatment in heavily patients with KRAS mutant NSCLC indicated VS-6063 monotherapy demonstrated modest clinical activity [41].
VS-6062 is an ATP-competitive FAK and Pyk2 dual inhibitor, which displayed significant inhibitory potency of FAK and Pyk2 (IC50 = 1.5 and 14 nM, respectively) [42]. VS-6062 potently blocked FAK-Y397 phosphorylation in a dose-dependent manner in Ewing sarcoma cell lines which resulted in the downregulation of its downstream targets AKT/mTOR and CAS activity. VS-6062 could suppress the growth and colony formation, and induced apoptosis in Ewing sarcoma cell lines [43]. The combined treatment of VS-6062 with sunitinib could potently inhibit angiogenesis and tumor proliferation on liver solid tumor models [44]. In addition, VS-6062 has been reported to be an effective antimetastatic and antiangiogenic agent, which could potently suppress EOC and endothelial cell migration and endothelial cell tube formation in vitro [45]. The results of phase I trial of VS-6062 in ninety-nine patients with advanced solid tumors indicated that VS-6062 showed an acceptable safety profile and displayed time- and dose-dependent nonlinear PK [46].
VS-4718 is a highly effective and selective FAK inhibitor (IC50 = 1.5 nM) [47]. VS-4718 displayed potent inhibition activity with IC50 values ranging from 0.25 μM to 3.53 μM in vitro in a pediatric preclinical testing program (PPTP) and exhibited excellent tolerance in PPTP preclinical xenografts in vivo [48]. In addition, it has been reported that VS-4718 could antagonize the multidrug resistance in ABCB1- and ABCG2-overexpressing cancer cells through competitively interacting with ABCB1 and ABCG2 and affecting the activity of ABCB1 and ABCG2, which indicated that VS-4718 combines with other anticancer drugs may antagonize the multidrug resistance [49,50,51,52]. The phase 1 clinical trial of VS-4718 combined with paclitaxel and gemcitabine for the treatment of advanced pancreatic cancer has been completed [53].
CEP-37440 is a potent FAK and ALK dual inhibitor with strong and selective inhibition activity of FAK and ALK (IC50 = 2.0 and 3.1 nM, respectively). CEP-37440 effectively suppressed the proliferation of Sup-M2 and Karpas-299 cells (NPM-ALK+ ALCL cell lines) with IC50 values 84 and 131 nM, respectively. CEP-37440 displayed favorable in vitro ADME properties and acceptable oral bioavailability in CD-1 mice, Sprague−Dawley rats, and cynomolgus monkeys. In addition, CEP-37440 is effective in preclinical models of FC-IBC02, SUM190, and KPL4 with GC50 values of 91, 900, and 890 nM, respectively [22,54]. CEP37440 is currently undergoing a human clinical phase I trial.
CEP-37440 was discovered by the optimization of lead compound 1 which is an ALK inhibitor containing a diaminopyrimidine core motif [55,56,57] (Figure 2A). When morpholine was switched to N-methyl piperazine (compound 2), the stability of compound 2 in rat liver microsomes was significantly improved without eroding the potency or selectivity. Based on these promising results of compound 2, the strategy is to further increase the inhibitory potency of ALK, maintaining the activity of auxiliary FAK, and improve the metabolic stability of human liver microsomes. Changing the substituent of N-methyl on the piperazine to a hydroxyethyl substituent (compound 3) showed more potent inhibitory activity against enzymes and cells. Based on compound 3, changing the bicycloheptene system to 2-(N-methylmethanesulfonamide) benzene ring gave compound 4, which exhibited similar ALK enzyme and cells inhibitory potency. When the 2-N, N-dimethylsulfonamide substituent was replaced with N-methylamide substituent (CEP-37440), it displayed acceptable oral bioavailability (47%) and demonstrated acceptable in vitro liver microsome stability across species (rat and human t1/2 > 40 min) and also exhibited great inhibitory potency of ALK and FAK. The molecular docking results also explained that compound CEP-37440 exhibited great dual ALK/FAK potency (Figure 2B) [22,58].

3.2. Pyrimidine FAK Inhibitors

In 2006, Novartis researchers first reported an ATP-competitive FAK inhibitor named as TAE-226 (Figure 3A). TAE-226 is a highly effective and orally active FAK and Pyk2 dual-target inhibitor [59]. TAE-226 exhibited a wide spectrum of antiproliferation activity against various human cancer cells with a mean GI50 value of 0.76 μM [60]. TAE-226 potently inhibited FAK-Y397 autophosphorylation and blocked FAK-mediated downstream signaling pathways [61]. TAE-226 showed a strong antitumor effect through suppressing migration and metastasis, and induced increased apoptosis in a variety of cancer cells, such as glioma, breast, ovarian, and neuroblastoma cells [62]. The crystal structure of TAE-226 complexed with the kinase domain of FAK has been determined (Figure 3B), which clearly demonstrates the interaction of TAE-226 with FAK [63]. However, in consideration of its serious side effects on sugar metabolism, TAE-226 never entered clinical trial but is usually compared with other newly developed FAK inhibitors as a positive drug [64].
PF-573228 (Figure 4) is the first selective and potent ATP-competitive FAK inhibitor developed by Pfizer [65], which potently suppressed the FAK activity with an IC50 of 4 nM and the phosphorylation of FAK on Tyr397 with IC50 values of 30–100 nM in F-G, SKOV-3 and PC3 cells. PF-573228 inhibited the chemotactic and haptotactic migration, resulting in concomitant with the inhibition of focal adhesion turnover in REF52 cells. PF-573228 could not inhibit the cell growth or induce cell apoptosis in REF52 and PC3 cells [66]. Based on the structural modifications of PF-573228, another two analogs VS-6062 and VS-6063 developed by Pfizer have been used in clinical trials.
2-Anilino-4-(benzimidazol-2-yl) pyrimidine 9 is a multi-kinase inhibitor with inhibitory potency of FAK, PLK1, Aurora B, and VEGF-R2 with IC50 values of 3.4, 1.2, 6, and 7.2 μM, respectively (Figure 5). Compound 9 also exhibited potent inhibition growth of SW-620, MDA-MB-435, OVCAR-3, CAKI-1, and MCF-7 cells at micromolar concentrations (IC50 values = 0.62, 0.19, 0.93, 0.86 and 0.47 μM, respectively) [67].
Farand’s group reported novel macrocycle diphenylpyrimidine derivatives as FAK inhibitors based on the lead compound VS-6062 in 2016 (Figure 6). The representative compound 10 showed strong antagonism against FAK and Pyk2 with IC50 values of 4.34 nM and 0.84 nM, respectively [68]. Among these compounds, compound 10 showed the best metabolic stability in the analysis of human microsomes.
Sulfonamide functional groups exist in many active compounds and exhibit antibacterial, anticancer, and antiviral activities [69,70,71]. Previous studies have shown that the introduction of functional sulfonamides into the C-2 aniline portion of the pyrimidine core could increase the inhibitory potency of Bruton’s tyrosine kinase (BTK), which is a successful and effective drug target for the treatment of B lymphocytic leukemia [72]. Therefore, Ma’s group synthesized novel diphenylpyrimidine derivatives (Figure 7) through the introduction of sulfonamide functional groups based on the famous FAK inhibitor TAE-226 to improve the anti-FAK activity and biological activity in 2017 [73].
These new compounds showed moderate anti-FAK enzyme activity compared to TAE-226 (IC50 = 5.5 nM). The most active compound 18 effectively inhibited FAK activity with an IC50 value of 86.7 nM. It is worth noting that compound 18 exhibited significantly inhibitory potency against pancreatic cancer cells ASPC-1 and PANC-1 with IC50 values of 3.92 and 0.53 μM, respectively, which were lower than that of TAE-226 (IC50 = 6.73 and >20 μM, respectively). Compound 18 also promoted the apoptosis of pancreatic cancer cells in a dose-dependent manner. Molecular docking results showed compound 18 bonded to the FAK enzyme tightly and the interaction with the FAK enzyme was similarly to TAE-226, which might explain compound 18 shows potent anti-FAK activity (Figure 8B). These results provide a theoretical basis for further optimizing the structure of novel sulfonamide compounds as FAK inhibitors.
In general, most of the potent FAK inhibitors have pyrimidine templates and N-methyl formamide substituted anilines, which are necessary to interact with amino acids Cys502, Asp564, and Leu567 of FAK protein to interfere with FAK activity [74,75,76]. Phosphorylated functional groups are very effective in improving the water solubility and biological activity of anticancer agents [77,78,79]. Ma’s group discovered that the introduction of phosphoryl groups on the C-2 aniline side chain of the pyrimidine core could improve its inhibitory activity of BTK and antiproliferation activity against B-cell leukemia cells [80,81,82,83]. In 2017, they designed and synthesized a series of novel diphenylpyrimidine analogues containing phosphamides as FAK inhibitors [84] (Figure 9).
Most of the phosphamide-containing diphenylpyrimidine analogs showed good activity against FAK, which were equivalent or showed stronger inhibition than that of TAE-226 (IC50 = 5.5 nM). Compound 24 exhibited stronger anti-FAK activity (IC50 = 4.65 nM), which was stronger than that of TAE-226. Importantly, compound 24 also exhibited a potent capacity to decrease the growth of AsPC-1 and BxPC-3 cells with IC50 values 1.66 and 0.57 μM, respectively, which was lower than that of TAE-226 (IC50 = 6.73 and 1.03 μM, respectively). Interestingly, compound 24 was not active against normal HPDE6-C7 cells (IC50 > 20 μM). In addition, compound 24 induced the apoptosis of AsPC-1 cells in dose and time-dependent manners and arrested AsPC-1 cells in a G2/M phase. The docking results indicated that compound 24 could well dock into the ATP-binding pocket of the FAK enzyme which is similar to lead compound TAE-226 (Figure 10). It is worth noting that compound 24 forms three strong hydrogen bonds, which are produced by the N-1 atom in the pyrimidine core with amino acid Cys502, the carbonyl group in N-methylbenzamide with the amino acid Asp564, and the nitrogen atom in N-methylbenzamide with Arg505 and Gly506 through a water molecule. Notably, the morpholine substituent and the ethoxyl group of compound 24 could form strong contacts with the amino acid Gly505 and Glu506 through the polar-induced forces.
Small molecule inhibitors targeting FAK are usually designed to bind residues that surround the ATP-binding pocket of FAK, including TAE-226, PF562271, and CEP-37440, etc. These inhibitors are composed of pyrimidine core and N-methylbenzamide functional group. The co-crystal structure of the complex of FAK and TAE226 shows that the hydrophilic functional group on its C-2-aniline side chain is beneficial to strengthen the contact with the amino acids Gln438 and Gly505 in the FAK protein-binding pocket [63,85]. In fact, efforts in the structural modification of pyrimidine scaffolds have found several FAK inhibitors that are more effective than TAE226, such as compounds 18 and 24 (Figure 8 and Figure 10), which indicated that structural modifications such as the introduction of sulfonate or phosphonate groups at C-2-aniline side chain are beneficial to increase the binding affinity with FAK. Therefore, Ma’s group introduced the N-morpholine carboxamide functional group on the aniline side chain at the C-2 position of the pyrimidine core to design novel carboxamide-diphenylpyrimidines with strong hydrogen bond affinity to FAK in 2019 (Figure 11) [86].
Most carboxamide-diphenylpyrimidines displayed high inhibitory potency of FAK activity with IC50 values ranging from 2.58 to 75.8 nM. Among them, compound 25 exhibited equal anti-FAK activity (IC50 = 5.17 nM) to the lead compound TAE-226 (IC50 = 5.5 nM). Significantly, compound 25 also displayed potent antiproliferation potency against AsPC-1, BxPC-3, and MCF-7/ADR cells (IC50 = 0.105, 0.09 and 0.59 μM, respectively), which were much lower than that of the lead compound TAE-226 (IC50 = 6.73 μM, >10 and >10 μM, respectively). Compound 25 potently inhibited FAK phosphorylation in a dose-dependent manner and completely inhibited it at a concentration of 50 nM for 72 h in AsPC-1 cells, while the lead compound TAE-226 completely inhibited FAK phosphorylation at a concentration of 100 nM. In addition, compound 25 also exhibited great antitumor efficacy in an AsPC-1 cancer xenograft mouse model. The results of molecule docking clearly suggested that compound 25 could tightly and similarly bind to the active site of FAK as the compound TAE-226 (Figure 12). Notably, compound 25 formed a strong hydrogen bond between the oxygen atom of the amide group and amino acid Asp564, which is similar to that of TAE-226 but also formed a new strong hydrogen bond through the carbonyl group with amino acid Gln438.
Recently, the discovery of irreversible covalent inhibitors targeting kinases has attracted great interest [87,88], which shows unique advantages, for example, irreversible covalent inhibitors could prolong the target retention time and obtain long-lasting inhibition without continuous administration, as well as reduce the development of drug-resistant mutations and better avoid resistance [89]. The crystal structure of FAK protein with TAE-226 clearly indicates that Cys427 on the glycine-rich loop of FAK locates close to the ATP-binding site. Therefore, this cysteine residue could be used as the nucleophilic group for the design of covalent FAK inhibitors. Chen’s group successfully discovered novel FAK inhibitors by the introduction of an acrylamide moiety to the pyrimidine scaffold of TAE-226 in 2018 (Figure 13) [90]. These compounds with appropriate linkers could ensure that acrylamide group can be in proximity to Cys427 of FAK protein and the 2,4-diphenylamine pyrimidine as a core scaffold to maintain the binding affinity with the FAK protein.
These compounds exhibited superior inhibitory potency against FAK with IC50 values ranging from 0.6 to 4.7 nM, which were lower than that of the lead compound TAE-226. Compound 30 bearing a squarate group displayed better inhibitory activity than compounds 32 and 33. Compound 30 also exhibited excellent antiproliferation activity of squamous cell carcinoma (SCC) cells with a similar ED50 (IC50 = 1.73 μM) to that of VS-4718 (IC50 = 1.49 μM). Besides, compound 30 significantly blocked Tyr397 phosphorylation and the autophosphorylation of FAK at low concentrations in SCC cells, which was comparable to VS-4718. The cocrystal structure of the FAK kinase domain with compound 30 shows that 30 makes a covalent bond with Cys 427 of the FAK kinase domain by means of its carbamate linker (Figure 14).
Molecular hybridization strategy is a common method of designing new drugs based on the combination of different biologically active parts to produce novel hybrids with higher affinity and effectiveness [91]. Before 2019, there was no report on 2,4-diarylaminopyrimidine dithiocarbamate hybrid compounds. The combination of 2,4-diarylaminopyrimidine and dithiocarbamate into one compound exhibited the potential to enhance its anti-FAK activity and antiproliferative effects [92,93]. Yin’s group designed a class of 2,4-diarylaminopyrimidines as novel FAK inhibitors by incorporating the dithiocarbamate moiety in 2019 (Figure 15) [94]. It is found that the different structural units of R have a great influence on FAK and cell inhibitory activity. Among of these compounds, compound 34 exhibited the highest inhibitory potency of FAK (inhibition rate = 73% at 1 μM). Compound 34 was selected as a new starting point for further optimization and retained the N,N-dimethylaminoethyl group. However, the introduction of a strong electron-withdrawing group, such as trifluoromethyl (compound 35), could significantly increase the FAK-inhibitory activity. Compound 35 exhibited more potent inhibitory activity against FAK (IC50 = 0.07 nM) compared to TAE-226 and excellent antiproliferative effects against HCT-116, PC-3, U87-MG, and MCF-7 cell lines with IC50 values of 1, 30, 60, and 20 nM, respectively, which were significantly lower than that of the lead compound TAE-226 (IC50 = 290, 1680, 1660, and 560 nM, respectively). Moreover, compound 35 significantly induced apoptosis by arresting cell cycle in G2/M phase in both MCF-7 and HCT-116 cell lines.
In recent years, with the development of polypharmacology and a deep understanding of tumor pathogenesis, multi-targeting antitumor agents have been considered as an important strategy to overcome drug resistance and improve therapeutic effects. Based on a fragment-based drug design strategy, Chen’s group discovered a new class of diphenylpyrimidine derivatives as potent FAK and EGFRT790M dual inhibitors in 2020 (Figure 16) [95]. Epidermal growth factor receptor (EGFR) tyrosine kinase has been proved to be a valuable clinical target for anticancer therapy, especially for NSCLC [96,97]. However, the T790M mutation (threonine to methionine) within the ATP site of the EGFR frequently occurs in NSCLC patients (about 60%), and these patients develop resistance to drugs that target EGFR, which leads to the failure of EGFR-targeted therapy [98,99]. In addition, novel EGFRT790M inhibitors such as CO-1686 (Phase III) and AZD-9291 consist of the core scaffold of pyrimidine. Therefore, they designed and synthesized the first novel dual FAK and EGFRT790M inhibitors using a fragment-based drug design strategy based on the lead compound TAE-226 and Rociletinib. Fortunately, most of them exhibited great inhibitory potency of FAK and EGFR T790M.
Among these compounds, compound 41 not only exhibited high inhibitory activity of FAK kinase (IC50 = 1.03 nM) and EGFR T790M (IC50 = 3.89 nM) but also displayed excellent antiproliferative activity against AsPC-1, BxPC-3, Panc-1 three FAK-overexpressing pancreatic cancer cells (IC50 = 0.909, 0.761, 1.218 μM, respectively) and two drug-resistant cancer cell lines MCF-7/ADR and H1975 cells (IC50 = 0.909, 0.761, 1.218 μM, respectively), which were significantly lower than that of the lead compound TAE-226. In addition, compound 41 also showed the efficiency in the in vivo assessment conducted in a FAK-driven human AsPC-1 cell xenograft mouse model.
Chen’s group reported another new class of irreversible covalent FAK inhibitors based on compound 30 in 2020 (Figure 17) [100]. Among them, compound 45 exhibited more effective and selective inhibition potency of FAK with an IC50 value of 0.6 nM than that of TAE-226. Compound 45 potently suppressed the proliferative activity of three human glioblastoma cell lines U-87 MG, A172, and U251 with IC50 values of 7.2, 0.89, and 1.2 μM, respectively, which were lower than that of the lead compound TAE-226 (IC50 = 2.9, 8.3 and 6.3 μM, respectively). Compound 45 could significantly suppress the autophosphorylation of FAK in U-87 MG cells at low concentrations and block the phosphorylation of FAK-Tyr397 in a dose-dependent manner. In addition, compound 45 also significantly reduced the rate of cell migration and blocked U-87 cells in the G2/M phase.
Positron emission tomography (PET) is an advanced clinical imaging technology in the field of nuclear medicine. In recent years, many groups discovered novel small molecule PET-imaging agents through 18F labeling strategies based on anticancer agents with dual biological properties. Zhang’s group first reported F-18-labelled 2,4-diaminopyrimidines as FAK inhibitors and tumor-imaging agents (Figure 18) [101]. These compounds displayed superior inhibition potency against FAK with IC50 values ranging from 5.0 nM to 205.1 nM. Interestingly, compounds 52 and 53 which were labeled by 18F exhibited respective IC50 values of 5.0 nM and 21.6 nM. In addition, compound 52 exhibited promising biodistribution data in sarcoma S180-bearing mice, with tumor/blood, tumor/muscle, and tumor/bone ratios of 1.17, 2.99, and 2.19, respectively.
Zhang’s group reported novel 2,4-diaminopyrimidine derivatives FAK as tumor radiotracers in 2021 (Figure 19) [102]. Among them, compounds 54, 55, and 58 exhibited potently inhibitory activity against FAK with IC50 values of 3.0, 0.6, and 3.2 nM, respectively. Replacement of the chlorine atom in the pyrimidine ring with other groups assisted in penetrating the protein pocket and maintaining a low IC50 level. Furthermore, compound 58 with a FAK titer was radiologically labeled [18F]. Compound 58 presented high radiochemical purity (>98%), high in vitro biostability, and good tumor uptake values in S180-bearing mice, with a tumor/muscle ratio of 2.08 and a tumor/bone ratio of 2.48 at 30 min of injection.

