Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Review

Focal adhesion kinase: from biological functions to therapeutic strategies

verfasst von: Ximin Tan, Yuheng Yan, Bin Song, Shuangli Zhu, Qi Mei, Kongming Wu

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

Abstract

Focal adhesion kinase (FAK), a nonreceptor cytoplasmic tyrosine kinase, is a vital participant in primary cellular functions, such as proliferation, survival, migration, and invasion. In addition, FAK regulates cancer stem cell activities and contributes to the formation of the tumor microenvironment (TME). Importantly, increased FAK expression and activity are strongly associated with unfavorable clinical outcomes and metastatic characteristics in numerous tumors. In vitro and in vivo studies have demonstrated that modulating FAK activity by application of FAK inhibitors alone or in combination treatment regimens could be effective for cancer therapy. Based on these findings, several agents targeting FAK have been exploited in diverse preclinical tumor models. This article briefly describes the structure and function of FAK, as well as research progress on FAK inhibitors in combination therapies. We also discuss the challenges and future directions regarding anti-FAK combination therapies.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ATE
Atezolizumab
CAFs
Cancer-associated fibroblast
CDK
Cyclin-dependent kinase
CIK
Cytokine-induced killer
CRC
Colorectal cancer
CSCs
Cancer stem cells
ECM
Extracellular matrix
EMT
Epithelial–mesenchymal transformation
EGFR
Epidermal growth factor receptor
FADD
Fas-associated death domain
FAK
Focal adhesion kinase
FAKi
Focal adhesion kinase inhibitor
FAT
Focal adhesion-targeting
FERM
Four-point-one-ezrin-radixin-moesin
HNSCC
Neck squamous cell carcinoma
iCCA
Intrahepatic cholangiocarcinoma
IGF-1
Insulin-like growth factor-1
IGF-1R
Insulin-like growth factor-1 receptor system
JNK
Jun NH2-terminal kinase
LncRNA
Long non‑coding RNA
MaCSC
Mammary cancer stem cells
Mdm2
Murine double minute2
MUCL1
Mucin-like 1
NES
Nuclear export signal
NES2
Nuclear export signal 2
NSCLC
Non-small cell lung cancer
PDAC
Pancreatic ductal adenocarcinoma
PDGF
Platelet-derived growth factor
PDGFR
Platelet-derived growth factor receptor
PTK2
Protein tyrosine kinase 2
PRRs
Proline-rich regions
PyK2
Proline-rich tyrosine kinase 2
RIP
Receptor-interacting protein
RTKs
Receptor tyrosine kinases
Runx1
Runt-related transcription factor 1
SH3
Src homology 3
TLS
Tumor infiltrating lymphocytes
TM4LFM5
Tetraspanin transmembrane 4 L6 family member 5
TME
Tumor microenvironment
TNBC
Triple-negative breast cancer
UM
Uveal melanoma
VEGFR
Vascular endothelial growth factor receptor
VEGFR2
Vascular endothelial growth factor receptor 2
YAP
Yes-associated protein/yes-related protein

Background

Focal adhesion kinase is a tyrosine kinase composed of 1052 amino acids with a molecular weight of 125kD [1]. FAK is a crucial regulator of vital cellular processes, including cell adhesion [2], migration [3], proliferation [4], and survival [5]. Such processes have significant implications for the development and progression of cancer. Moreover, multiple studies have confirmed FAK upregulation in a diverse range of human malignancies, including colorectal, lung, ovarian, neck, bladder, breast, and esophageal cancers [6]. In addition, by promoting tumor angiogenesis, epithelial-mesenchymal transformation, cancer stemness, and immunomodulatory capacity [79], FAK significantly contributes to malignant progression. The available evidence suggests that FAK represents a promising target for cancer therapy. This article aims to provide an overview of the significant impact of FAK on both cancer cells and cancer-associated cells within tumors while also offering a comprehensive discussion of the recent advances made in developing therapeutic agents that target FAK and the combination of these agents with other approaches.

FAK structure and activation

Molecular structure of FAK

FAK is composed of three distinct domains: a four-point-one-ezrin-radixin-moesin (FERM) domain, a kinase domain, and a C-terminal focal adhesion-targeting (FAT) domain [10]. To target different sites on FAK, inhibitors specific to each domain have been developed (Fig. 1). The FERM domain is further divided into F1, F2, and F3 subdomains. F1 contains a nuclear export sequence, and F2 contains a nuclear localization sequence [11]. As the names indicate, these sequences are important for the nuclear transport of FAK. Over the past few years, research has emphasized the significance of nuclear FAK in the regulation of gene expression; it interacts with distinct E3 ligases to induce the degradation of transcription factors [12]. For example, the F1 and F2 lobes interact with p53, followed by the combination of the F3 lobe with murine double minute2 (Mdm2) and subsequent p53 ubiquitination and degradation, thus facilitating cancer cell proliferation and inhibiting apoptosis [13]. Nuclear FAK also alters expression of GATA4 and IL-33, suppressing the inflammatory response and inducing immune escape [1417]. Aside from controlling FAK transportation from the cytoplasm to the nucleus, the FERM domain also has a crucial function in triggering the activation of FAK. For example, the link between the FERM domain and the kinase domain prevents FAK activation by blocking the autophosphorylation of Y397, which is the sole autophosphorylation site of FAK [18]. The FERM domain contains binding sites for numerous proteins, such as integrin [19], growth factor receptors [20, 21], and G-protein coupled estrogen receptors [22], and such interactions trigger downstream signaling cascades and induces FAK activation. The KAKTLRK sequence, which is in the F2 lobe, also participates in FAK activation [23].
The bilobal central kinase domain, which contains catalytic sites and ATP-binding sites, is highly homological with other tyrosine kinases, particularly proline-rich tyrosine kinase 2 (Pyk2), and serves as the major component for FAK enzymatic activity [24]. Nuclear export signal 2 (NES2), which is regarded as the only biologically active nuclear export signal (NES) exerting nuclear export activities, is located in the central kinase domain [25].
The C-terminal region, which resembles a bundle composed of four helices, is primarily comprised of the FAT domain and has a significant role in controlling the activation of FAK. Through its interaction with proteins associated with focal adhesions such as paxillin and talin, the FAT domain prompts the recruitment and activation of FAK at the site of focal adhesions [26, 27]. This interaction is pivotal for FA assembly and turnover, which influences cell motility. In contrast, other studies have indicated that the FAT domain functions as an inhibitor of FAK activation by competing with the FERM domain for the binding of specific intracellular receptors and inducing dephosphorylation [28].
In addition to the three major domains, there are some special residues regulating FAK activation and function. FAK contains three proline-rich regions (PRRs), PRR1 localizes in the N-terminal domain, and PRR2/3 localizes in the C-terminal domain next to the FAT region. PRRs attach to proteins that contain Src homology 3 (SH3) domains, including small GTPases and p130Cas, to regulate kinase activity and support the cytoskeleton [29]. Additionally, there are at minimum six tyrosine residues that undergo phosphorylation throughout the entire region: Y397, Y407, Y576, Y577, Y861, and Y925. The autophosphorylation of Y397, which is situated at the N-terminus of the FERM structural domain, is vital for the activation of FAK [30]. Phosphorylated Y397 provides a binding site for Src-family kinases and other proteins containing the SH2 domain [31], which also interact with other tyrosine residues and are pivotal for FAK catalytic activity. Located in the activation loop of the kinase domain [32, 33], Y576 and Y577 positively regulate FAK kinase function, while another kinase domain-located tyrosine phosphorylation site, Y407, inhibits FAK activity. Additionally, Y861 and Y925 are in the C-terminal domains. Phosphorylated Y925 interacts with the SH2 domain of the adaptor protein GRB2 to trigger downstream Ras/MAPK signaling and induce integrin internalization and focal adhesion disassembly [34]. This FAK-GRB2-MAPK linkage was also demonstrated to be essential for tumor angiogenesis [35].

The mechanism and regulation of FAK activation

FAK is a vital mediator in the transmission of signals from the extracellular matrix (ECM) to the cell cytoplasm. It regulates pivotal cellular functions, including cell survival, proliferation, migration, and invasion (Fig. 2). In response to upstream stimuli, FAK initiates downstream signaling cascades, thereby prompting a series of events. Additionally, FAK is versatile as a signaling molecule because it can demonstrate kinase-dependent or kinase-independent activity [36]. Stimuli, integrin signaling activation, in particular, disrupts the inhibitory interaction between FERM and kinase domains, which leads to FAK dimerization and causes subsequent Y397 autophosphorylation [28]. The extensively phosphorylated Y397 has high affinity for the SH2 domain of kinases belonging to the Src family. When this interaction occurs, these kinases bind to and phosphorylate Y576 and Y577, which are positioned in the activation loop, to achieve complete activation of FAK and endow it with its enzymatic function [24].
The activation of FAK is regulated by various internal and external factors, which mainly target the FERM domain to induce conformational changes, thus relieving the autoinhibitory structure between the FERM and the kinase domain and stimulating FAK activation. In addition to integrin, the uncovered binding partners of the FERM domain include extracellular matrix, phosphoinositide lipids, diacylglycerol kinase α, serine/threonine kinase PKCθ, and membrane-associated proteins such as tetraspanin transmembrane 4 L6 family member 5 (TM4LFM5) and EMP2 [24, 28, 3739]. Intriguingly, glutathione peroxidase-1 can bind to FAK and prevent H2O2-induced oxidative inactivation of FAK [40]. Furthermore, growth factor receptors, including the Met receptor for hepatocyte growth factor, epidermal growth factor receptor, and platelet-derived growth factor receptor, enable conformational changes in the FERM domain by phosphorylating either the Y397 or Y197 sites [41, 42]. Other stimuli, such as elevated intracellular pH and increased matrix stiffness or forces, which occur during cancer progression, also trigger Y397 phosphorylation and FAK activation [43, 44]. Recently, it was revealed that FAK expression and activation are epigenetically regulated [45]. For example, microRNA miR-15b-5p inhibits FAK expression by binding to the 3′UTR of FAK mRNA. Moreover, intercellular adhesion molecule-1 suppresses miR-15b-5p activity and stimulates endothelial cell proliferation and migration [46]. The interaction between long noncoding RNA (lncRNA) MIR4435‑2HG and ganglioside synthesis enzyme ST8SIA1 induces the activation of FAK and downstream AKT/β‑catenin signaling, thus promoting prostate cancer cell viability [45]. Epidermal growth factor and IL-6, which are highly activated in glioblastoma, also initiate FAK activation [47].