3.3. FAK Inhibitors of Pyrimidine Heterocyclic Compounds

2,4-Diaminopyrimidine is the main scaffold of FAK inhibitors, which exhibited the inhibitory activity against FAK and cancer cells. Therefore, many groups adopted the scaffold hopping strategy, replacing the core pyrimidine scaffold with pyrimidine heterocyclics, such as 7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine scaffold and thieno[3,2-d]pyrimidine scaffold to design novel FAK inhibitors.
7H-pyrrolo[2,3-d]pyrimidine derivative 59 (Figure 20) is a novel FAK inhibitor reported by Cheng’s group in 2019 [103]. They focused on the replacement of the 2,4-diaminopyrimidine core with the pyrimidine heterocyclic scaffold. They assumed it is feasible to introduce additional interactions with Glu500 by simply adding a hydrogen donor in this region, which would lead to the hinge binding part containing three donor-acceptor interactions, while the pyrimidine contains two cases. Besides, they introduced various substituents of R1 with hydrogen bond acceptors to form hydrogen bond interactions with Asp564 of the DFG motif. The introduction of dimethyl phosphine oxide moiety at R1 was favorable to increase activity against FAK. In addition, they further explored SAR at R2 with the dimethyl phosphine oxide substituted phenyl group at R1 position. A further modification is to introduce a substituted benzene ring or a substituted pyrazole ring at R2 to improve the efficacy and adjust the properties, and finally, focus on the R3 position of the phenyl group in position R2 on the pyrimidine. Since the para-substitution carrier R3 points to the solvent region, the introduction of hydrophilic fragments at the R3 position can help improve activity. A class of fragment-containing derivatives with different hydrophilic properties were discovered, and most of these compounds showed strong inhibitory potency against FAK. They also designed and synthesized compounds 61, 62, 63, and 64 containing thieno[2,3-d]pyrimidine or thieno[3,2-d]pyrimidine scaffold based on representative compounds 59 and 60 as FAK inhibitors.
Among them, compounds 59 and 60 exhibited great inhibitory potency of FAK with IC50 values of 5.4 and 23.1 nM, respectively. Compared with compounds 59 and 60, the inhibitory activity of thieno-pyrimidine analogs 61, 62, 63, and 64 against FAK was reduced by more than five times. These results indicate that the hydrogen bond between 7H-pyrrolo[2,3-d]pyrimidine and Glu500 helps to obtain high efficiency. In addition, compound 59 displayed inhibition activity against A549 cells with an IC50 value of 3.2 µM, and also induced A549 cells apoptosis and suppressed its migration in a dose-dependent manner. Moreover, compound 59 exhibited good metabolic stability in mouse, rat, and human liver microsomes. The results of the docking study indicated compound 59 was anchored to the hinge region via the canonical donor-acceptor-donor hydrogen-bonding motif between the nitrogen molecules on the 7H-pyrrolo[2,3-d]pyrimidine-2,4-diamine moiety and the backbone of residues Glu500 and Cys502 (Figure 21). The hydrophobic interaction by hydrophobic side chains of the Ala 452 and Leu 553 helps increase further stabilization. The dimethyl phosphine oxide moiety interacts with Asp564 of the DFG motif through hydrogen bonding.
Thieno [3,2-d]pyrimidine derivative 67 is a potent FAK inhibitor reported by Cheng’s group in 2020 [104] (Figure 22). Based on a cyclization strategy, Cheng’s group used 2,7-disubstituted-thieno[3,2-d]pyrimidine scaffold to mimic the bioactive conformation of the well-known diaminopyrimidine scaffold of FAK inhibitors. First, they focused on the R1 moiety by fixing the R2 moiety as a phosphonate group. In R1 moiety, different fragments containing hydrogen bond acceptors were introduced to form hydrogen bond interactions with Asp564 of the DFG motif. Compound 65 contains a 2-methoxy group at the R1 position, which exhibited the strongest inhibitory potency on FAK (IC50 = 134 nM). Then, Cheng’s group performed SAR detection at R2. The introduction of the N-substituted formamide group (compound 66, IC50 = 38.8 nM) produced twice the effectiveness of compound 65. Based on compound 66, the substituents of the benzene ring were changed to further explore the SAR. The introduction of F at 3-position of benzene ring maintained high efficacy against FAK (IC50 = 28.8 nM). In addition, they also explored the influence of the changes on the solubilizing groups at the R6 position.
The optimized piperidine-4-yl-substituted compound 67 has a high inhibitory activity against FAK (IC50 = 28.2 nM). Compound 67 also exhibited excellent antiproliferation activity against U-87MG, A549, and MDA-MB-231 cells (IC50 = 0.16, 0.27, and 0.19 µM, respectively), which were significantly lower than that of the lead compound TAE-226 (IC50 = 1.67, 1.16, and 4.06 µM, respectively). In addition, compound 67 could significantly induce cell apoptosis, arrest cell cycle at the G0/G1 phase, and suppress migration in MDA-MB-231 cells. The results of the docking study elucidate the possible binding modes of compound 67 and provide a structural basis for the further design of FAK inhibitors (Figure 23).
Gray’s group reported a new class of FAK inhibitors with a tricyclic benzopyrimidodiazepinone core in 2021 [105]. Previous studies suggested that the tricyclic benzopyrimidodiazepinone scaffold is a privileged scaffold for the design of kinase inhibitors, for example, BJG-01-181 (compound 72) was identified as a FAK inhibitor (IC50 = 62.2 nM) in the biochemical kinase assay. Therefore, they explored the structure–activity relationships of this novel scaffold to improve the inhibitory potency of FAK (Figure 24). In addition, the N-methylated compounds usually have better pharmacokinetic (PK) properties than the secondary amides [106], so they designed N-methylated tricyclic benzopyrimidodiazepinone core to provide compounds including compounds 74, 75, and 76 with enhanced FAK inhibitory potency. Based on compound 73, they further explored SAR of the substituent group on benzopyrimidodiazepinone core including compounds 77 and 78.
Compound 73, an N-methyl matched-pair with compound 72, was synthesized and exhibited a three-fold improvement against FAK with an IC50 value of 20.2 nM. A variety of modifications provided inhibitors 74, 75, and 76 with IC50 values in the 30–60 nM range. N-ethylation at any position of the central ring retained nearly identical inhibitory potency of FAK as compound 73. In addition, compound 73 exhibited potent activity in 3D-culture breast and gastric cancer models, and favorable pharmacokinetic properties in mice. The results of the docking study suggested that compound 73 revealed two hydrogen-bonding interactions with FAK protein, which might explain the potent inhibitory of compound 73 (Figure 25).

3.4. FAK Inhibitors with a Triazine Scaffold

Triazine ring is often reported as an important scaffold for anticancer drugs [107,108]. For example, furazil and dioxadet, which contain different amino groups at positions 2, 4, or 6, have been reported as anticancer drugs. Because the chlorine atom of the triazine nucleus can easily be substituted successively with nucleophilic groups, resulting in a variety of substitutions, many groups develop new FAK inhibitors with a triazine scaffold.
Pyrrolo[2,1-f][1,2,4]triazine 82 is a potent inhibitor of dual FAK and JAK2 developed by Zificsak’s group in 2012 [109]. Pyrrolo[2,1-f][1,2,4]triazine usually was designed as a core scaffold of ALK inhibitors, and Zificsak’s group screened 79 showing potent inhibitory activity against JAK2 (IC50 = 0.2 nM) and good potency of FAK (IC50 = 17 nM). In addition, bioavailable ALK inhibitor 80 showed potent FAK activity but modest inhibition of JAK2. Therefore, Zificsak’s group designed a new class of pyrrolo[2,1-f][1,2,4]triazines as dual inhibitors of FAK and JAK2 based on the compounds 79 and 80 (Figure 26). Compound 81 showed strongest inhibitory effect on JAK (IC50 = 2.0 nM) and FAK (IC50 = 14 nM). Compound 81 was further optimized by introducing a chlorine atom into the pyrrolo [2,1-f][1,2,4]triazine ring to obtain compound 82. Progress has been made in FAK and JAK2 dual inhibitors, which showed potent inhibitory activity against both FAK and JAK2. In addition, compound 82 potently inhibited p-STAT3 and p-FAK phosphorylation in CWR22 tumor mice xenografts.
Diarylamino-1,3,5-triazine derivative 87 was identified as a novel FAK inhibitor reported by Chen’s group in 2013 [110] (Figure 27). Chen’s group first introduced a 3,4,5-trimethoxyanilino group and a methanesulfonamide phenyl group on the 1,3,5-triazine ring to obtain compound 85 with poor activity against FAK (IC50 = 41.9 µM). Next, they removed the chlorine atom of the triazine ring based on compound 85 to give compound 86, which showed approximately eight-fold inhibitory activity against FAK (IC50 = 5.1 µM). Substituting an amide moiety (compound 87) for the complex sulfonamide group (compound 86) could significantly increase the inhibitory activity 13 times against FAK (IC50 = 0.4 µM). Chen’s group also explored the influence of the changes of the 3,4,5-trimethoxyanilino group on FAK activity (compounds 88, 89, 90, and 91).
Among of these compounds, compound 88 exhibited high inhibitory activity against both FAK (IC50 = 0.31 µM) and HUVEC cells growth (IC50 = 1.5 µM). In addition, the crystal structure of compound 88 in the FAK protein was resolved (PDB ID: 4brx), which indicated compound 88 interacting with the FAK kinase domain is highly similar to that of the lead compound TAE-226 (Figure 28).
Chen’s group reported novel imidazo[1,2-a][1,3,5]triazines as FAK inhibitors in 2015 [111]. Based on the studies of triazine FAK inhibitor 87, they designed novel compounds by the fusing imidazole ring with the triazine to give an imidazo[1,2-a][1,3,5]triazine (Figure 29). Imidazo[1,2-a][1,3,5]triazine 92 exhibited stronger binding affinity to FAK (2-fold) than that of compound 87, showing specific contributions of imidazo[1,2-a][1,3,5]triazine. SAR was further explored and Chen’s group designed compounds 94 and 95 with a CH2NH2 group or CH2NHAlloc group in the para position of the simple phenyl ring, which exhibited more potent inhibitory activity of FAK (IC50 = 120 and 50 nM, respectively) than that of compound 87.
Compared to TAE-226, compound 95 significantly inhibited the autophosphorylation of FAK Y397 in HCT-116, MDA-MB-231, U-87MG, and PC-3 cells. In addition, compound 95 exhibited excellent antiproliferation activity against HCT-116, and PC-3 cells (IC50 = 0.52, and 0.29 μM, respectively). Compound 95 also potently inhibited the cell-matrix adhesion, migration, and invasion of U87-MG cells in a dose-dependent manner. The results of the molecular docking studies suggested that compound 95 is very similar as observed for compound 87 (Figure 30). However, imidazo[1,2-a] ring could make additional interactions, most notably with Met499, CH2NH group forms a hydrogen bond interaction with Ile-428, and the CO of the alloc group in compound 95 also could produce an additional interaction with Glu506, which might explain the increased binding affinity of compound 95 to FAK.
Although the interaction mode of TAE-226 and compound 88 is very similar, compound 88 exhibited lower in vitro potency of FAK about 45 times than that of TAE-226. Chen’s group proposed that one major reason from X-ray structure is the missing chlorine atom in 1,3,5-triazine compounds, which in TAE-226 makes van der Waals interactions with Met499 of FAK kinase, therefore, Chen’s group developed 1,2,4-triazine compounds as a novel scaffold for FAK inhibitors which can contain a chlorine atom at position 6 of 1,2,4-triazine core in 2017 (Figure 31) [112].
Among these compounds, compound 98 containing a 2-OCH3-4-morpholino group showed the best anti-FAK activity with an IC50 value of 230 nM and excellent antiproliferation activity against HCT-116 cells (IC50 = 0.19 µM). The results of the docking studies suggested that the van der Waals interactions between the chlorine atom and Met499 of FAK protein might not be important to the binding of compound 98 with FAK protein, suggesting that the weaker hydrogen bonds between the amino 1,2,4-triazine and the hinge region of FAK may play an important role in the inhibitory activity of compound 98 (Figure 32).

3.5. Other Types of FAK Inhibitors

Many groups also reported five-membered heterocyclic compounds containing nitrogen as FAK inhibitor (Figure 33), including 1,3,4-thiadiazole derivatives 102 and 103 [113,114], 1,3,4-oxadiazole derivatives 104 and 105 [115,116], 2-styryl-5-nitroimidazole derivatives 106 [117], 1,2,4-triazole derivative 107 [118]. Most of these compounds exhibited inhibitory activity and antiproliferative activity at the micromolar levels.
Zhu’s group reported a new class of novel 1,3,4-thiadiazoles as FAK inhibitors. The most active compound 102 inhibited the activity of FAK and HEPG2 cells with an EC50 value of 10.79 and 10.28 μM, respectively (Figure 33). Furthermore, Zhu’s group designed and synthesized 1,3,4-thiadiazol-2-amide derivatives with more potent inhibitory potency of FAK and cancer cells growth compared to 102 [114]. The most active compound 103 exhibited better inhibitory activity against FAK with an IC50 value of 5.32 μM and inhibited proliferation of MCF-7 and B16-F10 cells with IC50 values of 0.45 and 0.31 μM, respectively [113]. The results of docking studies suggested that compounds 102 and 103 could be well embedded in the ATP-binding pocket of FAK through a hydrogen bond, π–σ interaction, and π–cation interaction (Figure 34).
Zhu’s group designed a new class of 1,3,4-oxadiazole-2(3H)-thiones as FAK inhibitors by replacing the 1,3,4-thiadiazole core of compounds 102 and 103 with further modifications. The most active compound 104 exhibited high inhibitory potency of FAK at submicromolar level (IC50 = 0.78 μM) and good inhibition of HepG2 and SM1116 cell growth (IC50 = 5.78 and 47.15 μM, respectively) [115]. Altintop’s group also reported a new class of oxadiazole-based derivatives as FAK inhibitors. The most active compound 105 exhibited significantly the inhibitory activity of FAK (Phospho-Tyr397) with an IC50 value of 19.5 μM in C6 cells [116]. Compound 105 also showed potent anticancer activity against C6 cells (IC50 = 4.63 μM) and caused higher caspase-3 activation and morphological changes.
In 2014, Zhu’s group also designed novel 2-styryl-5-nitroimidazole derivatives as FAK inhibitors with antiproliferative activity [117]. Compound 106 exhibited potently inhibitory activity against FAK (IC50 = 0.45 μM), and A549 and HeLa cells growth with IC50 values of 0.45, 3.11 μM, respectively. In addition, compound 106 induced HeLa cells apoptosis in a dose-dependent manner. Compound 106 could decrease the phosphorylation of PI3K, Akt, JNK, and STAT3, resulting in apoptosis induction and cell cycle arrest. The results of molecular docking revealed that compound 106 could well fit within the ATP-binding site of FAK. In 2021, Hayallah’s group reported a new class of 1,2,4-triazole derivatives as FAK inhibitors with potential anticancer activity [118]. The most active compound 107 inhibited the activity of FAK with an IC50 value of 18.10 nM and inhibited HepG2 and Hep3B cells growth with IC50 values of 3.78 and 4.83 μM, respectively.
Miki’s group reported 1,5-dihydropyrazolo[4,3-c][1,2]benzothiazines as novel FAK inhibitors by high-throughput screening (HTS) (Figure 35). The HTS of the internal compound library of this research group gave more than ten hit compounds of different chemical classes, and further modification gives compounds 109 and 110. Compound 109 and 110 inhibited the FAK activity with IC50 values 0.64 and 4.2 μM, respectively [119].

4. The Discovery of FAK-Targeting PROTACs and Their Therapeutic Significance

The proteolytic targeting chimera (PROTAC) is a new chemical knockout technique for post-translational proteins. PROTAC is a heterogeneous bifunctional small molecule containing two recognition parts: one specifically binds to E3 ubiquitin ligase, and the other specifically binds to the target protein. The PROTAC molecule can drive the E3 ubiquitin ligase to the target protein, which leads to the ubiquitination of the target protein, which leads to proteasome-mediated degradation (Figure 36) [120,121].
Crew’s group reported potent FAK selective degraders in 2018 [122]. In this work, they designed FAK degraders by conjugating a FAK ligand derived from VS-6063 (defactinib) through slight modifications with a peptide-based VHL ligand or thalidomide through ether linkers (Figure 37). All of these FAK PROTACs exhibited good inhibitory potency against FAK and could significantly induce the degradation of FAK at low concentrations. Among the FAK-PROTACs, compound 111 exhibited potent activity, which showed an IC50 value of 6.5 nM, DC50 value of 3 nM, and Dmax over 99%. Compound 111 displayed better selectivity to FAK than VS-6063. In addition, compound 111 displayed significant effects in suppressing the FAK downstream signaling pathways, which could highly induce the degradation of FAK in a dose-dependent manner with only 34% total FAK remaining at 10 nM and 5% at 50 nM, which outperformed VS-6063 (defactinib). In addition, compound 111 significantly reduces p-paxillin levels by as much as 85–90% at 50 nM, p-Akt suppression of 93% at 1 μM, and is more effective than VS-6063. Compound 111 also significantly inhibited the migration and invasion of MDA-MB-23 cells.
Ettmayer’s group reported a new class of FAK-PROTACs represented by compounds 113 and 114 in 2019, by tethering a previously identified FAK inhibitor compound 112 through an ether linker with a peptide-based VHL ligand and pomalidomide, respectively [123] (Figure 38). Both of FAK-PROTACs (compound 113 and 114) showed comparable degradation potencies and efficacies with mean pDC50 values of 7.45 and 7.08 nM, respectively. The CRBN-based PROTAC 113 potently degraded FAK (DC50 = 27 nM) with a Dmax of 95% at the chosen experimental conditions, while the VHL-based PROTAC 114 only achieved a Dmax of 80% and was less potent (DC50 = 243 nM) in the A549 cells. However, although the potent depletion of FAK, both FAK-PROTACs (compound 113 and 114) did not exhibit antiproliferation activity against the tested cell lines in either short or long-term assays, which indicated that scaffolding function of FAK may not be required for the proliferation of the tested cell lines.
Rao’s group reported a new class of FAK-PROTACs in 2020, by tethering a previously identified FAK inhibitors VS-6062 or VS-6063 through different ether linker with a CRBN E3 ligand pomalidomide [124] (Figure 39). They noticed that shorter diethylene or triethylene glycol linkers exhibited higher degradation activity of FAK. Among them, compound 115 caused rapid and significant degradation of FAK with DC50 values at the scale of picomolar in TM3, PA1, MDA-MB-436, LNCaP, and Ramos cells (DC50 = 310, 80, 330, 370, and 40 pM, respectively). Compound 115 showed a significant inhibitory effect on the autophosphorylation of FAK (pFAKtyr397) below 1 nM, but VS-6062 showed an inhibitory effect on pFAKtyr397 only at 3 μM. However, the efficient knockdown of FAK by compound 115 did not more severely affect the proliferation of the tested cell lines.
Gray’s group reported a new class of FAK-PROTACs by tethering a previously identified FAK inhibitor VS-4718 through an ether linker with a CRBN E3 ligand pomalidomide in a 2018 patent. All of them could effectively produce degradation of FAK with a Dmax of 85–100% at 100 nM, and compound 120, 121, 122, and 123 only achieved it at 10 nM [125] (Figure 40).