The functions of FAK on tumor cells

Tumor cell proliferation, apoptosis and survival

The involvement of FAK in tumor growth has been extensively investigated in human breast cancer. The PI3K/AKT/mTOR signaling pathway has been widely recognized as one of the most commonly disrupted pathways in cancer [48, 49], and is correlated with FAK-mediated tumor cell growth. The ablation of FAK reduced Wnt1-driven basal-like breast cancer growth and promotes apoptosis by downregulating AKT-mTOR signaling [50]. In addition, activation of FAK by insulin-like growth factor-1 (IGF-1) and its receptor system (IGF-1R) initiates the PI3K-AKT-YAP (yes-associated protein/yes-related protein) signaling cascade, which modulates the expression of genes targeted by YAP. This mechanism is implicated in the expansion of aggressive triple-negative breast cancer cells [51]. A similar result was observed in intrahepatic cholangiocarcinoma (iCCA) [52]. FAK activation, which is required for Y357 phosphorylation of YAP, strongly promotes AKT/YAP-driven mouse iCCA initiation [52].
FAK was also confirmed to stimulate cell cycle progression and promote cancer proliferation by targeting various cyclins and cyclin-dependent kinase (CDK) inhibitors [53]. Among them, cyclin D1, together with key CDK inhibitors p21 and p27, are the most extensively studied downstream targets of FAK, regulating the cell cycle transition from G1 to S phase. It was reported that knockdown of Mucin-like 1 (MUCL1) in HER2-amplified breast cancer resulted in FAK/Jun NH2-terminal kinase (JNK) signaling blockade and subsequent G1/S phase arrest, which was mediated by decreased cyclin D levels as well as increased p21 and p27 levels [54]. A mechanistic study revealed that integrin signaling through FAK activated the ERK pathway, which stimulated the transcriptional activation of cyclin D [55]. In addition to p21 and p27, it was also demonstrated that FAK ablation in glioblastoma repressed the expression of the autophagy cargo receptor p62/SQSTM-1, the inhibition of which post transcriptionally upregulated p27 expression to mediate G1 phase arrest and induced a cell senescence-like state [56]. Moreover, p62 synergized with its downstream target SKP2 to inhibit p21 and p27 activity. Nevertheless, contradictory results were observed in vascular smooth muscle cells: SKP2 was degraded by nuclear FAK to inhibit cell proliferation by promoting p21 and p27 expression [57]. It has been acknowledged that nuclear FAK acts as a scaffold for protein interactions and regulates specific gene transcription. In skin squamous cell carcinoma, nuclear FAK was reported to interact with runt-related transcription factor 1 (Runx1) and recruit Runx1 regulatory proteins such as sin3a to inhibit the transcription of insulin-like growth factor binding protein 3, which induces cell cycle arrest at the G1 phase by suppressing the expression of cyclins and CDKs and increasing p21 expression [58]. Likewise, the nuclear activation of FAK in colon cancer cells by fibrin resulted in a decline in p53, along with its subsequent targets, such as 14-3-3σ and p21, ultimately stimulating cell proliferation while repressing senescence [59].
FAK plays a vital role in sustaining cancer cell survival and regulating cell apoptosis [6063], anoikis [6468], autophagy [68], and senescence [69]. The disruption of FAK-mediated signaling in breast cancer cells stimulated the Fas-associated death domain (FADD) and caspase-8 apoptotic pathways, which induced cell apoptosis and inhibited anchorage-independent survival [70]. Additionally, components of death receptor pathways, including the FAK-binding partner death domain kinase receptor-interacting protein (RIP), are involved in this process [61]. The antiapoptotic effect of FAK was further confirmed to be associated with FAK-dependent activation of the phosphatidylinositide 3′-OH-kinase-AKT survival pathway, concomitant with the subsequent stimulation of NF-kB and inhibitor-of-apoptosis proteins [71]. Moreover, nuclear FAK also plays an important role in regulating cancer cell apoptosis. Nuclear FAK interacts with the N-terminal transactivation domain of p53 through its N-terminal fragment; this attenuates p53 transcriptional activity and inhibits p53-mediated apoptosis to promote cell survival [72]. Anoikis, a distinct type of apoptosis that occurs in normal epithelial and endothelial cells, is also negatively regulated by FAK signaling In human breast cancer, the mechanism of FAK-induced cell resistance to anoikis was reported to be correlated with the increased activity of NF-kB, which is induced by the functional interaction between the N-terminal domain of FAK and TRAF2, a RING finger adaptor protein [65]. Notably, integrin endocytosis has gradually been uncovered to be critical for FAK activation depending on endosome antigen-1 and small GTPase Rab21, and FAK activation ultimately promotes anoikis resistance and anchorage-independent cell growth [66, 67].

Cell migration and invasion

Integrin aggregation in the ECM plays a crucial role in inducing FAK signaling, which is fundamental for cell motility and cytoskeletal reorganization. Chemotactic signals activate integrins, leading to FAK activation and the formation of focal adhesion complexes, which subsequently trigger the polymerization of actin filaments toward the cytoplasmic membrane [73, 74]. The FAK/Src complex and kinase activity lead to p130Cas phosphorylation, promoting the formation of Cas/Crk complexes, which significantly influence cell migration [75]. Additionally, MLCK-mediated focal adhesion disassembly and JNK-mediated paxillin phosphorylation promote cytoskeleton reorganization [76, 77]. FAK’s associations with PI3-kinase and/or Grb7 govern intracellular signaling pathways correlated with cellular mobility [78, 79]. Furthermore, integrin-mediated FAK signaling critically controls adhesion dynamics during cell migration. The formation of FAK/Src complexes at focal adhesion sites enhances ERK2 activity, resulting in the activation of Calpain 2 [8082]. FAK’s effects on small GTPases impact cytoskeletal reorganization and adhesion stabilization [83]. RhoA, Rac1, and Cdc42, among small GTPases, play a crucial role in cytoskeletal reorganization and tumorigenesis [84, 85]. In this regard, RhoA influences cell‒cell or cell–ECM associations by inducing shifts within the cytoskeleton, while Rac1 initiates actin polymerization, enabling membrane folding, whereas Cdc42 initiates actin filament production in the generation of filopodia [8688]. These investigations present compelling proof that the expression of FAK and the activation of FAK signaling pathways are mainly mediated by Rho GTPases, indicating the indispensable role of FAK in cytoskeletal reorganization.

Epithelial–mesenchymal transformation

Epithelial–mesenchymal transition (EMT) is a physiological process in normal embryonic development and tissue regeneration. However, aberrant reactivation of EMT is associated with malignant properties of tumor cells, including migration, invasiveness, increased tumor stemness, and resistance to chemotherapy and immunotherapy [88, 89]. Multiple studies have established the role of FAK in promoting EMT and increasing cell invasion and metastasis [90, 91]. EMT is regulated by FAK-mediated alterations in E-cadherin expression, a key molecule in the process [9294]. Evidence provided by Gayrard et al. illustrates that SRC-FAK-mediated reconstruction of actomyosin results in the loosening of E-cadherin junctions without disrupting those involving β-associated proteins [94]. Avizienyte’s team confirmed the importance of FAK phosphorylation in the decrease in E-cadherin induced by Src in colon cancer cells [91]. The investigation carried out by Hauck’s team suggests that restraining FAK function restricts cell invasion stimulated by Src and obstructs the metastasis and invasion aimed by FAK-targeted drugs [95]. The downregulation of KIF26A clearly enhances EMT and decreases E-cadherin expression by augmenting the binding of c-MYC to the promoter section of FAK [96]. Slug expression, which balances EMT and cellular migration, is triggered by TGF-β1 in squamous cell carcinoma cells. However, when FAK inhibitors are administered, such an effect is alleviated [97]. These findings underline the crucial role of FAK in EMT, invasion, and metastasis. Nonetheless, additional research is required to clarify the downstream molecular mechanisms by which FAK regulates EMT. These mechanisms include E-cadherin-mediated cell‒cell adhesion, integrin-ECM-based adhesion, and the collaboration of both these mechanisms.

FAK in cancer stem cells

In various tumor types, FAK has been found to contribute to the activities of cancer stem cells (CSCs), particularly in breast cancer [98, 99]. Loss of FAK leads to a decrease in mammary cancer stem cells (MaCSCs) and suppresses their protumorigenic functions [100]. FAK facilitates the formation of a ternary complex with connexin and NANOG, which sustains CSC self-renewal and maintenance in triple-negative breast cancer [101]. By interrupting the interaction between FAK and endophilin A2, the stem-like population, gene signature, self-renewal, and tumorigenicity of mammary CSCs can be suppressed [102]. In triple-negative breast cancer (TNBC), inhibiting FAK genetically or with drugs decreases anchorage-independent spheroid cell growth, reduces chemotherapy-dependent CSC enrichment, and delays metastatic outgrowth [99, 103]. A recent study reported that inhibiting FAK, a protein found in head and neck squamous cell carcinoma (HNSCC), could significantly reduce the expression of stem cell markers, including Oct4, Sox2, and Nanog, leading to a decrease in cell self-renewal [104]. Additionally, FAK-mediated signaling pathways have been discovered to play a crucial role in regulating CSC properties in esophageal squamous cell carcinoma [105].

The effects of FAK on tumor-associated cells

Immune cells

Tumor-associated macrophage and regulatory T cells (Treg) are major inhibitory cells for anti-cancer immune response [106108]. The importance of FAK expression in the regulation of the tumor environment has been emphasized in current research (Fig. 3). Enhancement of the expression of several chemokines occurs due to the elevation of FAK levels in tumors, which promotes TME remodeling by recruiting immunosuppressive cells and secreting cytokines [109]. FAK inhibitors have been shown to suppress leucocyte and macrophage infiltration and the growth of breast cancer [110, 111] and pancreatic ductal adenocarcinoma (PDAC) tumors in mouse models [112]. FAK signaling enhances the expression of homing signals such as CCL5, CCL7, CXCL10, and TGFβ2 [113], which play a crucial role in recruiting Tregs [114]. FAK contributes to the increased expression of IL-33 [115], an alarmin cytokine, produced by stromal and epithelial cells, which binds to ST2L on immune cells and enhances the transcription of chemokine genes such as CCL5 [116]. The increased production of CCL5 leads to the recruitment of Tregs and other immune cells, promoting immunosuppression. However, recruited Tregs promote tumor survival by depleting CD8+ cytotoxic cells [117]. CD28 is a costimulatory molecule on T cells that enhances T-cell activation and proliferation. Amplification of CD28+ T cells within the TME can enhance their antitumor effects and facilitate tumor cell elimination. FAK depletion can lead to tumor regression by increasing the number of CD28+ T cells in the TME [118]. FAK overexpression slows tumor growth and promotes natural killer cell infiltrations while FAK knockdown promotes tumor growth and suppresses natural killer cell infiltrations [119].

Cancer-associated fibroblasts (CAFs)

In the TME, CAFs are key stromal cells that play a critical role in tumor cell initiation, survival, proliferation, and metastasis through the secretion of various cytokines, growth factors, hormones, and ECM proteins [120]. For example, increased lumican expression in gastric CAFs promotes FAK activation via β1 integrin, promoting the invasion of gastric cancer cells [121]. PDAC cells activate CAFs and promote cancer stemness through increased expression of type I collagen via β1 integrin-FAK signaling [122]. Inhibition of FAK reduces CAF recruitment and TME fibrosis [123]. This reduces the stemness of PDAC cells [124] and suppresses breast cancer metastasis while increasing the levels of tumor suppressor microRNAs in exosomes [125]. Emerging evidence highlights the pivotal role of CAFs in governing tumor metabolic processes via FAK-regulated pathways. Notably, breast and pancreatic cancer patients exhibiting diminished FAK expression experience a significant decline in overall survival. Furthermore, experimental studies using mouse models have demonstrated that the depletion of FAK in CAFs actively promotes tumor growth. Mechanistically, this phenomenon can be attributed to the activation of protein kinase A within CAFs, resulting from the deficiency of FAK. Consequently, this activation leads to a pronounced enhancement of glycolysis in tumor cells [126]. The remodeling of the TME is facilitated by FAK, which functions as a crucial regulator in the TME. Therefore, a comprehensive understanding of the impact of FAK on tumor progression and TME remodeling could reveal new opportunities for cancer therapy.

Endothelial cells (ECs)

Integrins and growth factor receptors mediate the signals involved in angiogenesis. FAK is activated by integrin-mediated cell adhesion and associates with several proteins that contain the SH2 structural domain, including Src, Grb7, the p85 subunit of PI3K, and phospholipase C-g [127]. FAK interacts with epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), and platelet-derived growth factor receptor (PDGFR) via its N-terminus, and VEGF induces FAK phosphorylation through VEGFR activation, promoting angiogenesis [128]. When platelet-derived growth factor (PDGF) binds to PDGFR, it phosphorylates FAK, which activates endothelial cells, stromal cells, and CEPs, leading to matrix metalloproteinase-mediated breakdown of the ECM and angiogenesis [129]. FAK inhibition is a promising anticancer treatment strategy to hinder cell migration, invasion, proliferation, and angiogenesis. Inhibition of the phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2), Src, and FAK through Sema3A led to substantial reductions in tumor growth and angiogenesis in tongue SSC-9 cells, illustrating the potential of this approach for cancer therapy [63]. TAE226, a potent FAK inhibitor, effectively suppressed the growth of OSCC xenografts and angiogenesis in mice [130]. Moreover, FRNK, a negative inhibitor of FAK, was revealed to impede FAK phosphorylation, thereby reducing EGF-induced MMP-9 expression and ultimately hindering the invasion of follicular thyroid cancer cells [131]. These collective findings affirm the pivotal role of FAK in inducing cell invasion and angiogenesis and its potential as an attractive target for antiangiogenic therapy in cancer treatment.