5. Concluding Remarks and Prospects

FAK is an intracellular non-receptor tyrosine kinase that promotes tumor cell growth by controlling cell adhesion, migration, proliferation, and survival. Therefore, targeting FAK is considered to be a promising cancer therapy with small molecules.
At present, a variety of FAK inhibitors have been proven to exhibit good antitumor activity. A comprehensive analysis of the mode of action of various FAK inhibitors shows that most of the inhibitor cores are composed of five-membered or six-membered nitrogen-containing heterocycles, which can be selectively inserted into the ATP-binding pocket of FAK and interact with the kinase hinge. The nitrogen atoms on the heterocycle could form key hydrogen bonds with the amino acid residues of FAK, and the heterocycle itself can form hydrophobic interactions with the surrounding residues. At the same time, the interaction between the branch chain of the core and FAK can further enhance the activity of the inhibitor, and the modification of the branch chain can also improve the drug ability of the inhibitor. FAK inhibitors have special effects on the FAK protein through the active groups in the structure, inhibiting the phosphorylation of phosphorylation sites (such as Tyr 397, Tyr 576, and Tyr 577) in the FAK structure, and inhibiting the overexpression of FAK, thereby exerting antitumor effect. So far, there is no approved FAK inhibitor in the market. Therefore, there is an urgent need to develop new FAK inhibitors with higher antitumor activity to exert stronger antitumor effects. Combining more anticancer therapies to treat human cancer is a frequently used strategy. The joint research of FAK inhibitors and other anticancer drugs will also be in the direction of FAK inhibitor development. PROTAC technology has also been applied to the development of FAK drugs. Several potential FAK PROTACs have been reported, which opens a new window for FAK-targeted therapy.

Funding

This research received no external funding.