Targeting FAK in combination therapies

FAK is considered a potential target for effective cancer therapy, and its key role in various types of cancer has been well established. Recently, FAK inhibitors have gained attention as novel and promising combination therapy partners (Fig. 3). Presented here is a summary of preclinical (Table 1) and clinical trials (Table 2) concerning FAK inhibitors. The primary focus is on combination trials incorporating FAK as a therapeutic target (Table 3) to assess its efficacy and its contribution to combination therapy.
Table 1
Summary of FAK inhibitors in preclinical trials
Name
Alternative names
Target(s)
Cancer types
In vitro
In vivo
References
VS-6062
PF-562, 271
FAK, Pyk2
Glioma
+
+
[175]
Colon cancer
+
+
[175]
Breast cancer
+
+
[175]
Pancreatic cancer
+
+
[112, 175]
Prostate cancer
+
+
[175177]
Lung cancer
+
+
[175, 178]
Hepatocellular cancer
+
+
[179]
Thyroid tumor
+
+
[180]
B16
+
[181]
Ovarian cancer
+
+
[182]
Ewing sarcoma
+
+
[183]
VS-6063
Defactinib, PF-04554878
FAK, Pyk2
Pancreatic cancer
+
+
[156, 184]
Lung cancer
+
+
[5, 164, 165]
Prostate cancer
+
+
[188]
Ovarian cancer
+
+
[189, 190]
Esophageal cancer
+
+
[191]
Endometrial cancer
+
+
[192]
BI-853520
IN10018, ifebemtinib
FAK
Breast cancer
+
+
[193]
KRAS G12C mutant cancer
+
+
[145]
Pancreatic cancer
+
+
[162]
Ovarian cancer
+
+
[145, 194]
Prostate cancer
+
+
[195]
PF-573228
PF-228
FAK
Pancreatic cancer
+
+
[196]
Glioma
+
[56]
Lung cancer
+
[197]
TAE226
NVP-226
FAK, IGF-IR
Breast cancer
+
+
[198, 199]
Glioma
+
+
[200, 201]
Esophageal cancer
+
+
[202, 203]
GSK2256098
FAK
Pancreatic cancer
+
[204]
Ovarian cancer
+
[205]
PF-431396
FAK, Pyk2
Pancreatic cancer
 + 
 + 
[196]
Malignant pleural mesothelioma
+
+
[196]
VS-4718
PND-1186
FAK, Pyk2
Breast cancer/ovarian cancer
+
+
[206]
Pancreatic cancer
+
[123]
Y15
FAK
Breast cancer
+
+
[207]
Lung cancer
+
+
[208]
Y11
FAK
Colon cancer/breast cancer
+
+
[176, 204]
C4
FAK-VEGFR3 interaction
Breast cancer
+
+
[209]
R2
FAK-p53 interaction
CRC
+
+
[210]
Table 2
Summary of FAK inhibitors in clinical trials
Name
Alternative names
Target(s)
Cancer types
Status
Phase
NCT number
VS-6063
Defactinib, PF-04554878
FAK, Pyk2
Advanced non-hematologic Malignancies
Completed
1
NCT00787033
Non-hematologic cancer
Completed
1
NCT01943292
NSCLC
Completed
2
NCT01951690
Recurrent skin cancer, squamous cell carcinoma of the skin, stage 0 chronic lymphocytic leukemia, stage I chronic lymphocytic Leukemia
Completed
2
NCT00563290
Ovarian cancer
Completed
1
NCT01778803
Solid tumor, pancreatic cancer
Completed
1
NCT02546531
NSCLC, low grade serous ovarian cancer, endometrioid carcinoma, pancreatic cancer
Recruiting
1
NCT03875820
Metastatic uveal melanoma
Recruiting
2
NCT04720417
Pancreas cancer
Recruiting
2
NCT04331041
Ovarian cancer
Recruiting
1
2
NCT03287271
PDAC
Recruiting
2
NCT03727880
Advanced lymphoma, advanced malignant solid neoplasm, hematopoietic and lymphoid cell neoplasm, refractory lymphoma, refractory malignant solid neoplasm, refractory plasma cell myeloma
Active, not recruiting
2
NCT04439331
Glioma
Not yet recruiting
Early 1
NCT05798507
Malignant pleural mesothelioma
Terminated
2
NCT02004028
CRC
Terminated
Early 1
NCT00835679
Relapsed malignant mesothelioma
Terminated
1
NCT02372227
NSCLC, mesothelioma, pancreatic neoplasms
Unknown status
1
2
NCT02758587
BI 853520
IN10018, ifebemtinib
FAK
Neoplasms
Completed
1
NCT01335269
Gastric cancer
Completed
1
NCT05327231
Metastatic melanoma
Recruiting
1
NCT04109456
Pancreatic cancer
Recruiting
1
2
NCT05827796
Platinum-resistant ovarian cancer
Recruiting
1
2
NCT05551507
Locally advanced or metastatic solid tumor
Active, not recruiting
1
2
NCT05830539
Solid tumor
Not yet recruiting
1
2
NCT05379946
GSK2256098
FAK
Solid tumor
Completed
1
NCT01138033
Cancer
Completed
1
NCT00996671
Pancreatic cancer, adenocarcinoma
Completed
2
NCT02428270
Advanced solid tumor
Completed
1
NCT01938443
Intracranial meningioma, recurrent meningioma, Nf2 gene mutation
Recruiting
2
NCT02523014
VS-4718
PND-1186
FAK, Pyk2
Pancreatic cancer
Terminated
1
NCT02651727
Non-hematologic cancer, metastatic cancer
Terminated
1
NCT01849744
Relapsed or refractory acute myeloid leukemia, relapsed or refractory B-cell acute lymphoblastic leukemia
Withdrawn
1
NCT02215629
CT-707
Conteltinib
FAK, ALK, Pyk2
Advanced pancreatic cancer
Recruiting
1
2
NCT05580445
NSCLC
Unknown status
1
NCT02695550
VS-6062
PF-00562271
FAK, Pyk2
Head and neck neoplasm, prostatic neoplasm, pancreatic neoplasm
Completed
1
NCT00666926
AMP-945
Narmafotinib
FAK
PDAC
Recruiting
1
2
NCT05355298
APG-2449
FAK, ALK, ROS1
Advanced solid cancer, NSCLC, esophageal cancer, ovarian cancer, malignant pleural mesothelioma
Recruiting
1
NCT03917043
Table 3
Combination agents of FAK inhibitors
Name
Alternative names
Target(s)
Combination agents
Preclinical trials
Clinical trials
Cancer types
References
Cancer types
NCT number
VS-6063
Defactinib, PF-04554878
FAK, Pyk2
Pembrolizumab (a PD1 inhibitor)
NSCLC, mesothelioma, pancreatic tumor
NCT02758587
PDAC
NCT03727880
Solid tumor, PDAC
NCT02546531
VS-6766 (a RAF/MEK inhibitor)
NSCLC, ovarian cancer, endometrioid carcinoma, pancreatic cancer
NCT03875820
Metastatic uveal melanoma
NCT04720417
Paclitaxel
Ovarian cancer
[189]
Ovarian cancer
NCT01778803
Pancreatic cancer
[156]
Ovarian cancer
NCT03287271
Gemcitabine
Lung cancer
[185]
Solid tumor, pancreatic cancer
NCT02546531
VS-5584
NCT00835679 (a PI3K inhibitor)
Relapsed malignant mesothelioma
NCT02372227
Radiation therapy
Pancreatic cancer
[184]
Pancreas cancer
NCT04331041
Cetuximab
CRC
NCT00835679
BRD4 inhibitor
lung cancer
[186]
EGFR-TKI
Lung cancer
[187]
Docetaxel
Prostate cancer
[188]
BI 853520
IN10018, ifebemtinib
FAK
Albumin-bound paclitaxel, gemcitabine, KN046
Pancreatic cancer
NCT05827796
Cobimetinib, atezolizumab
Metastatic melanoma
NCT04109456
PLD
Platinum-resistant ovarian cancer
NCT05551507
PLD, toripalimab
Locally advanced or metastatic solid tumor
NCT05830539
Docetaxel
Gastric cancer
NCT05327231
D-1553
Solid tumor
NCT05379946
KRAS G12C inhibitors
CRC, pancreatic cancer, NSCLC
[145]
Radiation therapy
Pancreatic cancer
[162]
Paclitaxel
Ovarian cancer
[194]
VS-6062
PF-562,271
FAK, Pyk2
Gemcitabine
Pancreatic cancer
[112]
Sunitinib
Hepatocellular cancer
[179]
anti-VEGF
Ovarian cancer
[182]
AZD-1152
Ewing sarcoma
[183]
VS-4718
PND-1186
FAK, Pyk2
Nab-paclitaxel, gemcitabine
Pancreatic cancer
NCT02651727
Gemcitabine, adoptive cell transfer (ACT), Anti-PD1/anti-CTLA4
Pancreatic cancer
[123]
GSK2256098
FAK
Trametinib (a MEK inhibitor)
Advanced solid tumor
NCT01938443
Pazopanib
Ovarian cancer
[205]
CT-707
Conteltinib
FAK, ALK, Pyk2
Toripalimab, gemcitabine
Advanced pancreatic cancer
NCT05580445
TAE226
NVP-226
FAK, IGF-IR
Radiation therapy
Glioma
[201]

Combination with immunotherapies

Antibodies to immune check point inhibitor that enhance the host immunologic activity against tumors have become standard of care in the treatment of many malignancies [132]. However, only a small percentage of patients have meaningful responses to these treatments. Searching for new pathways and molecules to improve responses and application of immune checkpoint inhibition therapy attracts great attention [133136]. In TNBC, PD-L1 expression is elevated and significantly correlated with FAK mRNA expression, highlighting the functional relationship between immune checkpoints and FAK [137, 138]. Anti-PD-L1 antibody atezolizumab augments the suppressive impact of FAK inhibitors on cell invasion and migration through the restraint of FAK phosphorylation [138]. Cytokine-induced killer (CIK) cells are used as a treatment approach in adoptive cellular immunotherapy and are highly regarded as a promising candidate for cancer immunotherapy [139]. FAK knockdown/inhibition increased the sensitivity of TNBC cells to CIK cells in coculture system by enhancing CIK-mediated cell death. FAK knockdown also decreased PD-L1 mRNA and protein expression in TNBC cells [140].
Combining the FAK inhibitor VS4718 with anti-PD1 therapy in hepatocellular carcinoma resulted in decreased macrophage numbers and increased CD8+ T-cell numbers [141]. Additionally, when FAK inhibitors were combined with agents that induce T-cell costimulatory pathways in skin squamous cell carcinoma, the tumors became more sensitive to FAK inhibitors, and this effect was mediated by CD80. This suppressed tumor formation and even drove complete regression [118].
Lu et al. constituted a nanodrug PLGA-FAKi by encapsulated FAK inhibitor using poly(lactic-co-glycolic) acid (PLGA). PLGA-FAKi treatment increased ovalbumin-specific CTLs (OVA-CTLs) infiltration into B16-OVA tumors, leading to reduced immunosuppression and increased tumor microvessel permeability, and further inhibited tumor growth when combined with OVA-CTLs [70]. In mice with HGSOC, the combination of FAK inhibitor and anti-TIGIT therapy was able to prolong survival rates, increase the level of CXCL13, which is associated with tumor infiltrating lymphocytes (TLS) formation, and promote B and T-cell enrichment [142]. Mechanical stretching has also been shown to have a positive effect on melanoma cells: it enhances M1 polarization and antitumor effects. This effect is associated with the FAK/NF-kB signaling pathway [143]. It was found that ABCB1, CXCR4, and FAK were overexpressed in non-small cell lung cancer (NSCLC) patients and cell lines [144]. Therefore, targeting CXCR4 and FAK could be a way to overcome DOX resistance and enhance the anti-invasive effects of CXCR4 and FAK inhibitors in NSCLC cells.

Combination with targeted therapies

Research suggests that co-treatment using KRAS G12C inhibitors and IN10018 is likely to benefit cancer patients with mutated KRAS G12C and may also prevent resistance to KRAS G12C inhibition by targeting dysregulated FAK-YAP signaling and fibrogenesis [145]. Uveal melanoma (UM) patients with unresponsive liver metastases have a druggable downstream signaling hub from GNAQ mutations that activates YAP1 via FAK [146]. Co-targeting FAK and MEK using this approach could lead to novel precision therapy and inhibit tumor growth in UM cells and UM xenograft models. It is important to note that FAK is overexpressed in tumors and activated in iCCA lesions, which in turn contribute to cancer initiation and progression through the YAP proto-oncogene [52]. iCCA growth was dramatically inhibited by combination of FAK and CDK4/6 inhibitor. Overall, the study proved the role of FAK-YAP signaling and suggests its potential as a target for precision therapy in inhibiting cancer growth using various approaches.
Co-targeting this pathway using the FAK inhibitor PF562271 and the BRAF inhibitor vemurafenib could represent a promising therapeutic approach for BRAF-mutant colorectal cancer (CRC) patients, as they exhibit synergistic antitumor effects in vitro and in vivo [147]. Notably, the combination of small-molecule inhibitors of β-catenin or FAK along with vemurafenib not only inhibits the proliferation of BRAF V600E colon cancer cells in vitro but also prevents tumor formation in xenograft mice [148]. Overall, these findings emphasize the potential of combination therapy using several inhibitors in treating cancers with mutations in BRAF and highlights the potential of FAK inhibitors in several therapeutic approaches.
One study found that using TAE226 and sorafenib together effectively reduces hepatocellular carcinoma growth by changing gene expression and epigenetics through FAK nuclear interactome dysregulation [149]. Another study showed that combining VS-6063 and JQ1 to target integrins inhibits FAK signaling and PI3K/AKT, reducing survival in primary HGSOC tumors with co-amplification of FAK and c-Myc [150]. In squamous cell carcinoma cells with mutated FAK, HDAC and FAK inhibitors work synergistically to arrest cellular proliferation and tumor growth, emphasizing the importance of collaborations of multiple targets [151]. In addition, combining SFK/FAK inhibitors with osimertinib shows promise as a therapeutic approach to inhibit growth and resistance in EGFR-mutant lung cancer treatment [152]. Simultaneous targeting of the FAK and Janus kinase/STAT3 pathways produces a synergistic effect. This suggests that repressing STAT3 signals may overcome FAK inhibitor resistance in PDAC, as demonstrated in another study [153]. The studies described above provide evidence that FAK inhibitors, when combined with targeted therapy, present a new and promising avenue of tumor treatment.

Combination with chemotherapies

Chemotherapy is often ineffective against ovarian cancer; however, the hyaluronic acid-labeled two-in-one drug delivery system HA-PLGA-NPs, containing paclitaxel and FAK siRNA, has high binding efficiency to CD44-positive tumor cells, resulting in increased cytotoxicity and apoptosis in drug-resistant tumors, as demonstrated in experimental studies [154]. FAK inhibition has been identified to enhance chemotherapy sensitivity and promote anticancer effects, primarily through the activation of p53 transcriptional activity, making it a potential focal point for therapeutic strategies in gastric cancer management and a valuable prognostic indicator in clinical settings [155]. Inhibition of FAK, as observed in the study of phosphorylated kinases in PDAC, shows synergistic effects with nab-paclitaxel to reduce tumor growth and appears to be a promising potential treatment option [156]. Furthermore, endothelial cell focal adhesion kinase (EC-FAK) plays a significant role in the regulation of the chemotherapy response and the levels of endocrine factors, and the combination of FAK inhibitors with gemcitabine has the potential to serve as a promising strategy to control PDAC metastasis, as supported by studies that revealed reduced metastasis load and improved survival rates in gemcitabine-treated mice and patients with low levels of EC-FAK [157]. FAK regulates CSC activity in breast cancer, and inhibition of FAK suppresses self-renewal, leading to a reduced tumor size, thereby providing a promising strategy to improve survival by suppressing CSC activity; the approach is especially effective when combined with paclitaxel treatment [98]. In addition, inhibiting endothelial FAK enhances the response of B16 and CMT19T mouse tumors to adriamycin or radiotherapy by suppressing NF-κB activation and cytokine production, thereby improving the effectiveness of DNA damage therapy [158]. Furthermore, elevated EC-PY397-FAK expression levels are strongly correlated with advanced clinical parameters of breast cancer and poor treatment response and independently predict unfavorable five-year recurrence-free survival, which highlights the need to assess the role of FAK inhibitors in optimizing treatments and improve the response to various strategies [159].

Combination with radiotherapies

Combining FAK inhibitors with low-dose radiation in pancreatic cancer can regulate the TME through several mechanisms, including reducing hypoxia, boosting immune cell infiltration, and enhancing radiosensitivity [160]. FAK overexpression is known to be associated with treatment resistance and metastasis in pancreatic cancer. A database study identified VS-4718 as a potential inhibitor of FAK that can enhance radiosensitivity and inhibit ECM synthesis [161]. Combining FAK inhibition with radiotherapy may prove to be effective against this disease. Tests in a PDAC mouse model have shown that inhibiting FAK with IN10018 can enhance the anticancer effect of radiotherapy by decreasing suppressor granulocyte infiltration and increasing CD8+ T cell and macrophage [162]. This indicates that FAK inhibitors have the potential to enhance the radiosensitivity and immunomodulation of PDAC.
The presence of CSCs in high-grade DCIS is associated with disease recurrence and resistance to radiotherapy via the FAK/Wnt pathway, and the use of FAK inhibitors can reduce cellular self-renewal while enhancing the effects of radiation in breast cancer [163]. Moreover, inhibiting FAK decreased tumor cell adhesion in a glioblastoma/breast cancer cell and endothelial cell coculture model after radiotherapy [164]. The evidence suggests that FAK inhibitors hold promise for enhancing radiosensitivity.
Similarly, a study demonstrated that the combination of FAK inhibition and carbon ion irradiation was effective in inhibiting metastasis in tongue squamous cell carcinoma. The treatment decreased colony formation, increased apoptosis, and reduced migration and invasion in CAL27 cells [165]. Furthermore, in HPV-negative HNSCC cells, FAK inhibition led to enhanced radiosensitivity by inducing G2/M arrest and DNA damage. The study also revealed that lower protein tyrosine kinase 2 (PTK2)/FAK mRNA expression was linked to better disease-free survival. Therefore, PTK2/FAK could be a potential biomarker for HNSCC patients who are susceptible to relapse after radiotherapy [5]. According to the available data, FAK inhibitors have shown promising results in sensitizing cancer cells to the effects of ionizing radiation, which helps reduce tumor burden and recurrence rates.