Acknowledgments

This work was supported by the National Natural Sciences Foundations of China (No. 81703541 & U2004123 for Sai-Yang Zhang and No. 81673322 for Yan-Bing Zhang) and China Postdoctoral Science Foundation (No. 2018M632812 for Sai-Yang Zhang). Henan Association of Science and Technology (No. 2020HYTP056 for Sai-Yang Zhang, China) and Science and Technology Department of Henan Province (No. 20202310144, for Sai-Yang Zhang, China). The open fund of state key laboratory of Pharmaceutical Biotechnology, Nan-jing University, China (Grant no. KF-GN-202104).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cui, J.J. A new challenging and promising era of tyrosine kinase inhibitors. ACS Med. Chem. Lett. 2014, 5, 272–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kong, X.; Pan, P.; Sun, H.; Xia, H.; Wang, X.; Li, Y.; Hou, T. Drug Discovery Targeting Anaplastic Lymphoma Kinase (ALK). J. Med. Chem. 2019, 62, 10927–10954. [Google Scholar] [CrossRef]
  3. Manning, G.; Whyte, D.B.; Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase complement of the human genome. Science 2002, 298, 1912–1934. [Google Scholar] [CrossRef] [Green Version]
  4. Schaller, M.D.; Borgman, C.A.; Cobb, B.S.; Vines, R.R.; Reynolds, A.B.; Parsons, J.T. pp125FAK a structurally distinctive protein-tyrosine kinase associated with focal adhesions. Proc. Natl. Acad. Sci. USA 1992, 89, 5192–5196. [Google Scholar] [CrossRef] [Green Version]
  5. Zhao, J.; Guan, J.-L. Signal transduction by focal adhesion kinase in cancer. Cancer Metastasis Rev. 2009, 28, 35–49. [Google Scholar] [CrossRef]
  6. Schaller, M.D. The focal adhesion kinase. J. Endocrinol. 1996, 150, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Mitra, S.K.; Hanson, D.A.; Schlaepfer, D.D. Focal adhesion kinase: In command and control of cell motility. Nat. Rev. Mol. Cell Biol. 2005, 6, 56–68. [Google Scholar] [CrossRef]
  8. Parsons, J.T. Focal adhesion kinase: The first ten years. J. Cell Sci. 2003, 116, 1409–1416. [Google Scholar] [CrossRef] [Green Version]
  9. Hildebrand, J.D.; Schaller, M.D.; Parsons, J.T. Identification of sequences required for the efficient localization of the Focal Adhesion Kinase, pp125(FAK), to cellular focal adhesions. J. Cell Biol. 1993, 123, 993–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Schaller, M.D. Cellular functions of FAK kinases: Insight into molecular mechanisms and novel functions. J. Cell Sci. 2010, 123, 1007–1013. [Google Scholar] [CrossRef] [Green Version]
  11. Frame, M.C.; Patel, H.; Serrels, B.; Lietha, D.; Eck, M.J. The FERM domain: Organizing the structure and function of FAK. Nat. Rev. Mol. Cell Biol. 2010, 11, 802–814. [Google Scholar] [CrossRef]
  12. Béraud, C.; Dormoy, V.; Danilin, S.; Lindner, V.; Béthry, A.; Hochane, M.; Coquard, C.; Barthelmebs, M.; Jacqmin, D.; Lang, H.; et al. Targeting FAK scaffold functions inhibits human renal cell carcinoma growth. Int. J. Cancer 2015, 137, 1549–1559. [Google Scholar] [CrossRef]
  13. Pylayeva, Y.; Gillen, K.M.; Gerald, W.; Beggs, H.E.; Reichardt, L.F.; Giancotti, F.G. Ras- and PI3K-dependent breast tumorigenesis in mice and humans requires focal adhesion kinase signaling. J. Clin. Investig. 2009, 119, 252–266. [Google Scholar] [CrossRef] [Green Version]
  14. Siesser, P.M.F.; Hanks, S.K. The signaling and biological implications of FAK overexpression in cancer. Clin. Cancer Res. 2006, 12, 3233–3237. [Google Scholar] [CrossRef] [Green Version]
  15. Lee, B.Y.; Timpson, P.; Horvath, L.G.; Daly, R.J. FAK signaling in human cancer as a target for therapeutics. Pharmacol. Ther. 2015, 146, 132–149. [Google Scholar] [CrossRef] [PubMed]
  16. Lechertier, T.; Hodivala-Dilke, K. Focal Adhesion Kinase and tumour angiogenesis. J. Pathol. 2012, 226, 404–412. [Google Scholar] [CrossRef]
  17. Smith, C.S.; Golubovskaya, V.M.; Peck, E.; Xu, L.-H.; Monia, B.P.; Yang, X.; Cance, W.G. Effect of focal adhesion kinase (FAK) downregulation with FAK antisense oligonucleotides and 5-fluorouracil on the viability of melanoma cell lines. Melanoma Res. 2005, 15, 357–362. [Google Scholar] [CrossRef] [PubMed]
  18. Halder, J.; Landen, C.N., Jr.; Lutgendorf, S.K.; Li, Y.; Jennings, N.B.; Fan, D.; Nelkin, G.M.; Schmandt, R.; Schaller, M.D.; Sood, A.K. Focal adhesion kinase silencing augments docetaxel-mediated apoptosis in ovarian cancer cells. Clin. Cancer Res. 2005, 11, 8829–8836. [Google Scholar] [CrossRef] [Green Version]
  19. Demirbas, A.; Sahin, D.; Demirbas, N.; Karaoglu, S.A. Synthesis of some new 1,3,4-thiadiazol-2-ylmethyl-1,2,4-triazole derivatives and investigation of their antimicrobial activities. Eur. J. Med. Chem. 2009, 44, 2896–2903. [Google Scholar] [CrossRef] [PubMed]
  20. Kuhn, M.W.; Armstrong, S.A. Designed to kill: Novel menin-MLL inhibitors target MLL-rearranged leukemia. Cancer Cell 2015, 27, 431–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Shi, Q.; Hjelmeland, A.B.; Keir, S.T.; Song, L.; Wickman, S.; Jackson, D.; Ohmori, O.; Bigner, D.D.; Friedman, H.S.; Rich, J.N. A novel low-molecular weight inhibitor of focal adhesion kinase, TAE226, inhibits glioma growth. Mol. Carcinog. 2007, 46, 488–496. [Google Scholar] [CrossRef]
  22. Ott, G.R.; Cheng, M.; Learn, K.S.; Wagner, J.; Gingrich, D.E.; Lisko, J.G.; Curry, M.; Mesaros, E.F.; Ghose, A.K.; Quail, M.R.; et al. Discovery of Clinical Candidate CEP-37440, a Selective Inhibitor of Focal Adhesion Kinase (FAK) and Anaplastic Lymphoma Kinase (ALK). J. Med. Chem. 2016, 59, 7478–7496. [Google Scholar] [CrossRef] [PubMed]
  23. Shimizu, T.; Fukuoka, K.; Takeda, M.; Iwasa, T.; Yoshida, T.; Horobin, J.; Keegan, M.; Vaickus, L.; Chavan, A.; Padval, M.; et al. A first-in-Asian phase 1 study to evaluate safety, pharmacokinetics and clinical activity of VS-6063, a focal adhesion kinase (FAK) inhibitor in Japanese patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2016, 77, 997–1003. [Google Scholar] [CrossRef] [Green Version]
  24. Kang, Y.; Hu, W.; Ivan, C.; Dalton, H.J.; Miyake, T.; Pecot, C.V.; Zand, B.; Liu, T.; Huang, J.; Jennings, N.B.; et al. Role of Focal Adhesion Kinase in Regulating YB–1–Mediated Paclitaxel Resistance in Ovarian Cancer. JNCI J. Natl. Cancer Inst. 2013, 105, 1485–1495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Brown, N.F.; Williams, M.; Arkenau, H.T.; Fleming, R.A.; Tolson, J.; Yan, L.; Zhang, J.; Singh, R.; Auger, K.R.; Lenox, L.; et al. A study of the focal adhesion kinase inhibitor GSK2256098 in patients with recurrent glioblastoma with evaluation of tumor penetration of [11C]GSK2256098. Neuro Oncol. 2018, 20, 1634–1642. [Google Scholar] [CrossRef] [Green Version]
  26. Mak, G.; Soria, J.C.; Blagden, S.P.; Plummer, R.; Fleming, R.A.; Nebot, N.; Zhang, J.; Mazumdar, J.; Rogan, D.; Gazzah, A.; et al. A phase Ib dose-finding, pharmacokinetic study of the focal adhesion kinase inhibitor GSK2256098 and trametinib in patients with advanced solid tumours. Br. J. Cancer 2019, 120, 975–981. [Google Scholar] [CrossRef]
  27. Jones, S.F.; Siu, L.L.; Bendell, J.C.; Cleary, J.M.; Razak, A.R.A.; Infante, J.R.; Pandya, S.S.; Bedard, P.L.; Pierce, K.J.; Houk, B.; et al. A phase I study of VS-6063, a second-generation focal adhesion kinase inhibitor, in patients with advanced solid tumors. Investig. New Drugs 2015, 33, 1100–1107. [Google Scholar] [CrossRef] [PubMed]
  28. Stokes, J.B.; Adair, S.J.; Slack-Davis, J.K.; Walters, D.M.; Tilghman, R.W.; Hershey, E.D.; Lowrey, B.; Thomas, K.S.; Bouton, A.H.; Hwang, R.F.; et al. Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol. Cancer Ther. 2011, 10, 2135–2145. [Google Scholar] [CrossRef] [Green Version]
  29. Lu, Y.; Sun, H. Progress in the Development of Small Molecular Inhibitors of Focal Adhesion Kinase (FAK). J. Med. Chem. 2020, 63, 14382–14403. [Google Scholar] [CrossRef]
  30. Fu, Q.; Socki, R.A.; Niles, P.B. Evaluating reaction pathways of hydrothermal abiotic organic synthesis at elevated temperatures and pressures using carbon isotopes. Geochim. Cosmochim. Acta 2015, 154, 1–17. [Google Scholar] [CrossRef]
  31. Rosado, P.; Lequerica-Fernández, P.; Peña, I.; Alonso-Durán, L.; de Vicente, J.C. In oral squamous cell carcinoma, high FAK expression is correlated with low P53 expression. Virchows Arch. 2012, 461, 163–168. [Google Scholar] [CrossRef] [PubMed]
  32. Zheng, Y.; Gierut, J.; Wang, Z.; Miao, J.; Asara, J.M.; Tyner, A.L. Protein tyrosine kinase 6 protects cells from anoikis by directly phosphorylating focal adhesion kinase and activating AKT. Oncogene 2013, 32, 4304–4312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lim, S.-T.; Chen, X.L.; Lim, Y.; Hanson, D.A.; Vo, T.-T.; Howerton, K.; Larocque, N.; Fisher, S.J.; Schlaepfer, D.D.; Ilic, D. Nuclear FAK promotes cell proliferation and survival through FERM-enhanced p53 degradation. Mol. Cell 2008, 29, 9–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hamadi, A.; Bouali, M.; Dontenwill, M.; Stoeckel, H.; Takeda, K.; Rondé, P. Regulation of focal adhesion dynamics and disassembly by phosphorylation of FAK at tyrosine 397. J. Cell Sci. 2005, 118, 4415–4425. [Google Scholar] [CrossRef] [Green Version]
  35. Scheswohl, D.M.; Harrell, J.R.; Rajfur, Z.; Gao, G.; Campbell, S.L.; Schaller, M.D. Multiple paxillin binding sites regulate FAK function. J. Mol. Signal. 2008, 3, 1. [Google Scholar] [CrossRef] [Green Version]
  36. Lawson, C.; Lim, S.-T.; Uryu, S.; Chen, X.L.; Calderwood, D.A.; Schlaepfer, D.D. FAK promotes recruitment of talin to nascent adhesions to control cell motility. J. Cell Biol. 2012, 196, 223–232. [Google Scholar] [CrossRef] [Green Version]
  37. Subauste, M.C.; Pertz, O.; Adamson, E.D.; Turner, C.E.; Junger, S.; Hahn, K.M. Vinculin modulation of paxillin–FAK interactions regulates ERK to control survival and motility. J. Cell Biol. 2004, 165, 371–381. [Google Scholar] [CrossRef]
  38. Cho, J.-H.; Muralidharan, V.; Vila-Perello, M.; Raleigh, D.P.; Muir, T.W.; Palmer, A.G. Tuning protein autoinhibition by domain destabilization. Nat. Struct. Mol. Biol. 2011, 18, 550–555. [Google Scholar] [CrossRef] [Green Version]
  39. Golubovskaya, V.M.; Kwen, F.A.; Cance, W.G. Focal adhesion kinase and cancer. Histol. Histopathol. 2009, 24, 503–510. [Google Scholar]
  40. Zhang, J.; He, D.-H.; Zajac-Kaye, M.; Hochwald, S.N. A small molecule FAK kinase inhibitor, GSK2256098, inhibits growth and survival of pancreatic ductal adenocarcinoma cells. Cell Cycle 2014, 13, 3143–3149. [Google Scholar] [CrossRef] [Green Version]
  41. Gerber, D.E.; Camidge, D.R.; Morgensztern, D.; Cetnar, J.; Kelly, R.J.; Ramalingam, S.S.; Spigel, D.R.; Jeong, W.; Scaglioni, P.P.; Zhang, S.; et al. Phase 2 study of the focal adhesion kinase inhibitor defactinib (VS-6063) in previously treated advanced KRAS mutant non-small cell lung cancer. Lung Cancer 2020, 139, 60–67. [Google Scholar] [CrossRef]
  42. Roberts, W.; Ung, E.; Whalen, P.; Cooper, B.; Hulford, C.; Autry, C.; Richter, D.; Emerson, E.; Lin, J.; Kath, J.; et al. Antitumor Activity and Pharmacology of a Selective Focal Adhesion Kinase Inhibitor, PF-562,271. Cancer Res. 2008, 68, 1935–1944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Crompton, B.D.; Carlton, A.L.; Thorner, A.R.; Christie, A.L.; Du, J.; Calicchio, M.L.; Rivera, M.N.; Fleming, M.D.; Kohl, N.E.; Kung, A.L.; et al. High-Throughput Tyrosine Kinase Activity Profiling Identifies FAK as a Candidate Therapeutic Target in Ewing Sarcoma. Cancer Res. 2013, 73, 2873–2883. [Google Scholar] [CrossRef] [Green Version]