Conclusion

Cancer is a complex illness that arises due to diverse genetic and epigenetic alterations that cause abnormalities in multiple biological pathways. Although the development of molecularly targeted therapies has aided in their treatment, their frequent ineffectiveness and drug resistance pose significant challenges owing to recurrence and metastasis s; therefore, new therapeutic targets are urgently needed. Crucial cellular processes such as cell adhesion, migration, proliferation, and survival are regulated by FAK. Furthermore, FAK promotes cancer progression, including features such as tumor angiogenesis, EMT, cancer stemness, and immunomodulatory capacity [166, 167]. FAK is widely activated in multiple cancer types, such as colorectal, lung, ovarian, neck, bladder, breast, and esophageal cancers, and predicts a poor prognosis [98, 168, 169].
The FAK pathway has also been linked to the generation of CSCs [101, 103, 105, 114], which are responsible for tumor propagation, metastasis, and therapy resistance. FAK inhibition has been shown to decrease the number of CSCs, suggesting that FAK may represent a viable target for eliminating CSCs, thereby improving cancer therapy outcomes. In addition, the FAK signaling pathway plays a crucial role in regulating the complex TME [119, 125, 129, 170], which comprises cellular and noncellular components that promote tumor growth and metastasis. Gaining a comprehensive understanding of the involvement of FAK in tumor microenvironment (TME) remodeling is imperative in advancing cancer treatment outcomes to a higher level.
FAK has emerged as a promising target for cancer therapy owing to its key role in tumor cells and the TME. Various FAK inhibitors have demonstrated significant antitumor efficacy in diverse preclinical models and are currently being evaluated in clinical trials. Combining FAK inhibitors with standard cancer treatments has been shown to significantly enhance treatment efficacy and decrease chemotherapy resistance [12, 170], as FAK inhibition sensitizes cancer cells to chemotherapy, leading to better therapeutic outcomes. It is interesting that D-pinitol, a 3-methoxy analogue of d-chiro-inositol in soy foods and legumes, can reduce c-Src kinase activity and NF-kB activation through inhibiting FAK phosphorylation, resulting in decrease of prostate cancer metastasis [171, 172]. Recently, an effective FAK degradation agents have been developed that can selectively degrade FAK and showed outstanding inhibitory effects in triple-negative breast cancer and ovarian cancer cells [173, 174].
However, the clinical translation of FAK inhibitors has been hampered by several challenges. First, a uniform method for measuring the expression of FAK, whether phosphorylated FAK or total FAK, needs to be selected. Second, the selection of an appropriate FAK assay is necessary, and immunohistochemistry, western blotting, and RT‒PCR are the most commonly employed methodologies. Each method has advantages and limitations, and the selection must account for factors such as sensitivity, specificity, and reliability. Finally, there are challenges regarding combination therapies utilizing FAK, including reduced selectivity and specificity, drug resistance, and emerging molecular targets that impact efficacy. As a result, future research should aim to enhance the selectivity and specificity of FAK inhibitors and also develop novel combination therapies to overcome these therapeutic obstacles. By addressing these challenges, the clinical translational impact of FAK-targeted therapies in patients can be optimized, ultimately resulting in more effective and personalized cancer treatments.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Guan JL, Shalloway D. Regulation of focal adhesion-associated protein tyrosine kinase by both cellular adhesion and oncogenic transformation. Nature. 1992;358:690–2.PubMed Guan JL, Shalloway D. Regulation of focal adhesion-associated protein tyrosine kinase by both cellular adhesion and oncogenic transformation. Nature. 1992;358:690–2.PubMed
2.
Zurück zum Zitat Nader GPF, Ezratty EJ, Gundersen GG. FAK, talin and PIPKIγ regulate endocytosed integrin activation to polarize focal adhesion assembly. Nat Cell Biol. 2016;18:491–503.PubMed Nader GPF, Ezratty EJ, Gundersen GG. FAK, talin and PIPKIγ regulate endocytosed integrin activation to polarize focal adhesion assembly. Nat Cell Biol. 2016;18:491–503.PubMed
3.
Zurück zum Zitat Fan T, Chen J, Zhang L, Gao P, Hui Y, Xu P, et al. Bit1 knockdown contributes to growth suppression as well as the decreases of migration and invasion abilities in esophageal squamous cell carcinoma via suppressing FAK-paxillin pathway. Mol Cancer. 2016;15:23.PubMedPubMedCentral Fan T, Chen J, Zhang L, Gao P, Hui Y, Xu P, et al. Bit1 knockdown contributes to growth suppression as well as the decreases of migration and invasion abilities in esophageal squamous cell carcinoma via suppressing FAK-paxillin pathway. Mol Cancer. 2016;15:23.PubMedPubMedCentral
4.
Zurück zum Zitat Balsas P, Palomero J, Eguileor Á, Rodríguez ML, Vegliante MC, Planas-Rigol E, et al. SOX11 promotes tumor protective microenvironment interactions through CXCR4 and FAK regulation in mantle cell lymphoma. Blood. 2017;130:501–13.PubMed Balsas P, Palomero J, Eguileor Á, Rodríguez ML, Vegliante MC, Planas-Rigol E, et al. SOX11 promotes tumor protective microenvironment interactions through CXCR4 and FAK regulation in mantle cell lymphoma. Blood. 2017;130:501–13.PubMed
5.
Zurück zum Zitat Skinner HD, Giri U, Yang L, Woo SH, Story MD, Pickering CR, et al. Proteomic profiling identifies PTK2/FAK as a driver of radioresistance in HPV-negative head and neck cancer. Clin Cancer Res. 2016;22:4643–50.PubMedPubMedCentral Skinner HD, Giri U, Yang L, Woo SH, Story MD, Pickering CR, et al. Proteomic profiling identifies PTK2/FAK as a driver of radioresistance in HPV-negative head and neck cancer. Clin Cancer Res. 2016;22:4643–50.PubMedPubMedCentral
6.
Zurück zum Zitat Yoon H, Dehart JP, Murphy JM, Lim S-TS. Understanding the roles of FAK in cancer: inhibitors, genetic models, and new insights. J Histochem Cytochem. 2015;63:114–28.PubMed Yoon H, Dehart JP, Murphy JM, Lim S-TS. Understanding the roles of FAK in cancer: inhibitors, genetic models, and new insights. J Histochem Cytochem. 2015;63:114–28.PubMed
7.
Zurück zum Zitat Chuang HH, Zhen YY, Tsai YC, Chuang CH, Hsiao M, Huang MS, et al. FAK in cancer: from mechanisms to therapeutic strategies. Int J Mol Sci. 2022;23:1726.PubMedPubMedCentral Chuang HH, Zhen YY, Tsai YC, Chuang CH, Hsiao M, Huang MS, et al. FAK in cancer: from mechanisms to therapeutic strategies. Int J Mol Sci. 2022;23:1726.PubMedPubMedCentral
8.
Zurück zum Zitat Rigiracciolo DC, Cirillo F, Talia M, Muglia L, Gutkind JS, Maggiolini M, et al. Focal adhesion kinase fine tunes multifaced signals toward breast cancer progression. Cancers. 2021;13:645.PubMedPubMedCentral Rigiracciolo DC, Cirillo F, Talia M, Muglia L, Gutkind JS, Maggiolini M, et al. Focal adhesion kinase fine tunes multifaced signals toward breast cancer progression. Cancers. 2021;13:645.PubMedPubMedCentral
9.
Zurück zum Zitat Golubovskaya VM. Targeting FAK in human cancer: from finding to first clinical trials. Front Biosci. 2014;19:687–706. Golubovskaya VM. Targeting FAK in human cancer: from finding to first clinical trials. Front Biosci. 2014;19:687–706.
10.
Zurück zum Zitat Tremblay L, Hauck W, Aprikian AG, Begin LR, Chapdelaine A, Chevalier S. Focal adhesion kinase (pp125FAK) expression, activation and association with paxillin and p50CSK in human metastatic prostate carcinoma. Int J Cancer. 1996;68:164–71.PubMed Tremblay L, Hauck W, Aprikian AG, Begin LR, Chapdelaine A, Chevalier S. Focal adhesion kinase (pp125FAK) expression, activation and association with paxillin and p50CSK in human metastatic prostate carcinoma. Int J Cancer. 1996;68:164–71.PubMed
11.
Zurück zum Zitat Ossovskaya V, Lim ST, Ota N, Schlaepfer DD, Ilic D. FAK nuclear export signal sequences. FEBS Lett. 2008;582:2402–6.PubMedPubMedCentral Ossovskaya V, Lim ST, Ota N, Schlaepfer DD, Ilic D. FAK nuclear export signal sequences. FEBS Lett. 2008;582:2402–6.PubMedPubMedCentral
12.
Zurück zum Zitat Zhou J, Yi Q, Tang L. The roles of nuclear focal adhesion kinase (FAK) on cancer: a focused review. J Exp Clin Cancer Res. 2019;38:250.PubMedPubMedCentral Zhou J, Yi Q, Tang L. The roles of nuclear focal adhesion kinase (FAK) on cancer: a focused review. J Exp Clin Cancer Res. 2019;38:250.PubMedPubMedCentral
13.
Zurück zum Zitat Lim ST, Chen XL, Lim Y, Hanson DA, Vo TT, Howerton K, et al. Nuclear FAK promotes cell proliferation and survival through FERM-enhanced p53 degradation. Mol Cell. 2008;29:9–22.PubMedPubMedCentral Lim ST, Chen XL, Lim Y, Hanson DA, Vo TT, Howerton K, et al. Nuclear FAK promotes cell proliferation and survival through FERM-enhanced p53 degradation. Mol Cell. 2008;29:9–22.PubMedPubMedCentral
14.
Zurück zum Zitat Constanzo JD, Tang K-J, Rindhe S, Melegari M, Liu H, Tang X, et al. PIAS1-FAK interaction promotes the survival and progression of non-small cell lung cancer. Neoplasia. 2016;18:282–93.PubMedPubMedCentral Constanzo JD, Tang K-J, Rindhe S, Melegari M, Liu H, Tang X, et al. PIAS1-FAK interaction promotes the survival and progression of non-small cell lung cancer. Neoplasia. 2016;18:282–93.PubMedPubMedCentral
15.
Zurück zum Zitat Tai H-C, Chang A-C, Yu H-J, Huang C-Y, Tsai Y-C, Lai Y-W, et al. Osteoblast-derived WNT-induced secreted protein 1 increases VCAM-1 expression and enhances prostate cancer metastasis by down-regulating miR-126. Oncotarget. 2014;5:7589–98.PubMedPubMedCentral Tai H-C, Chang A-C, Yu H-J, Huang C-Y, Tsai Y-C, Lai Y-W, et al. Osteoblast-derived WNT-induced secreted protein 1 increases VCAM-1 expression and enhances prostate cancer metastasis by down-regulating miR-126. Oncotarget. 2014;5:7589–98.PubMedPubMedCentral
16.
Zurück zum Zitat Serrels B, McGivern N, Canel M, Byron A, Johnson SC, McSorley HJ, et al. IL-33 and ST2 mediate FAK-dependent antitumor immune evasion through transcriptional networks. Sci Signal. 2017;10: eaan8355.PubMedPubMedCentral Serrels B, McGivern N, Canel M, Byron A, Johnson SC, McSorley HJ, et al. IL-33 and ST2 mediate FAK-dependent antitumor immune evasion through transcriptional networks. Sci Signal. 2017;10: eaan8355.PubMedPubMedCentral
17.
Zurück zum Zitat Lim S-T, Miller NLG, Chen XL, Tancioni I, Walsh CT, Lawson C, et al. Nuclear-localized focal adhesion kinase regulates inflammatory VCAM-1 expression. J Cell Biol. 2012;197:907–19.PubMedPubMedCentral Lim S-T, Miller NLG, Chen XL, Tancioni I, Walsh CT, Lawson C, et al. Nuclear-localized focal adhesion kinase regulates inflammatory VCAM-1 expression. J Cell Biol. 2012;197:907–19.PubMedPubMedCentral
18.
Zurück zum Zitat Frame MC, Patel H, Serrels B, Lietha D, Eck MJ. The FERM domain: organizing the structure and function of FAK. Nat Rev Mol Cell Biol. 2010;11:802–14.PubMed Frame MC, Patel H, Serrels B, Lietha D, Eck MJ. The FERM domain: organizing the structure and function of FAK. Nat Rev Mol Cell Biol. 2010;11:802–14.PubMed
19.
Zurück zum Zitat Cooper J, Giancotti FG. Integrin signaling in cancer: mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell. 2019;35:347–67.PubMedPubMedCentral Cooper J, Giancotti FG. Integrin signaling in cancer: mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell. 2019;35:347–67.PubMedPubMedCentral
20.
Zurück zum Zitat Sun X, Meng L, Qiao W, Yang R, Gao Q, Peng Y, et al. Vascular endothelial growth factor A/Vascular endothelial growth factor receptor 2 axis promotes human dental pulp stem cell migration via the FAK/PI3K/Akt and p38 MAPK signalling pathways. Int Endod J. 2019;52:1691–703.PubMed Sun X, Meng L, Qiao W, Yang R, Gao Q, Peng Y, et al. Vascular endothelial growth factor A/Vascular endothelial growth factor receptor 2 axis promotes human dental pulp stem cell migration via the FAK/PI3K/Akt and p38 MAPK signalling pathways. Int Endod J. 2019;52:1691–703.PubMed
21.
Zurück zum Zitat Shen T, Guo Q. EGFR signaling pathway occupies an important position in cancer-related downstream signaling pathways of Pyk2. Cell Biol Int. 2019;44:2–13.PubMedPubMedCentral Shen T, Guo Q. EGFR signaling pathway occupies an important position in cancer-related downstream signaling pathways of Pyk2. Cell Biol Int. 2019;44:2–13.PubMedPubMedCentral
22.
Zurück zum Zitat Rigiracciolo DC, Santolla MF, Lappano R, Vivacqua A, Cirillo F, Galli GR, et al. Focal adhesion kinase (FAK) activation by estrogens involves GPER in triple-negative breast cancer cells. J Exp Clin Cancer Res. 2019;38:58.PubMedPubMedCentral Rigiracciolo DC, Santolla MF, Lappano R, Vivacqua A, Cirillo F, Galli GR, et al. Focal adhesion kinase (FAK) activation by estrogens involves GPER in triple-negative breast cancer cells. J Exp Clin Cancer Res. 2019;38:58.PubMedPubMedCentral
23.
Zurück zum Zitat Goñi GM, Epifano C, Boskovic J, Camacho-Artacho M, Zhou J, Bronowska A, et al. Phosphatidylinositol 4,5-bisphosphate triggers activation of focal adhesion kinase by inducing clustering and conformational changes. Proc Natl Acad Sci USA. 2014;111:E3177–86.PubMedPubMedCentral Goñi GM, Epifano C, Boskovic J, Camacho-Artacho M, Zhou J, Bronowska A, et al. Phosphatidylinositol 4,5-bisphosphate triggers activation of focal adhesion kinase by inducing clustering and conformational changes. Proc Natl Acad Sci USA. 2014;111:E3177–86.PubMedPubMedCentral
24.
Zurück zum Zitat Lim Y, Lim ST, Tomar A, Gardel M, Bernard-Trifilo JA, Chen XL, et al. PyK2 and FAK connections to p190Rho guanine nucleotide exchange factor regulate RhoA activity, focal adhesion formation, and cell motility. J Cell Biol. 2008;180:187–203.PubMedPubMedCentral Lim Y, Lim ST, Tomar A, Gardel M, Bernard-Trifilo JA, Chen XL, et al. PyK2 and FAK connections to p190Rho guanine nucleotide exchange factor regulate RhoA activity, focal adhesion formation, and cell motility. J Cell Biol. 2008;180:187–203.PubMedPubMedCentral
25.
Zurück zum Zitat Mousson A, Sick E, Carl P, Dujardin D, De Mey J, Rondé P. Targeting focal adhesion kinase using inhibitors of protein–protein interactions. Cancers. 2018;10:278.PubMedPubMedCentral Mousson A, Sick E, Carl P, Dujardin D, De Mey J, Rondé P. Targeting focal adhesion kinase using inhibitors of protein–protein interactions. Cancers. 2018;10:278.PubMedPubMedCentral
26.
Zurück zum Zitat Rashid M, Belmont J, Carpenter D, Turner CE, Olson EC. Neural-specific deletion of the focal adhesion adaptor protein paxillin slows migration speed and delays cortical layer formation. Development. 2017;144:4002–14.PubMedPubMedCentral Rashid M, Belmont J, Carpenter D, Turner CE, Olson EC. Neural-specific deletion of the focal adhesion adaptor protein paxillin slows migration speed and delays cortical layer formation. Development. 2017;144:4002–14.PubMedPubMedCentral
27.
Zurück zum Zitat Baumann K. Cell adhesion: FAK or talin: who goes first? Nat Rev Mol Cell Biol. 2012;13:138.PubMed Baumann K. Cell adhesion: FAK or talin: who goes first? Nat Rev Mol Cell Biol. 2012;13:138.PubMed
28.
Zurück zum Zitat Brami-Cherrier K, Gervasi N, Arsenieva D, Walkiewicz K, Boutterin MC, Ortega A, et al. FAK dimerization controls its kinase-dependent functions at focal adhesions. Embo J. 2014;33:356–70.PubMedPubMedCentral Brami-Cherrier K, Gervasi N, Arsenieva D, Walkiewicz K, Boutterin MC, Ortega A, et al. FAK dimerization controls its kinase-dependent functions at focal adhesions. Embo J. 2014;33:356–70.PubMedPubMedCentral
29.
Zurück zum Zitat Liu Y, Loijens JC, Martin KH, Karginov AV, Parsons JT. The association of ASAP1, an ADP ribosylation factor-GTPase activating protein, with focal adhesion kinase contributes to the process of focal adhesion assembly. Mol Biol Cell. 2002;13:2147–56.PubMedPubMedCentral Liu Y, Loijens JC, Martin KH, Karginov AV, Parsons JT. The association of ASAP1, an ADP ribosylation factor-GTPase activating protein, with focal adhesion kinase contributes to the process of focal adhesion assembly. Mol Biol Cell. 2002;13:2147–56.PubMedPubMedCentral
30.
Zurück zum Zitat Schaller MD, Hildebrand JD, Shannon JD, Fox JW, Vines RR, Parsons JT. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent binding of pp60src. Mol Cell Biol. 1994;14:1680–8.PubMedPubMedCentral Schaller MD, Hildebrand JD, Shannon JD, Fox JW, Vines RR, Parsons JT. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2-dependent binding of pp60src. Mol Cell Biol. 1994;14:1680–8.PubMedPubMedCentral
31.
Zurück zum Zitat Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci. 2003;116:1409–16.PubMed Parsons JT. Focal adhesion kinase: the first ten years. J Cell Sci. 2003;116:1409–16.PubMed
32.
Zurück zum Zitat Calalb MB, Polte TR, Hanks SK. Tyrosine phosphorylation of focal adhesion kinase at sites in the catalytic domain regulates kinase activity: a role for Src family kinases. Mol Cell Biol. 1995;15:954–63.PubMedPubMedCentral Calalb MB, Polte TR, Hanks SK. Tyrosine phosphorylation of focal adhesion kinase at sites in the catalytic domain regulates kinase activity: a role for Src family kinases. Mol Cell Biol. 1995;15:954–63.PubMedPubMedCentral
33.
Zurück zum Zitat Lim Y, Park H, Jeon J, Han I, Kim J, Jho EH, et al. Focal adhesion kinase is negatively regulated by phosphorylation at tyrosine 407. J Biol Chem. 2007;282:10398–404.PubMed Lim Y, Park H, Jeon J, Han I, Kim J, Jho EH, et al. Focal adhesion kinase is negatively regulated by phosphorylation at tyrosine 407. J Biol Chem. 2007;282:10398–404.PubMed
34.
Zurück zum Zitat Schlaepfer DD, Hanks SK, Hunter T, van der Geer P. Integrin-mediated signal transduction linked to Ras pathway by GRB2 binding to focal adhesion kinase. Nature. 1994;372:786–91.PubMed Schlaepfer DD, Hanks SK, Hunter T, van der Geer P. Integrin-mediated signal transduction linked to Ras pathway by GRB2 binding to focal adhesion kinase. Nature. 1994;372:786–91.PubMed
35.
Zurück zum Zitat Mitra SK, Mikolon D, Molina JE, Hsia DA, Hanson DA, Chi A, et al. Intrinsic FAK activity and Y925 phosphorylation facilitate an angiogenic switch in tumors. Oncogene. 2006;25:5969–84.PubMed Mitra SK, Mikolon D, Molina JE, Hsia DA, Hanson DA, Chi A, et al. Intrinsic FAK activity and Y925 phosphorylation facilitate an angiogenic switch in tumors. Oncogene. 2006;25:5969–84.PubMed
36.
Zurück zum Zitat Mitra SK, Hanson DA, Schlaepfer DD. Focal adhesion kinase: in command and control of cell motility. Nat Rev Mol Cell Biol. 2005;6:56–68.PubMed Mitra SK, Hanson DA, Schlaepfer DD. Focal adhesion kinase: in command and control of cell motility. Nat Rev Mol Cell Biol. 2005;6:56–68.PubMed
37.
Zurück zum Zitat Cai X, Lietha D, Ceccarelli DF, Karginov AV, Rajfur Z, Jacobson K, et al. Spatial and temporal regulation of focal adhesion kinase activity in living cells. Mol Cell Biol. 2008;28:201–14.PubMed Cai X, Lietha D, Ceccarelli DF, Karginov AV, Rajfur Z, Jacobson K, et al. Spatial and temporal regulation of focal adhesion kinase activity in living cells. Mol Cell Biol. 2008;28:201–14.PubMed
38.
Zurück zum Zitat Zhang N, Zhu H-P, Huang W, Wen X, Xie X, Jiang X, et al. Unraveling the structures, functions and mechanisms of epithelial membrane protein family in human cancers. Exp Hematol Oncol. 2022;11:69.PubMedPubMedCentral Zhang N, Zhu H-P, Huang W, Wen X, Xie X, Jiang X, et al. Unraveling the structures, functions and mechanisms of epithelial membrane protein family in human cancers. Exp Hematol Oncol. 2022;11:69.PubMedPubMedCentral
39.
Zurück zum Zitat Jung O, Choi S, Jang SB, Lee SA, Lim ST, Choi YJ, et al. Tetraspan TM4SF5-dependent direct activation of FAK and metastatic potential of hepatocarcinoma cells. J Cell Sci. 2012;125:5960–73.PubMedPubMedCentral Jung O, Choi S, Jang SB, Lee SA, Lim ST, Choi YJ, et al. Tetraspan TM4SF5-dependent direct activation of FAK and metastatic potential of hepatocarcinoma cells. J Cell Sci. 2012;125:5960–73.PubMedPubMedCentral
40.
Zurück zum Zitat Lee E, Choi A, Jun Y, Kim N, Yook JI, Kim SY, et al. Glutathione peroxidase-1 regulates adhesion and metastasis of triple-negative breast cancer cells via FAK signaling. Redox Biol. 2020;29: 101391.PubMed Lee E, Choi A, Jun Y, Kim N, Yook JI, Kim SY, et al. Glutathione peroxidase-1 regulates adhesion and metastasis of triple-negative breast cancer cells via FAK signaling. Redox Biol. 2020;29: 101391.PubMed
41.
Zurück zum Zitat Sieg DJ, Hauck CR, Ilic D, Klingbeil CK, Schaefer E, Damsky CH, et al. FAK integrates growth-factor and integrin signals to promote cell migration. Nat Cell Biol. 2000;2:249–56.PubMed Sieg DJ, Hauck CR, Ilic D, Klingbeil CK, Schaefer E, Damsky CH, et al. FAK integrates growth-factor and integrin signals to promote cell migration. Nat Cell Biol. 2000;2:249–56.PubMed
42.
Zurück zum Zitat Chen TH, Chan PC, Chen CL, Chen HC. Phosphorylation of focal adhesion kinase on tyrosine 194 by Met leads to its activation through relief of autoinhibition. Oncogene. 2011;30:153–66.PubMed Chen TH, Chan PC, Chen CL, Chen HC. Phosphorylation of focal adhesion kinase on tyrosine 194 by Met leads to its activation through relief of autoinhibition. Oncogene. 2011;30:153–66.PubMed
43.
Zurück zum Zitat Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139:891–906.PubMedPubMedCentral Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139:891–906.PubMedPubMedCentral
44.