  44. Bagi, C.M.; Christensen, J.; Cohen, D.P.; Roberts, W.G.; Wilkie, D.; Swanson, T.; Tuthill, T.; Andresen, C.J. Sunitinib and PF-562,271 (FAK/Pyk2 inhibitor) effectively block growth and recovery of human hepatocellular carcinoma in a rat xenograft model. Cancer Biol. Ther. 2009, 8, 856–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Stone, R.L.; Baggerly, K.A.; Armaiz-Pena, G.N.; Kang, Y.; Sanguino, A.M.; Thanapprapasr, D.; Dalton, H.J.; Bottsford-Miller, J.; Zand, B.; Akbani, R.; et al. Focal adhesion kinase. Cancer Biol. Ther. 2014, 15, 919–929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Infante, J.R.; Camidge, D.R.; Mileshkin, L.R.; Chen, E.X.; Hicks, R.J.; Rischin, D.; Fingert, H.; Pierce, K.J.; Xu, H.; Roberts, W.G.; et al. Safety, Pharmacokinetic, and Pharmacodynamic Phase I Dose-Escalation Trial of PF-00562271, an Inhibitor of Focal Adhesion Kinase, in Advanced Solid Tumors. J. Clin. Oncol. 2012, 30, 1527–1533. [Google Scholar] [CrossRef]
  47. Tanjoni, I.; Walsh, C.; Uryu, S.; Tomar, A.; Nam, J.-O.; Mielgo, A.; Lim, S.-T.; Liang, C.; Koenig, M.; Patel, N.; et al. PND-1186 FAK inhibitor selectively promotes tumor cell apoptosis in three-dimensional environments. Cancer Biol. Ther. 2010, 9, 764–777. [Google Scholar] [CrossRef] [Green Version]
  48. Kurmasheva, R.T.; Gorlick, R.; Kolb, E.A.; Keir, S.T.; Maris, J.M.; Lock, R.B.; Carol, H.; Kang, M.; Reynolds, C.P.; Wu, J.; et al. Initial testing of VS-4718, a novel inhibitor of focal adhesion kinase (FAK), against pediatric tumor models by the Pediatric Preclinical Testing Program. Pediatric Blood Cancer 2017, 64, e26304. [Google Scholar] [CrossRef] [Green Version]
  49. Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug resistance in cancer: Role of ATP–dependent transporters. Nat. Rev. Cancer 2002, 2, 48–58. [Google Scholar] [CrossRef] [Green Version]
  50. Eckford, P.D.W.; Sharom, F.J. ABC Efflux Pump-Based Resistance to Chemotherapy Drugs. Chem. Rev. 2009, 109, 2989–3011. [Google Scholar] [CrossRef]
  51. Stavrovskaya, A.A.; Stromskaya, T.P. Transport proteins of the ABC family and multidrug resistance of tumor cells. Biochemistry 2008, 73, 592–604. [Google Scholar] [CrossRef] [PubMed]
  52. Ji, N.; Yang, Y.; Cai, C.-Y.; Lei, Z.-N.; Wang, J.-Q.; Gupta, P.; Teng, Q.-X.; Chen, Z.-S.; Kong, D.; Yang, D.-H. VS-4718 Antagonizes Multidrug Resistance in ABCB1- and ABCG2-Overexpressing Cancer Cells by Inhibiting the Efflux Function of ABC Transporters. Front. Pharmacol. 2018, 9, 1236. [Google Scholar] [CrossRef] [PubMed]
  53. Schaller, M.D.; Frisch, S.M. PND-1186 FAK inhibitor selectively promotes tumor cell apoptosis in three-dimensional environments. Cancer Biol. Ther. 2010, 9, 791–793. [Google Scholar] [CrossRef] [Green Version]
  54. Salem, I.; Alsalahi, M.; Chervoneva, I.; Aburto, L.D.; Addya, S.; Ott, G.R.; Ruggeri, B.A.; Cristofanilli, M.; Fernandez, S.V. The effects of CEP-37440, an inhibitor of focal adhesion kinase, in vitro and in vivo on inflammatory breast cancer cells. Breast Cancer Res. 2016, 18, 37. [Google Scholar] [CrossRef] [Green Version]
  55. Song, Z.; Yang, Y.; Liu, Z.; Peng, X.; Guo, J.; Yang, X.; Wu, K.; Ai, J.; Ding, J.; Geng, M.; et al. Discovery of Novel 2,4-Diarylaminopyrimidine Analogues (DAAPalogues) Showing Potent Inhibitory Activities against Both Wild-type and Mutant ALK Kinases. J. Med. Chem. 2015, 58, 197–211. [Google Scholar] [CrossRef]
  56. Huang, W.-S.; Liu, S.; Zou, D.; Thomas, M.; Wang, Y.; Zhou, T.; Romero, J.; Kohlmann, A.; Li, F.; Qi, J.; et al. Discovery of Brigatinib (AP26113), a Phosphine Oxide-Containing, Potent, Orally Active Inhibitor of Anaplastic Lymphoma Kinase. J. Med. Chem. 2016, 59, 4948–4964. [Google Scholar] [CrossRef] [PubMed]
  57. Ott, G.R.; Tripathy, R.; Cheng, M.; McHugh, R.; Anzalone, A.V.; Underiner, T.L.; Curry, M.A.; Quail, M.R.; Lu, L.; Wan, W.; et al. Discovery of a potent inhibitor of anaplastic lymphoma kinase with in vivo antitumor activity. ACS Med. Chem. Lett. 2010, 1, 493–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Bossi, R.T.; Saccardo, M.B.; Ardini, E.; Menichincheri, M.; Rusconi, L.; Magnaghi, P.; Orsini, P.; Avanzi, N.; Borgia, A.L.; Nesi, M.; et al. Crystal Structures of Anaplastic Lymphoma Kinase in Complex with ATP Competitive Inhibitors. Biochemistry 2010, 49, 6813–6825. [Google Scholar] [CrossRef] [PubMed]
  59. Wang, Z.G.; Fukazawa, T.; Nishikawa, T.; Watanabe, N.; Sakurama, K.; Motoki, T.; Takaoka, M.; Hatakeyama, S.; Omori, O.; Ohara, T.; et al. TAE226, a dual inhibitor for FAK and IGF-IR, has inhibitory effects on mTOR signaling in esophageal cancer cells. Oncol. Rep. 2008, 20, 1473–1477. [Google Scholar]
  60. Fukami, S.; Tomioka, D.; Murakami, Y.; Honda, T.; Hatakeyama, S. Pharmacological profiling of a dual FAK/IGF-1R kinase inhibitor TAE226 in cellular and in vivo tumor models. BMC Res. Notes 2019, 12, 347. [Google Scholar] [CrossRef]
  61. Hao, H.F.; Naomoto, Y.; Bao, X.H.; Watanabe, N.; Sakurama, K.; Noma, K.; Tomono, Y.; Fukazawa, T.; Shirakawa, Y.; Yamatsuji, T.; et al. Progress in researches about focal adhesion kinase in gastrointestinal tract. World J. Gastroenterol. 2009, 15, 5916–5923. [Google Scholar] [CrossRef]
  62. Tai, Y.-L.; Chen, L.-C.; Shen, T.-L. Emerging Roles of Focal Adhesion Kinase in Cancer. BioMed Res. Int. 2015, 2015, 690690. [Google Scholar] [CrossRef] [PubMed]
  63. Lietha, D.; Eck, M.J. Crystal structures of the FAK kinase in complex with TAE226 and related bis-anilino pyrimidine inhibitors reveal a helical DFG conformation. PLoS ONE 2008, 3, e3800. [Google Scholar] [CrossRef] [Green Version]
  64. Alexander, S.; Walter, F. Clinical Importance and Potential Use of Small Molecule Inhibitors of Focal Adhesion Kinase. Anti Cancer Agents Med. Chem. 2011, 11, 593–599. [Google Scholar]
  65. Schultze, A.; Fiedler, W. Therapeutic potential and limitations of new FAK inhibitors in the treatment of cancer. Expert Opin. Investig. Drugs 2010, 19, 777–788. [Google Scholar] [CrossRef]
  66. Slack-Davis, J.K.; Martin, K.H.; Tilghman, R.W.; Iwanicki, M.; Ung, E.J.; Autry, C.; Luzzio, M.J.; Cooper, B.; Kath, J.C.; Roberts, W.G.; et al. Cellular Characterization of a Novel Focal Adhesion Kinase Inhibitor. J. Biol. Chem. 2007, 282, 14845–14852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Determann, R.; Dreher, J.; Baumann, K.; Preu, L.; Jones, P.G.; Totzke, F.; Schachtele, C.; Kubbutat, M.H.; Kunick, C. 2-Anilino-4-(benzimidazol-2-yl)pyrimidines-a multikinase inhibitor scaffold with antiproliferative activity toward cancer cell lines. Eur. J. Med. Chem. 2012, 53, 254–263. [Google Scholar] [CrossRef]
  68. Farand, J.; Mai, N.; Chandrasekhar, J.; Newby, Z.E.; van Veldhuizen, J.; Loyer-Drew, J.; Venkataramani, C.; Guerrero, J.; Kwok, A.; Li, N.; et al. Selectivity switch between FAK and Pyk2: Macrocyclization of FAK inhibitors improves Pyk2 potency. Bioorg. Med. Chem. Lett. 2016, 26, 5926–5930. [Google Scholar] [CrossRef] [PubMed]
  69. Winum, J.-Y.; Scozzafava, A.; Montero, J.-L.; Supuran, C.T. Therapeutic potential of sulfamides as enzyme inhibitors. Med. Res. Rev. 2006, 26, 767–792. [Google Scholar] [CrossRef]
  70. Scozzafava, A.; Supuran, C.T. Carbonic anhydrase and matrix metalloproteinase inhibitors: Sulfonylated amino acid hydroxamates with MMP inhibitory properties act as efficient inhibitors of CA isozymes I, II, and IV, and N-hydroxysulfonamides inhibit both these zinc enzymes. J. Med. Chem. 2000, 43, 3677–3687. [Google Scholar] [CrossRef]
  71. Fukuoka, K.; Usuda, J.; Iwamoto, Y.; Fukumoto, H.; Nakamura, T.; Yoneda, T.; Narita, N.; Saijo, N.; Nishio, K. Mechanisms of action of the novel sulfonamide anticancer agent E7070 on cell cycle progression in human non-small cell lung cancer cells. Investig. New Drugs 2001, 19, 219–227. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, H.; Qu, M.; Xu, L.; Han, X.; Wang, C.; Shu, X.; Yao, J.; Liu, K.; Peng, J.; Li, Y.; et al. Design and synthesis of sulfonamide-substituted diphenylpyrimidines (SFA-DPPYs) as potent Bruton’s tyrosine kinase (BTK) inhibitors with improved activity toward B-cell lymphoblastic leukemia. Eur. J. Med. Chem. 2017, 135, 60–69. [Google Scholar] [CrossRef]
  73. Qu, M.; Liu, Z.; Zhao, D.; Wang, C.; Zhang, J.; Tang, Z.; Liu, K.; Shu, X.; Yuan, H.; Ma, X. Design, synthesis and biological evaluation of sulfonamide-substituted diphenylpyrimidine derivatives (Sul-DPPYs) as potent focal adhesion kinase (FAK) inhibitors with antitumor activity. Bioorg. Med. Chem. 2017, 25, 3989–3996. [Google Scholar] [CrossRef] [PubMed]
  74. Shanthi, E.; Krishna, M.H.; Arunesh, G.M.; Reddy, K.V.; Kumar, J.S.; Viswanadhan, V.N. Focal adhesion kinase inhibitors in the treatment of metastatic cancer: A patent review. Expert Opin. Ther. Pat. 2014, 24, 1077–1100. [Google Scholar] [CrossRef]
  75. Cao, F.Y.; Zhou, X.P.; Su, J.; Yang, X.H.; Mu, F.H.; Shen, J.Y.; Sun, W. Chemical Structure Characteristics and Bioactivity of Small Molecule FAK Inhibitors. Anti Cancer Agents Med. Chem. 2016, 16, 934–941. [Google Scholar] [CrossRef] [PubMed]
  76. Sulzmaier, F.J.; Jean, C.; Schlaepfer, D.D. FAK in cancer: Mechanistic findings and clinical applications. Nat. Rev. Cancer 2014, 14, 598–610. [Google Scholar] [CrossRef] [Green Version]
  77. Lee, M.Y.; Kim, M.H.; Kim, J.; Kim, S.H.; Kim, B.T.; Jeong, I.H.; Chang, S.; Kim, S.H.; Chang, S.-Y. Synthesis and SAR of sulfonyl- and phosphoryl amidine compounds as anti-resorptive agents. Bioorg. Med. Chem. Lett. 2010, 20, 541–545. [Google Scholar] [CrossRef]
  78. Huttunen, K.M.; Rautio, J. Prodrugs—An efficient way to breach delivery and targeting barriers. Curr. Top. Med. Chem. 2011, 11, 2265–2287. [Google Scholar] [CrossRef]
  79. Kamiyama, A.; Nakajima, M.; Han, L.; Wada, K.; Mizutani, M.; Tabuchi, Y.; Kojima-Yuasa, A.; Matsui-Yuasa, I.; Suzuki, H.; Fukuyama, K.; et al. Phosphonate-based irreversible inhibitors of human γ-glutamyl transpeptidase (GGT). GGsTop is a non-toxic and highly selective inhibitor with critical electrostatic interaction with an active-site residue Lys562 for enhanced inhibitory activity. Bioorg. Med. Chem. 2016, 24, 5340–5352. [Google Scholar] [CrossRef]
  80. Ge, Y.; Yang, H.; Wang, C.; Meng, Q.; Li, L.; Sun, H.; Zhen, Y.; Liu, K.; Li, Y.; Ma, X. Design and synthesis of phosphoryl-substituted diphenylpyrimidines (Pho-DPPYs) as potent Bruton’s tyrosine kinase (BTK) inhibitors: Targeted treatment of B lymphoblastic leukemia cell lines. Bioorg. Med. Chem. 2017, 25, 765–772. [Google Scholar] [CrossRef] [PubMed]
  81. Zhao, D.; Huang, S.; Qu, M.; Wang, C.; Liu, Z.; Li, Z.; Peng, J.; Liu, K.; Li, Y.; Ma, X.; et al. Structural optimization of diphenylpyrimidine derivatives (DPPYs) as potent Bruton’s tyrosine kinase (BTK) inhibitors with improved activity toward B leukemia cell lines. Eur. J. Med. Chem. 2017, 126, 444–455. [Google Scholar] [CrossRef] [PubMed]
  82. Ge, Y.; Jin, Y.; Wang, C.; Zhang, J.; Tang, Z.; Peng, J.; Liu, K.; Li, Y.; Zhou, Y.; Ma, X. Discovery of Novel Bruton’s Tyrosine Kinase (BTK) Inhibitors Bearing a N,9-Diphenyl-9H-purin-2-amine Scaffold. ACS Med. Chem. Lett. 2016, 7, 1050–1055. [Google Scholar] [CrossRef] [Green Version]
  83. Liu, H.-M.; Lee, C.-W. Reply to Letter to the Editor “Concern on article “Predicting procedure successful rate and 1-year patency after endovascular recanalization for chronic carotid artery occlusion by CT angiography”. Int. J. Cardiol. 2017, 229, 60. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, H.; Wu, B.; Ge, Y.; Huang, J.; Song, S.; Wang, C.; Yao, J.; Liu, K.; Li, Y.; Li, Y.; et al. Phosphamide-containing diphenylpyrimidine analogues (PA-DPPYs) as potent focal adhesion kinase (FAK) inhibitors with enhanced activity against pancreatic cancer cell lines. Bioorg. Med. Chem. 2017, 25, 6313–6321. [Google Scholar] [CrossRef] [PubMed]
  85. Wu, F.; Xu, T.; He, G.; Ouyang, L.; Han, B.; Peng, C.; Song, X.; Xiang, M. Discovery of Novel Focal Adhesion Kinase Inhibitors Using a Hybrid Protocol of Virtual Screening Approach Based on Multicomplex-Based Pharmacophore and Molecular Docking. Int. J. Mol. Sci. 2012, 13, 15668–15678. [Google Scholar] [CrossRef]
  86. Wang, L.; Ai, M.; Yu, J.; Jin, L.; Wang, C.; Liu, Z.; Shu, X.; Tang, Z.; Liu, K.; Luo, H.; et al. Structure-based modification of carbonyl-diphenylpyrimidines (Car-DPPYs) as a novel focal adhesion kinase (FAK) inhibitor against various stubborn cancer cells. Eur. J. Med. Chem. 2019, 172, 154–162. [Google Scholar] [CrossRef] [PubMed]