Zurück zum Zitat Choi CH, Webb BA, Chimenti MS, Jacobson MP, Barber DL. pH sensing by FAK-His58 regulates focal adhesion remodeling. J Cell Biol. 2013;202:849–59.PubMedPubMedCentral Choi CH, Webb BA, Chimenti MS, Jacobson MP, Barber DL. pH sensing by FAK-His58 regulates focal adhesion remodeling. J Cell Biol. 2013;202:849–59.PubMedPubMedCentral
45.
Zurück zum Zitat Xing P, Wang Y, Zhang L, Ma C, Lu J. Knockdown of lncRNA MIR44352HG and ST8SIA1 expression inhibits the proliferation, invasion and migration of prostate cancer cells in vitro and in vivo by blocking the activation of the FAK/AKT/betacatenin signaling pathway. Int J Mol Med. 2021;47(6):93.PubMedPubMedCentral Xing P, Wang Y, Zhang L, Ma C, Lu J. Knockdown of lncRNA MIR44352HG and ST8SIA1 expression inhibits the proliferation, invasion and migration of prostate cancer cells in vitro and in vivo by blocking the activation of the FAK/AKT/betacatenin signaling pathway. Int J Mol Med. 2021;47(6):93.PubMedPubMedCentral
46.
Zurück zum Zitat Gu W, Zhang L, Zhang X, Wang B, Shi X, Hu K, et al. MiR-15p-5p mediates the coordination of ICAM-1 and FAK to promote endothelial cell proliferation and migration. Inflammation. 2022;45:1402–17.PubMed Gu W, Zhang L, Zhang X, Wang B, Shi X, Hu K, et al. MiR-15p-5p mediates the coordination of ICAM-1 and FAK to promote endothelial cell proliferation and migration. Inflammation. 2022;45:1402–17.PubMed
47.
Zurück zum Zitat Nunez RE, Del Valle MM, Ortiz K, Almodovar L, Kucheryavykh L. Microglial cytokines induce invasiveness and proliferation of human glioblastoma through Pyk2 and FAK activation. Cancers. 2021;13(24):6160.PubMedPubMedCentral Nunez RE, Del Valle MM, Ortiz K, Almodovar L, Kucheryavykh L. Microglial cytokines induce invasiveness and proliferation of human glioblastoma through Pyk2 and FAK activation. Cancers. 2021;13(24):6160.PubMedPubMedCentral
48.
Zurück zum Zitat Janku F, Yap TA, Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol. 2018;15:273–91.PubMed Janku F, Yap TA, Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol. 2018;15:273–91.PubMed
49.
Zurück zum Zitat Luo X, Cao M, Gao F, He X. YTHDF1 promotes hepatocellular carcinoma progression via activating PI3K/AKT/mTOR signaling pathway and inducing epithelial-mesenchymal transition. Exp Hematol Oncol. 2021;10:35.PubMedPubMedCentral Luo X, Cao M, Gao F, He X. YTHDF1 promotes hepatocellular carcinoma progression via activating PI3K/AKT/mTOR signaling pathway and inducing epithelial-mesenchymal transition. Exp Hematol Oncol. 2021;10:35.PubMedPubMedCentral
50.
Zurück zum Zitat Paul R, Luo M, Mo X, Lu J, Yeo SK, Guan J-L. FAK activates AKT-mTOR signaling to promote the growth and progression of MMTV-Wnt1-driven basal-like mammary tumors. Breast Cancer Res. 2020;22:59.PubMedPubMedCentral Paul R, Luo M, Mo X, Lu J, Yeo SK, Guan J-L. FAK activates AKT-mTOR signaling to promote the growth and progression of MMTV-Wnt1-driven basal-like mammary tumors. Breast Cancer Res. 2020;22:59.PubMedPubMedCentral
51.
Zurück zum Zitat Rigiracciolo DC, Nohata N, Lappano R, Cirillo F, Talia M, Scordamaglia D, et al. IGF-1/IGF-1R/FAK/YAP transduction signaling prompts growth effects in triple-negative breast cancer (TNBC) cells. Cells. 2020;9(4):1010.PubMedPubMedCentral Rigiracciolo DC, Nohata N, Lappano R, Cirillo F, Talia M, Scordamaglia D, et al. IGF-1/IGF-1R/FAK/YAP transduction signaling prompts growth effects in triple-negative breast cancer (TNBC) cells. Cells. 2020;9(4):1010.PubMedPubMedCentral
52.
Zurück zum Zitat Song X, Xu H, Wang P, Wang J, Affo S, Wang H, et al. Focal adhesion kinase (FAK) promotes cholangiocarcinoma development and progression via YAP activation. J Hepatol. 2021;75:888–99.PubMedPubMedCentral Song X, Xu H, Wang P, Wang J, Affo S, Wang H, et al. Focal adhesion kinase (FAK) promotes cholangiocarcinoma development and progression via YAP activation. J Hepatol. 2021;75:888–99.PubMedPubMedCentral
53.
Zurück zum Zitat Zhao JH, Reiske H, Guan JL. Regulation of the cell cycle by focal adhesion kinase. J Cell Biol. 1998;143:1997–2008.PubMedPubMedCentral Zhao JH, Reiske H, Guan JL. Regulation of the cell cycle by focal adhesion kinase. J Cell Biol. 1998;143:1997–2008.PubMedPubMedCentral
54.
Zurück zum Zitat Conley SJ, Bosco EE, Tice DA, Hollingsworth RE, Herbst R, Xiao Z. HER2 drives Mucin-like 1 to control proliferation in breast cancer cells. Oncogene. 2016;35:4225–34.PubMedPubMedCentral Conley SJ, Bosco EE, Tice DA, Hollingsworth RE, Herbst R, Xiao Z. HER2 drives Mucin-like 1 to control proliferation in breast cancer cells. Oncogene. 2016;35:4225–34.PubMedPubMedCentral
55.
Zurück zum Zitat Zhao J, Pestell R, Guan JL. Transcriptional activation of cyclin D1 promoter by FAK contributes to cell cycle progression. Mol Biol Cell. 2001;12:4066–77.PubMedPubMedCentral Zhao J, Pestell R, Guan JL. Transcriptional activation of cyclin D1 promoter by FAK contributes to cell cycle progression. Mol Biol Cell. 2001;12:4066–77.PubMedPubMedCentral
56.
Zurück zum Zitat Alza L, Nàger M, Visa A, Cantí C, Herreros J. FAK inhibition induces glioblastoma cell senescence-like state through p62 and p27. Cancers. 2020;12:1086.PubMedPubMedCentral Alza L, Nàger M, Visa A, Cantí C, Herreros J. FAK inhibition induces glioblastoma cell senescence-like state through p62 and p27. Cancers. 2020;12:1086.PubMedPubMedCentral
57.
Zurück zum Zitat Jeong K, Murphy JM, Ahn EE, Lim SS. FAK in the nucleus prevents VSMC proliferation by promoting p27 and p21 expression via Skp2 degradation. Cardiovasc Res. 2022;118:1150–63.PubMed Jeong K, Murphy JM, Ahn EE, Lim SS. FAK in the nucleus prevents VSMC proliferation by promoting p27 and p21 expression via Skp2 degradation. Cardiovasc Res. 2022;118:1150–63.PubMed
58.
Zurück zum Zitat Canel M, Byron A, Sims AH, Cartier J, Patel H, Frame MC, et al. Nuclear FAK and Runx1 cooperate to regulate IGFBP3, cell-cycle progression, and tumor growth. Cancer Res. 2017;77:5301–12.PubMedPubMedCentral Canel M, Byron A, Sims AH, Cartier J, Patel H, Frame MC, et al. Nuclear FAK and Runx1 cooperate to regulate IGFBP3, cell-cycle progression, and tumor growth. Cancer Res. 2017;77:5301–12.PubMedPubMedCentral
59.
Zurück zum Zitat Sharma BK, Mureb D, Murab S, Rosenfeldt L, Francisco B, Cantrell R, et al. Fibrinogen activates focal adhesion kinase (FAK) promoting colorectal adenocarcinoma growth. J Thromb Haemost. 2021;19:2480–94.PubMedPubMedCentral Sharma BK, Mureb D, Murab S, Rosenfeldt L, Francisco B, Cantrell R, et al. Fibrinogen activates focal adhesion kinase (FAK) promoting colorectal adenocarcinoma growth. J Thromb Haemost. 2021;19:2480–94.PubMedPubMedCentral
60.
Zurück zum Zitat Kim EY, Cha YJ, Jeong S, Chang YS. Overexpression of CEACAM6 activates Src-FAK signaling and inhibits anoikis, through homophilic interactions in lung adenocarcinomas. Transl Oncol. 2022;20:101402.PubMedPubMedCentral Kim EY, Cha YJ, Jeong S, Chang YS. Overexpression of CEACAM6 activates Src-FAK signaling and inhibits anoikis, through homophilic interactions in lung adenocarcinomas. Transl Oncol. 2022;20:101402.PubMedPubMedCentral
61.
Zurück zum Zitat Kurenova E, Xu LH, Yang X, Baldwin AS, Craven RJ, Hanks SK, et al. Focal adhesion kinase suppresses apoptosis by binding to the death domain of receptor-interacting protein. Mol Cell Biol. 2004;24:4361–71.PubMedPubMedCentral Kurenova E, Xu LH, Yang X, Baldwin AS, Craven RJ, Hanks SK, et al. Focal adhesion kinase suppresses apoptosis by binding to the death domain of receptor-interacting protein. Mol Cell Biol. 2004;24:4361–71.PubMedPubMedCentral
62.
Zurück zum Zitat Huang C, Wang Y, Huang J-H, Liu W. Sema3A drastically suppresses tumor growth in oral cancer Xenograft model of mice. BMC Pharmacol Toxicol. 2017;18:55.PubMedPubMedCentral Huang C, Wang Y, Huang J-H, Liu W. Sema3A drastically suppresses tumor growth in oral cancer Xenograft model of mice. BMC Pharmacol Toxicol. 2017;18:55.PubMedPubMedCentral
63.
Zurück zum Zitat Liao M, Qin R, Huang W, Zhu H-P, Peng F, Han B, et al. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: a revisited perspective from molecular mechanisms to targeted therapies. J Hematol Oncol. 2022;15:44.PubMedPubMedCentral Liao M, Qin R, Huang W, Zhu H-P, Peng F, Han B, et al. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: a revisited perspective from molecular mechanisms to targeted therapies. J Hematol Oncol. 2022;15:44.PubMedPubMedCentral
64.
Zurück zum Zitat Frisch SM, Vuori K, Ruoslahti E, Chan-Hui PY. Control of adhesion-dependent cell survival by focal adhesion kinase. J Cell Biol. 1996;134:793–9.PubMed Frisch SM, Vuori K, Ruoslahti E, Chan-Hui PY. Control of adhesion-dependent cell survival by focal adhesion kinase. J Cell Biol. 1996;134:793–9.PubMed
65.
Zurück zum Zitat da Silva SD, Xu B, Maschietto M, Marchi FA, Alkailani MI, Bijian K, et al. TRAF2 Cooperates with Focal Adhesion Signaling to Regulate Cancer Cell Susceptibility to Anoikis. Mol Cancer Ther. 2019;18:139–46.PubMed da Silva SD, Xu B, Maschietto M, Marchi FA, Alkailani MI, Bijian K, et al. TRAF2 Cooperates with Focal Adhesion Signaling to Regulate Cancer Cell Susceptibility to Anoikis. Mol Cancer Ther. 2019;18:139–46.PubMed
66.
Zurück zum Zitat Alanko J, Mai A, Jacquemet G, Schauer K, Kaukonen R, Saari M, et al. Integrin endosomal signalling suppresses anoikis. Nat Cell Biol. 2015;17:1412–21.PubMedPubMedCentral Alanko J, Mai A, Jacquemet G, Schauer K, Kaukonen R, Saari M, et al. Integrin endosomal signalling suppresses anoikis. Nat Cell Biol. 2015;17:1412–21.PubMedPubMedCentral
67.
Zurück zum Zitat Alanko J, Ivaska J. Endosomes: Emerging Platforms for Integrin-Mediated FAK Signalling. Trends Cell Biol. 2016;26:391–8.PubMed Alanko J, Ivaska J. Endosomes: Emerging Platforms for Integrin-Mediated FAK Signalling. Trends Cell Biol. 2016;26:391–8.PubMed
68.
Zurück zum Zitat Zhao M, Finlay D, Kwong E, Liddington R, Viollet B, Sasaoka N, et al. Cell adhesion suppresses autophagy via Src/FAK-mediated phosphorylation and inhibition of AMPK. Cell Signal. 2022;89:110170.PubMed Zhao M, Finlay D, Kwong E, Liddington R, Viollet B, Sasaoka N, et al. Cell adhesion suppresses autophagy via Src/FAK-mediated phosphorylation and inhibition of AMPK. Cell Signal. 2022;89:110170.PubMed
69.
Zurück zum Zitat Lu D, Wang Y, Zhang T, Wang F, Li K, Zhou S, et al. Metabolic radiolabeling and in vivo PET imaging of cytotoxic T lymphocytes to guide combination adoptive cell transfer cancer therapy. J Nanobiotechnol. 2021;19:175. Lu D, Wang Y, Zhang T, Wang F, Li K, Zhou S, et al. Metabolic radiolabeling and in vivo PET imaging of cytotoxic T lymphocytes to guide combination adoptive cell transfer cancer therapy. J Nanobiotechnol. 2021;19:175.
70.