  87. Sanderson, K. Irreversible kinase inhibitors gain traction. Nat. Rev. Drug Discov. 2013, 12, 649–651. [Google Scholar] [CrossRef]
  88. Singh, J.; Petter, R.C.; Baillie, T.A.; Whitty, A. The resurgence of covalent drugs. Nat. Rev. Drug Discov. 2011, 10, 307–317. [Google Scholar] [CrossRef]
  89. Barf, T.; Kaptein, A. Irreversible protein kinase inhibitors: Balancing the benefits and risks. J. Med. Chem. 2012, 55, 6243–6262. [Google Scholar] [CrossRef]
  90. Yen-Pon, E.; Li, B.; Acebrón-Garcia-de-Eulate, M.; Tomkiewicz-Raulet, C.; Dawson, J.; Lietha, D.; Frame, M.C.; Coumoul, X.; Garbay, C.; Etheve-Quelquejeu, M.; et al. Structure-Based Design, Synthesis, and Characterization of the First Irreversible Inhibitor of Focal Adhesion Kinase. ACS Chem. Biol. 2018, 13, 2067–2073. [Google Scholar] [CrossRef]
  91. Viegas-Junior, C.; Danuello, A.; Bolzani, V.d.; Barreiro, E.J.; Fraga, C.A.M. Molecular hybridization: A useful tool in the design of new drug prototypes. Curr. Med. Chem. 2007, 14, 1829–1852. [Google Scholar] [CrossRef]
  92. Fu, D.-J.; Zhang, L.; Song, J.; Mao, R.-W.; Zhao, R.-H.; Liu, Y.-C.; Hou, Y.-H.; Li, J.-H.; Yang, J.-J.; Jin, C.-Y.; et al. Design and synthesis of formononetin-dithiocarbamate hybrids that inhibit growth and migration of PC-3 cells via MAPK/Wnt signaling pathways. Eur. J. Med. Chem. 2017, 127, 87–99. [Google Scholar] [CrossRef] [Green Version]
  93. Fu, D.-J.; Zhang, S.-Y.; Liu, Y.-C.; Zhang, L.; Liu, J.-J.; Song, J.; Zhao, R.-H.; Li, F.; Sun, H.-H.; Liu, H.-M.; et al. Design, synthesis and antiproliferative activity studies of novel dithiocarbamate–chalcone derivates. Bioorg. Med. Chem. Lett. 2016, 26, 3918–3922. [Google Scholar] [CrossRef]
  94. Su, Y.; Li, R.; Ning, X.; Lin, Z.; Zhao, X.; Zhou, J.; Liu, J.; Jin, Y.; Yin, Y. Discovery of 2,4-diarylaminopyrimidine derivatives bearing dithiocarbamate moiety as novel FAK inhibitors with antitumor and anti-angiogenesis activities. Eur. J. Med. Chem. 2019, 177, 32–46. [Google Scholar] [CrossRef]
  95. Ai, M.; Wang, C.; Tang, Z.; Liu, K.; Sun, X.; Ma, T.; Li, Y.; Ma, X.; Li, L.; Chen, L. Design and synthesis of diphenylpyrimidine derivatives (DPPYs) as potential dual EGFR T790M and FAK inhibitors against a diverse range of cancer cell lines. Bioorg. Chem. 2020, 94, 103408. [Google Scholar] [CrossRef] [PubMed]
  96. Fang, S.; Wang, Z. EGFR mutations as a prognostic and predictive marker in non-small-cell lung cancer. Drug Des. Dev. Ther. 2014, 8, 1595–1611. [Google Scholar]
  97. Jotte, R.M.; Spigel, D.R. Advances in molecular-based personalized non-small-cell lung cancer therapy: Targeting epidermal growth factor receptor and mechanisms of resistance. Cancer Med. 2015, 4, 1621–1632. [Google Scholar] [CrossRef] [PubMed]
  98. Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Jänne, P.A.; Kocher, O.; Meyerson, M.; Johnson, B.E.; Eck, M.J.; Tenen, D.G.; Halmos, B. EGFR Mutation and Resistance of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
  99. Kwak, E.L.; Sordella, R.; Bell, D.W.; Godin-Heymann, N.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Driscoll, D.R.; Fidias, P.; Lynch, T.J.; et al. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc. Natl. Acad. Sci. USA 2005, 102, 7665–7670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Li, B.; Li, Y.; Tomkiewicz-Raulet, C.; Dao, P.; Lietha, D.; Yen-Pon, E.; Du, Z.; Coumoul, X.; Garbay, C.; Etheve-Quelquejeu, M.; et al. Design, Synthesis, and Biological Evaluation of Covalent Inhibitors of Focal Adhesion Kinase (FAK) against Human Malignant Glioblastoma. J. Med. Chem. 2020, 63, 12707–12724. [Google Scholar] [CrossRef]
  101. Qi, Y.; Li, Y.; Fang, Y.; Qiang, B.; Gao, H.; Wang, S.; Zhang, H. Design, synthesis, and biological evaluation of F-18-labelled 2, 4-diaminopyrimidine-type FAK-targeted inhibitors as potential tumour imaging agents. Bioorg. Med. Chem. Lett. 2020, 30, 127452. [Google Scholar] [CrossRef]
  102. Qi, Y.; Li, Y.; Fang, Y.; Gao, H.; Qiang, B.; Wang, S.; Zhang, H. Design, Synthesis, Biological Evaluation, and Molecular Docking of 2,4-Diaminopyrimidine Derivatives Targeting Focal Adhesion Kinase as Tumor Radiotracers. Mol. Pharm. 2021, 18, 1634–1642. [Google Scholar] [CrossRef]
  103. Wang, R.; Chen, Y.; Zhao, X.; Yu, S.; Yang, B.; Wu, T.; Guo, J.; Hao, C.; Zhao, D.; Cheng, M. Design, synthesis and biological evaluation of novel 7H-pyrrolo[2,3-d]pyrimidine derivatives as potential FAK inhibitors and anticancer agents. Eur. J. Med. Chem. 2019, 183, 111716. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, R.; Yu, S.; Zhao, X.; Chen, Y.; Yang, B.; Wu, T.; Hao, C.; Zhao, D.; Cheng, M. Design, synthesis, biological evaluation and molecular docking study of novel thieno[3,2-d]pyrimidine derivatives as potent FAK inhibitors. Eur. J. Med. Chem. 2020, 188, 112024. [Google Scholar] [CrossRef] [PubMed]
  105. Groendyke, B.J.; Nabet, B.; Mohardt, M.L.; Zhang, H.; Peng, K.; Koide, E.; Coffey, C.R.; Che, J.; Scott, D.A.; Bass, A.J.; et al. Discovery of a Pyrimidothiazolodiazepinone as a Potent and Selective Focal Adhesion Kinase (FAK) Inhibitor. ACS Med. Chem. Lett. 2021, 12, 30–38. [Google Scholar] [CrossRef]
  106. Groendyke, B.J.; Powell, C.E.; Feru, F.; Gero, T.W.; Li, Z.; Szabo, H.; Pang, K.; Feutrill, J.; Chen, B.; Li, B.; et al. Benzopyrimidodiazepinone inhibitors of TNK2. Bioorg. Med. Chem. Lett. 2020, 30, 126948. [Google Scholar] [CrossRef]
  107. Song, J.; Cui, X.-X.; Wu, B.-W.; Li, D.; Wang, S.-H.; Shi, L.; Zhu, T.; Zhang, Y.-B.; Zhang, S.-Y. Discovery of 1,2,4-triazine-based derivatives as novel neddylation inhibitors and anticancer activity studies against gastric cancer MGC-803 cells. Bioorg. Med. Chem. Lett. 2020, 30, 126791. [Google Scholar] [CrossRef] [PubMed]
  108. Fu, D.-J.; Song, J.; Hou, Y.-H.; Zhao, R.-H.; Li, J.-H.; Mao, R.-W.; Yang, J.-J.; Li, P.; Zi, X.-L.; Li, Z.-H.; et al. Discovery of 5,6-diaryl-1,2,4-triazines hybrids as potential apoptosis inducers. Eur. J. Med. Chem. 2017, 138, 1076–1088. [Google Scholar] [CrossRef]
  109. Zificsak, C.A.; Gingrich, D.E.; Breslin, H.J.; Dunn, D.D.; Milkiewicz, K.L.; Theroff, J.P.; Thieu, T.V.; Underiner, T.L.; Weinberg, L.R.; Aimone, L.D.; et al. Optimization of a novel kinase inhibitor scaffold for the dual inhibition of JAK2 and FAK kinases. Bioorg. Med. Chem. Lett. 2012, 22, 133–137. [Google Scholar] [CrossRef]
  110. Dao, P.; Jarray, R.; le Coq, J.; Lietha, D.; Loukaci, A.; Lepelletier, Y.; Hadj-Slimane, R.; Garbay, C.; Raynaud, F.; Chen, H. Synthesis of novel diarylamino-1,3,5-triazine derivatives as FAK inhibitors with anti-angiogenic activity. Bioorg. Med. Chem. Lett. 2013, 23, 4552–4556. [Google Scholar] [CrossRef]
  111. Dao, P.; Smith, N.; Tomkiewicz-Raulet, C.; Yen-Pon, E.; Camacho-Artacho, M.; Lietha, D.; Herbeuval, J.P.; Coumoul, X.; Garbay, C.; Chen, H. Design, synthesis, and evaluation of novel imidazo[1,2-a][1,3,5]triazines and their derivatives as focal adhesion kinase inhibitors with antitumor activity. J. Med. Chem. 2015, 58, 237–251. [Google Scholar] [CrossRef] [PubMed]
  112. Dao, P.; Lietha, D.; Etheve-Quelquejeu, M.; Garbay, C.; Chen, H. Synthesis of novel 1,2,4-triazine scaffold as FAK inhibitors with antitumor activity. Bioorg. Med. Chem. Lett. 2017, 27, 1727–1730. [Google Scholar] [CrossRef]
  113. Yang, X.-H.; Xiang, L.; Li, X.; Zhao, T.-T.; Zhang, H.; Zhou, W.-P.; Wang, X.-M.; Gong, H.-B.; Zhu, H.-L. Synthesis, biological evaluation, and molecular docking studies of 1,3,4-thiadiazol-2-amide derivatives as novel anticancer agents. Bioorg. Med. Chem. 2012, 20, 2789–2795. [Google Scholar] [CrossRef]
  114. Sun, J.; Yang, Y.-S.; Li, W.; Zhang, Y.-B.; Wang, X.-L.; Tang, J.-F.; Zhu, H.-L. Synthesis, biological evaluation and molecular docking studies of 1,3,4-thiadiazole derivatives containing 1,4-benzodioxan as potential antitumor agents. Bioorg. Med. Chem. Lett. 2011, 21, 6116–6121. [Google Scholar] [CrossRef] [PubMed]
  115. Sun, J.; Ren, S.-Z.; Lu, X.-Y.; Li, J.-J.; Shen, F.-Q.; Xu, C.; Zhu, H.-L. Discovery of a series of 1,3,4-oxadiazole-2(3H)-thione derivatives containing piperazine skeleton as potential FAK inhibitors. Bioorg. Med. Chem. 2017, 25, 2593–2600. [Google Scholar] [CrossRef] [PubMed]
  116. Altıntop, M.D.; Sever, B.; Çiftçi, G.A.; Turan-Zitouni, G.; Kaplancıklı, Z.A.; Özdemir, A. Design, synthesis, in vitro and in silico evaluation of a new series of oxadiazole-based anticancer agents as potential Akt and FAK inhibitors. Eur. J. Med. Chem. 2018, 155, 905–924. [Google Scholar] [CrossRef]
  117. Duan, Y.-T.; Yao, Y.-F.; Huang, W.; Makawana, J.A.; Teraiya, S.B.; Thumar, N.j.; Tang, D.-J.; Tao, X.-X.; Wang, Z.-C.; Jiang, A.-Q.; et al. Synthesis, biological evaluation, and molecular docking studies of novel 2-styryl-5-nitroimidazole derivatives containing 1,4-benzodioxan moiety as FAK inhibitors with anticancer activity. Bioorg. Med. Chem. 2014, 22, 2947–2954. [Google Scholar] [CrossRef]
  118. Mustafa, M.; Abuo-Rahma, G.E.-D.A.; El-Hafeez, A.A.A.; Ahmed, E.R.; Abdelhamid, D.; Ghosh, P.; Hayallah, A.M. Discovery of antiproliferative and anti-FAK inhibitory activity of 1,2,4-triazole derivatives containing acetamido carboxylic acid skeleton. Bioorg. Med. Chem. Lett. 2021, 40, 127965. [Google Scholar] [CrossRef]
  119. Tomita, N.; Hayashi, Y.; Suzuki, S.; Oomori, Y.; Aramaki, Y.; Matsushita, Y.; Iwatani, M.; Iwata, H.; Okabe, A.; Awazu, Y.; et al. Structure-based discovery of cellular-active allosteric inhibitors of FAK. Bioorg. Med. Chem. Lett. 2013, 23, 1779–1785. [Google Scholar] [CrossRef] [PubMed]
  120. Sun, Y.; Zhao, X.; Ding, N.; Gao, H.; Wu, Y.; Yang, Y.; Zhao, M.; Hwang, J.; Song, Y.; Liu, W.; et al. PROTAC-induced BTK degradation as a novel therapy for mutated BTK C481S induced ibrutinib-resistant B-cell malignancies. Cell Res. 2018, 28, 779–781. [Google Scholar] [CrossRef]
  121. Raina, K.; Crews, C.M. Chemical Inducers of Targeted Protein Degradation*. J. Biol. Chem. 2010, 285, 11057–11060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Kargbo, R.B. Bifunctional Pyrimidines as Modulators of Focal Adhesion Kinase. ACS Med. Chem. Lett. 2020, 11, 409–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Popow, J.; Arnhof, H.; Bader, G.; Berger, H.; Ciulli, A.; Covini, D.; Dank, C.; Gmaschitz, T.; Greb, P.; Karolyi-Özguer, J.; et al. Highly Selective PTK2 Proteolysis Targeting Chimeras to Probe Focal Adhesion Kinase Scaffolding Functions. J. Med. Chem. 2019, 62, 2508–2520. [Google Scholar] [CrossRef] [PubMed]
  124. Gao, H.; Wu, Y.; Sun, Y.; Yang, Y.; Zhou, G.; Rao, Y. Design, Synthesis, and Evaluation of Highly Potent FAK-Targeting PROTACs. ACS Med. Chem. Lett. 2020, 11, 1855–1862. [Google Scholar] [CrossRef] [PubMed]
  125. Kargbo, R.B. Chemically Induced Degradation of FAK-ALK for Application in Cancer Therapeutics. ACS Med. Chem. Lett. 2020, 11, 1367–1368. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic diagram of the molecular structure of FAK protein.
Figure 1. Schematic diagram of the molecular structure of FAK protein.
Molecules 26 04250 g001
Figure 2. (A) The discovery of compound CEP-37440. (B) Glide docking of CEP37440 (gray) and compound 1 (yellow): left panel, ALK (PDB code 3LZT); right panel, FAK (PDB code 3BZ3).
Figure 2. (A) The discovery of compound CEP-37440. (B) Glide docking of CEP37440 (gray) and compound 1 (yellow): left panel, ALK (PDB code 3LZT); right panel, FAK (PDB code 3BZ3).
Molecules 26 04250 g002
Figure 3. (A) Chemical structure of TAE-226. (B) Crystal structure of TAE-226 complexed with the kinase domain of FAK (PDB ID: 2JKK). Hydrogen bonds are shown in yellow dash lines. The residues that can form hydrogen bonds with TAE-226 are shown in yellow.
Figure 3. (A) Chemical structure of TAE-226. (B) Crystal structure of TAE-226 complexed with the kinase domain of FAK (PDB ID: 2JKK). Hydrogen bonds are shown in yellow dash lines. The residues that can form hydrogen bonds with TAE-226 are shown in yellow.
Molecules 26 04250 g003
Figure 4. Chemical structure of PF-573228.
Figure 4. Chemical structure of PF-573228.
Molecules 26 04250 g004
Figure 5. Chemical structure and inhibition activity of compound 9.
Figure 5. Chemical structure and inhibition activity of compound 9.
Molecules 26 04250 g005
Figure 6. Macrocyclic double-target inhibitors of FAK and Pyk2 were obtained by cyclization.
Figure 6. Macrocyclic double-target inhibitors of FAK and Pyk2 were obtained by cyclization.
Molecules 26 04250 g006
Figure 7. Discovery of diphenylpyrimidine derivatives with sulfonamide functional groups as FAK inhibitors.
Figure 7. Discovery of diphenylpyrimidine derivatives with sulfonamide functional groups as FAK inhibitors.
Molecules 26 04250 g007
Figure 8. (A) Chemical structure of compound 18. (B) Predicted binding pose of compound 18 in FAK enzyme (PDB code: 2JKK).
Figure 8. (A) Chemical structure of compound 18. (B) Predicted binding pose of compound 18 in FAK enzyme (PDB code: 2JKK).
Molecules 26 04250 g008
Figure 9. The fragment-based drug design strategy of compound 24.
Figure 9. The fragment-based drug design strategy of compound 24.
Molecules 26 04250 g009
Figure 10. (A) Chemical structure of compound 24. (B) Predicted binding models of TAE-226 (red) and compound 24 (violet) in FAK enzyme (PDB code: 2JKK).
Figure 10. (A) Chemical structure of compound 24. (B) Predicted binding models of TAE-226 (red) and compound 24 (violet) in FAK enzyme (PDB code: 2JKK).
Molecules 26 04250 g010
Figure 11. Compound 25 was obtained by structural modification.
Figure 11. Compound 25 was obtained by structural modification.
Molecules 26 04250 g011
Figure 12. (A) Chemical structure of compound 25. (B) Predicted binding models of TAE-226 (red) and compound 25 (blue) in FAK enzyme (PDB code: 2JKK).
Figure 12. (A) Chemical structure of compound 25. (B) Predicted binding models of TAE-226 (red) and compound 25 (blue) in FAK enzyme (PDB code: 2JKK).
Molecules 26 04250 g012
Figure 13. The design strategy of first irreversible covalent FAK inhibitors.
Figure 13. The design strategy of first irreversible covalent FAK inhibitors.
Molecules 26 04250 g013
Figure 14. (A) Chemical structure of compound 30. (B) Crystal structure of compound 30 (PDB ID: 6GCX) linked the kinase domain of FAK.
Figure 14. (A) Chemical structure of compound 30. (B) Crystal structure of compound 30 (PDB ID: 6GCX) linked the kinase domain of FAK.
Molecules 26 04250 g014
Figure 15. The design strategy of 2,4-diarylaminopyrimidine derivatives by incorporating the dithiocarbamate moiety.
Figure 15. The design strategy of 2,4-diarylaminopyrimidine derivatives by incorporating the dithiocarbamate moiety.
Molecules 26 04250 g015
Figure 16. The designed strategy of dual FAK and EGFRT790M inhibitors.
Figure 16. The designed strategy of dual FAK and EGFRT790M inhibitors.
Molecules 26 04250 g016
Figure 17. Discovery of reversible valence FAK inhibitors. Compound 30 and compound 45 came from the same research group.
Figure 17. Discovery of reversible valence FAK inhibitors. Compound 30 and compound 45 came from the same research group.
Molecules 26 04250 g017
Figure 18. Design and synthesis of F-18-labelled 2, 4-diaminopyrimidine-type FAK-targeted inhibitors as potential tumor-imaging agents.
Figure 18. Design and synthesis of F-18-labelled 2, 4-diaminopyrimidine-type FAK-targeted inhibitors as potential tumor-imaging agents.
Molecules 26 04250 g018
Figure 19. Design of 2,4-diaminopyrimidine derivatives targeting focal adhesion kinase as tumor radiotracers.
Figure 19. Design of 2,4-diaminopyrimidine derivatives targeting focal adhesion kinase as tumor radiotracers.
Molecules 26 04250 g019
Figure 20. Compounds 59 and 61 replaces 2,4-diaminopyrimidine scaffolds by pyrrolo-pyrimidine 2,4-diamine and thieno-pyrimidine 2,4-diamine scaffolds.
Figure 20. Compounds 59 and 61 replaces 2,4-diaminopyrimidine scaffolds by pyrrolo-pyrimidine 2,4-diamine and thieno-pyrimidine 2,4-diamine scaffolds.
Molecules 26 04250 g020
Figure 21. (A) Chemical structure of compound 59. (B) Predicted binding mode of compound 59 in the ATP-binding site of FAK (PDB ID: 2JKK).
Figure 21. (A) Chemical structure of compound 59. (B) Predicted binding mode of compound 59 in the ATP-binding site of FAK (PDB ID: 2JKK).
Molecules 26 04250 g021
Figure 22. Design and synthesis of thieno [3, 2-d]pyrimidine derivatives as potent FAK inhibitors.
Figure 22. Design and synthesis of thieno [3, 2-d]pyrimidine derivatives as potent FAK inhibitors.
Molecules 26 04250 g022
Figure 23. The predicted docked pose of compound 67 (yellow sticks) in the FAK active site (PDB: 2JKK). Detailed interactions with the protein residues. Each dashed red line represents hydrogen bonds.
Figure 23. The predicted docked pose of compound 67 (yellow sticks) in the FAK active site (PDB: 2JKK). Detailed interactions with the protein residues. Each dashed red line represents hydrogen bonds.
Molecules 26 04250 g023
Figure 24. Discovery of a pyrimidothiazolodiazepinone as a potent and selective focal adhesion kinase (FAK) inhibitor.
Figure 24. Discovery of a pyrimidothiazolodiazepinone as a potent and selective focal adhesion kinase (FAK) inhibitor.
Molecules 26 04250 g024
Figure 25. Docking of compound 73 with FAK (PDB 6I8Z). (A) Key interactions of compound 73 with active-site residues, notably the critical hydrogen-bond of the thiazole nitrogen with Asp564. (B) FAK surface map shows the U-shaped active site, with the 2-methyl group extending into a hydrophobic pocket.
Figure 25. Docking of compound 73 with FAK (PDB 6I8Z). (A) Key interactions of compound 73 with active-site residues, notably the critical hydrogen-bond of the thiazole nitrogen with Asp564. (B) FAK surface map shows the U-shaped active site, with the 2-methyl group extending into a hydrophobic pocket.
Molecules 26 04250 g025
Figure 26. Optimization of a novel kinase inhibitor scaffold for the dual inhibition of JAK2 and FAK kinases.
Figure 26. Optimization of a novel kinase inhibitor scaffold for the dual inhibition of JAK2 and FAK kinases.
Molecules 26 04250 g026
Figure 27. Designs and synthesis of diarylamino 1,3,5-triazine derivatives as FAK inhibitors.
Figure 27. Designs and synthesis of diarylamino 1,3,5-triazine derivatives as FAK inhibitors.
Molecules 26 04250 g027
Figure 28. (A). Chemical structure of compound 88. (B). Compound 88 is shown to bound to the active site of the FAK kinase (beige ribbon with activation looping cyan, PDB ID: 4brx). Key side chains and the inhibitors (yellow) are shown in stick representation.
Figure 28. (A). Chemical structure of compound 88. (B). Compound 88 is shown to bound to the active site of the FAK kinase (beige ribbon with activation looping cyan, PDB ID: 4brx). Key side chains and the inhibitors (yellow) are shown in stick representation.
Molecules 26 04250 g028
Figure 29. Designs and synthesis of imidazo [1,2-a][1,3,5]triazine cyclic FAK inhibitors.
Figure 29. Designs and synthesis of imidazo [1,2-a][1,3,5]triazine cyclic FAK inhibitors.
Molecules 26 04250 g029
Figure 30. Docking of compound 87 (A) and 95 (B) into the ATP-binding pocket of FAK (PDB ID: 4c7t).
Figure 30. Docking of compound 87 (A) and 95 (B) into the ATP-binding pocket of FAK (PDB ID: 4c7t).
Molecules 26 04250 g030
Figure 31. Design and synthesis of 1,2,4-triazine derivatives as FAK inhibitors.
Figure 31. Design and synthesis of 1,2,4-triazine derivatives as FAK inhibitors.
Molecules 26 04250 g031
Figure 32. (A) Chemical structure of compound 98. (B) compound 98 is shown bound to the active site of the FAK kinase (beige ribbon with activation looping cyan, PDB ID: 4brx). Key side chains and the inhibitors (yellow) are shown in stick representation.
Figure 32. (A) Chemical structure of compound 98. (B) compound 98 is shown bound to the active site of the FAK kinase (beige ribbon with activation looping cyan, PDB ID: 4brx). Key side chains and the inhibitors (yellow) are shown in stick representation.
Molecules 26 04250 g032
Figure 33. Nitrogen-containing 5-membered heterocyclic FAK inhibitors.
Figure 33. Nitrogen-containing 5-membered heterocyclic FAK inhibitors.
Molecules 26 04250 g033
Figure 34. Molecular docking modeling of compound 102 (A) and compound 103 (B) with FAK.
Figure 34. Molecular docking modeling of compound 102 (A) and compound 103 (B) with FAK.
Molecules 26 04250 g034
Figure 35. Design and synthesis of an allosteric inhibitor 110 targeting FAK.
Figure 35. Design and synthesis of an allosteric inhibitor 110 targeting FAK.
Molecules 26 04250 g035
Figure 36. (A) FAK protein schematic. FAK-PROTACs can act on both enzymatic and nonenzymatic functions of FAK, while FAK inhibitor only acts on the enzymatic function of FAK. (B) Schematic depiction of the PROTAC strategy.
Figure 36. (A) FAK protein schematic. FAK-PROTACs can act on both enzymatic and nonenzymatic functions of FAK, while FAK inhibitor only acts on the enzymatic function of FAK. (B) Schematic depiction of the PROTAC strategy.
Molecules 26 04250 g036
Figure 37. Bifunctional pyrimidine 111 as a proteolytic targeting chimera of focal adhesion kinase (FAK).
Figure 37. Bifunctional pyrimidine 111 as a proteolytic targeting chimera of focal adhesion kinase (FAK).
Molecules 26 04250 g037
Figure 38. Compounds 113 and 114 as a proteolytic targeting chimera of FAK.
Figure 38. Compounds 113 and 114 as a proteolytic targeting chimera of FAK.
Molecules 26 04250 g038
Figure 39. Compound 115 as a proteolytic targeting chimera of focal adhesion kinase.
Figure 39. Compound 115 as a proteolytic targeting chimera of focal adhesion kinase.
Molecules 26 04250 g039
Figure 40. The proteolytic targeting chimera of focal adhesion kinase.
Figure 40. The proteolytic targeting chimera of focal adhesion kinase.
Molecules 26 04250 g040
Table 1. All reported FAK inhibitors in clinical trials.
Table 1. All reported FAK inhibitors in clinical trials.
InhibitorStructureTarget (IC50)CompanyClinical TrialStatusIdentifier
GSK-2256098 Molecules 26 04250 i001FAK IC50 = 0.4 nMGlaxo Smith Klinephase Icompleted
completed
completed
NCT01138033
NCT00996671
NCT02551653
VS-6063
(PF-04554878)
(Defactinib)
Molecules 26 04250 i002FAK IC50 = 0.6 nM
Pyk2 IC50 = 0.6 nM
Verastemphase IIcompleted
completed
completed
NCT01943292
NCT00787033
NCT01951690
VS-6062
(PF-00562271)
Molecules 26 04250 i003FAK IC50 = 1.5 nM
Pyk2 IC50 = 14 nM
Verastemphase IcompletedNCT00666926
VS-4718
(PND-1186)
Molecules 26 04250 i004FAK IC50 = 1.5 nMVerastemphase Iterminated
withdrawn
NCT01849744
NCT02215629
CEP-37440 Molecules 26 04250 i005FAK IC50 = 2 nM
ALK IC50 = 3.1 nM
Teva Brandedphase IcompletedNCT01922752
BI-853520
(IN-10018)
UnknownFAKBoehringer Ingelheimphase Icompleted
completed
NCT01335269
NCT01905111
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pang, X.-J.; Liu, X.-J.; Liu, Y.; Liu, W.-B.; Li, Y.-R.; Yu, G.-X.; Tian, X.-Y.; Zhang, Y.-B.; Song, J.; Jin, C.-Y.; et al. Drug Discovery Targeting Focal Adhesion Kinase (FAK) as a Promising Cancer Therapy. Molecules 2021, 26, 4250. https://doi.org/10.3390/molecules26144250

AMA Style

Pang X-J, Liu X-J, Liu Y, Liu W-B, Li Y-R, Yu G-X, Tian X-Y, Zhang Y-B, Song J, Jin C-Y, et al. Drug Discovery Targeting Focal Adhesion Kinase (FAK) as a Promising Cancer Therapy. Molecules. 2021; 26(14):4250. https://doi.org/10.3390/molecules26144250

Chicago/Turabian Style

Pang, Xiao-Jing, Xiu-Juan Liu, Yuan Liu, Wen-Bo Liu, Yin-Ru Li, Guang-Xi Yu, Xin-Yi Tian, Yan-Bing Zhang, Jian Song, Cheng-Yun Jin, and et al. 2021. "Drug Discovery Targeting Focal Adhesion Kinase (FAK) as a Promising Cancer Therapy" Molecules 26, no. 14: 4250. https://doi.org/10.3390/molecules26144250

Article Metrics

Back to TopTop