Zurück zum Zitat Xu LH, Yang X, Bradham CA, Brenner DA, Baldwin AS, Craven RJ, et al. The focal adhesion kinase suppresses transformation-associated, anchorage-independent apoptosis in human breast cancer cells. Involvement of death receptor-related signaling pathways. J Biol Chem. 2000;275:30597–604.PubMed Xu LH, Yang X, Bradham CA, Brenner DA, Baldwin AS, Craven RJ, et al. The focal adhesion kinase suppresses transformation-associated, anchorage-independent apoptosis in human breast cancer cells. Involvement of death receptor-related signaling pathways. J Biol Chem. 2000;275:30597–604.PubMed
71.
Zurück zum Zitat Sonoda Y, Matsumoto Y, Funakoshi M, Yamamoto D, Hanks SK, Kasahara T. Anti-apoptotic role of focal adhesion kinase (FAK). Induction of inhibitor-of-apoptosis proteins and apoptosis suppression by the overexpression of FAK in a human leukemic cell line, HL-60. J Biol Chem. 2000;275:16309–15.PubMed Sonoda Y, Matsumoto Y, Funakoshi M, Yamamoto D, Hanks SK, Kasahara T. Anti-apoptotic role of focal adhesion kinase (FAK). Induction of inhibitor-of-apoptosis proteins and apoptosis suppression by the overexpression of FAK in a human leukemic cell line, HL-60. J Biol Chem. 2000;275:16309–15.PubMed
72.
Zurück zum Zitat Golubovskaya VM, Finch R, Cance WG. Direct interaction of the N-terminal domain of focal adhesion kinase with the N-terminal transactivation domain of p53. J Biol Chem. 2005;280:25008–21.PubMed Golubovskaya VM, Finch R, Cance WG. Direct interaction of the N-terminal domain of focal adhesion kinase with the N-terminal transactivation domain of p53. J Biol Chem. 2005;280:25008–21.PubMed
73.
Zurück zum Zitat Shi Q, Boettiger D. A novel mode for integrin-mediated signaling: tethering is required for phosphorylation of FAK Y397. Mol Biol Cell. 2003;14:4306–15.PubMedPubMedCentral Shi Q, Boettiger D. A novel mode for integrin-mediated signaling: tethering is required for phosphorylation of FAK Y397. Mol Biol Cell. 2003;14:4306–15.PubMedPubMedCentral
74.
Zurück zum Zitat Vicente-Manzanares M, Webb DJ, Horwitz AR. Cell migration at a glance. J Cell Sci. 2005;118:4917–9.PubMed Vicente-Manzanares M, Webb DJ, Horwitz AR. Cell migration at a glance. J Cell Sci. 2005;118:4917–9.PubMed
75.
Zurück zum Zitat Cary LA, Han DC, Polte TR, Hanks SK, Guan JL. Identification of p130Cas as a mediator of focal adhesion kinase-promoted cell migration. J Cell Biol. 1998;140:211–21.PubMedPubMedCentral Cary LA, Han DC, Polte TR, Hanks SK, Guan JL. Identification of p130Cas as a mediator of focal adhesion kinase-promoted cell migration. J Cell Biol. 1998;140:211–21.PubMedPubMedCentral
76.
Zurück zum Zitat Webb DJ, Donais K, Whitmore LA, Thomas SM, Turner CE, Parsons JT, et al. FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. Nat Cell Biol. 2004;6:154–61.PubMed Webb DJ, Donais K, Whitmore LA, Thomas SM, Turner CE, Parsons JT, et al. FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly. Nat Cell Biol. 2004;6:154–61.PubMed
77.
Zurück zum Zitat Huang C, Rajfur Z, Borchers C, Schaller MD, Jacobson K. JNK phosphorylates paxillin and regulates cell migration. Nature. 2003;424:219–23.PubMed Huang C, Rajfur Z, Borchers C, Schaller MD, Jacobson K. JNK phosphorylates paxillin and regulates cell migration. Nature. 2003;424:219–23.PubMed
78.
Zurück zum Zitat Chu P-Y, Huang L-Y, Hsu C-H, Liang C-C, Guan J-L, Hung T-H, et al. Tyrosine phosphorylation of growth factor receptor-bound protein-7 by focal adhesion kinase in the regulation of cell migration, proliferation, and tumorigenesis. J Biol Chem. 2009;284:20215–26.PubMedPubMedCentral Chu P-Y, Huang L-Y, Hsu C-H, Liang C-C, Guan J-L, Hung T-H, et al. Tyrosine phosphorylation of growth factor receptor-bound protein-7 by focal adhesion kinase in the regulation of cell migration, proliferation, and tumorigenesis. J Biol Chem. 2009;284:20215–26.PubMedPubMedCentral
79.
Zurück zum Zitat Shen T-L, Han DC, Guan J-L. Association of Grb7 with phosphoinositides and its role in the regulation of cell migration. J Biol Chem. 2002;277:29069–77.PubMed Shen T-L, Han DC, Guan J-L. Association of Grb7 with phosphoinositides and its role in the regulation of cell migration. J Biol Chem. 2002;277:29069–77.PubMed
80.
Zurück zum Zitat Carragher NO, Westhoff MA, Fincham VJ, Schaller MD, Frame MC. A novel role for FAK as a protease-targeting adaptor protein: regulation by p42 ERK and Src. Curr Biol. 2003;13:1442–50.PubMed Carragher NO, Westhoff MA, Fincham VJ, Schaller MD, Frame MC. A novel role for FAK as a protease-targeting adaptor protein: regulation by p42 ERK and Src. Curr Biol. 2003;13:1442–50.PubMed
81.
Zurück zum Zitat Kerstein PC, Patel KM, Gomez TM. Calpain-mediated proteolysis of Talin and FAK regulates adhesion dynamics necessary for axon guidance. J Neurosci. 2017;37:1568–80.PubMedPubMedCentral Kerstein PC, Patel KM, Gomez TM. Calpain-mediated proteolysis of Talin and FAK regulates adhesion dynamics necessary for axon guidance. J Neurosci. 2017;37:1568–80.PubMedPubMedCentral
82.
Zurück zum Zitat Chan KT, Bennin DA, Huttenlocher A. Regulation of adhesion dynamics by calpain-mediated proteolysis of focal adhesion kinase (FAK). J Biol Chem. 2010;285:11418–26.PubMedPubMedCentral Chan KT, Bennin DA, Huttenlocher A. Regulation of adhesion dynamics by calpain-mediated proteolysis of focal adhesion kinase (FAK). J Biol Chem. 2010;285:11418–26.PubMedPubMedCentral
83.
Zurück zum Zitat McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer—a new therapeutic opportunity. Nat Rev Cancer. 2005;5:505–15.PubMed McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer—a new therapeutic opportunity. Nat Rev Cancer. 2005;5:505–15.PubMed
84.
Zurück zum Zitat Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28:15–33.PubMed Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28:15–33.PubMed
85.
Zurück zum Zitat Wang T, Rao D, Yu C, Sheng J, Luo Y, Xia L, et al. RHO GTPase family in hepatocellular carcinoma. Exp Hematol Oncol. 2022;11:91.PubMedPubMedCentral Wang T, Rao D, Yu C, Sheng J, Luo Y, Xia L, et al. RHO GTPase family in hepatocellular carcinoma. Exp Hematol Oncol. 2022;11:91.PubMedPubMedCentral
86.
Zurück zum Zitat Krugmann S, Jordens I, Gevaert K, Driessens M, Vandekerckhove J, Hall A. Cdc42 induces filopodia by promoting the formation of an IRSp53: Mena complex. Curr Biol. 2001;11:1645–55.PubMed Krugmann S, Jordens I, Gevaert K, Driessens M, Vandekerckhove J, Hall A. Cdc42 induces filopodia by promoting the formation of an IRSp53: Mena complex. Curr Biol. 2001;11:1645–55.PubMed
87.
Zurück zum Zitat Ridley AJ. Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol. 2006;16:522–9.PubMed Ridley AJ. Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol. 2006;16:522–9.PubMed
88.
Zurück zum Zitat Huang Y, Hong W, Wei X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 2022;15:129.PubMedPubMedCentral Huang Y, Hong W, Wei X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 2022;15:129.PubMedPubMedCentral
89.
Zurück zum Zitat Dong B, Li S, Zhu S, Yi M, Luo S, Wu K. MiRNA-mediated EMT and CSCs in cancer chemoresistance. Exp Hematol Oncol. 2021;10:12.PubMedPubMedCentral Dong B, Li S, Zhu S, Yi M, Luo S, Wu K. MiRNA-mediated EMT and CSCs in cancer chemoresistance. Exp Hematol Oncol. 2021;10:12.PubMedPubMedCentral
90.
Zurück zum Zitat Canel M, Serrels A, Frame MC, Brunton VG. E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci. 2013;126:393–401.PubMed Canel M, Serrels A, Frame MC, Brunton VG. E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci. 2013;126:393–401.PubMed
91.
Zurück zum Zitat Avizienyte E, Wyke AW, Jones RJ, McLean GW, Westhoff MA, Brunton VG, et al. Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nat Cell Biol. 2002;4:632–8.PubMed Avizienyte E, Wyke AW, Jones RJ, McLean GW, Westhoff MA, Brunton VG, et al. Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling. Nat Cell Biol. 2002;4:632–8.PubMed
92.
Zurück zum Zitat Canel M, Serrels A, Miller D, Timpson P, Serrels B, Frame MC, et al. Quantitative in vivo imaging of the effects of inhibiting integrin signaling via Src and FAK on cancer cell movement: effects on E-cadherin dynamics. Cancer Res. 2010;70:9413–22.PubMedPubMedCentral Canel M, Serrels A, Miller D, Timpson P, Serrels B, Frame MC, et al. Quantitative in vivo imaging of the effects of inhibiting integrin signaling via Src and FAK on cancer cell movement: effects on E-cadherin dynamics. Cancer Res. 2010;70:9413–22.PubMedPubMedCentral
93.
Zurück zum Zitat Serrels A, Canel M, Brunton VG, Frame MC. Src/FAK-mediated regulation of E-cadherin as a mechanism for controlling collective cell movement: insights from in vivo imaging. Cell Adh Migr. 2011;5:360–5.PubMedPubMedCentral Serrels A, Canel M, Brunton VG, Frame MC. Src/FAK-mediated regulation of E-cadherin as a mechanism for controlling collective cell movement: insights from in vivo imaging. Cell Adh Migr. 2011;5:360–5.PubMedPubMedCentral
94.
Zurück zum Zitat Gayrard C, Bernaudin C, Déjardin T, Seiler C, Borghi N. Src- and confinement-dependent FAK activation causes E-cadherin relaxation and β-catenin activity. J Cell Biol. 2018;217:1063–77.PubMedPubMedCentral Gayrard C, Bernaudin C, Déjardin T, Seiler C, Borghi N. Src- and confinement-dependent FAK activation causes E-cadherin relaxation and β-catenin activity. J Cell Biol. 2018;217:1063–77.PubMedPubMedCentral
95.
Zurück zum Zitat Hauck CR, Hsia DA, Schlaepfer DD. The focal adhesion kinase—a regulator of cell migration and invasion. IUBMB Life. 2002;53:115–9.PubMed Hauck CR, Hsia DA, Schlaepfer DD. The focal adhesion kinase—a regulator of cell migration and invasion. IUBMB Life. 2002;53:115–9.PubMed
96.
Zurück zum Zitat Ma R-R, Zhang H, Chen H-F, Zhang G-H, Tian Y-R, Gao P. MiR-19a/miR-96-mediated low expression of KIF26A suppresses metastasis by regulating FAK pathway in gastric cancer. Oncogene. 2021;40:2524–38.PubMed Ma R-R, Zhang H, Chen H-F, Zhang G-H, Tian Y-R, Gao P. MiR-19a/miR-96-mediated low expression of KIF26A suppresses metastasis by regulating FAK pathway in gastric cancer. Oncogene. 2021;40:2524–38.PubMed
97.
Zurück zum Zitat Saito D, Kyakumoto S, Chosa N, Ibi M, Takahashi N, Okubo N, et al. Transforming growth factor-β1 induces epithelial-mesenchymal transition and integrin α3β1-mediated cell migration of HSC-4 human squamous cell carcinoma cells through Slug. J Biochem. 2013;153:303–15.PubMed Saito D, Kyakumoto S, Chosa N, Ibi M, Takahashi N, Okubo N, et al. Transforming growth factor-β1 induces epithelial-mesenchymal transition and integrin α3β1-mediated cell migration of HSC-4 human squamous cell carcinoma cells through Slug. J Biochem. 2013;153:303–15.PubMed
98.
Zurück zum Zitat Timbrell S, Aglan H, Cramer A, Foden P, Weaver D, Pachter J, et al. FAK inhibition alone or in combination with adjuvant therapies reduces cancer stem cell activity. NPJ Breast Cancer. 2021;7:65.PubMedPubMedCentral Timbrell S, Aglan H, Cramer A, Foden P, Weaver D, Pachter J, et al. FAK inhibition alone or in combination with adjuvant therapies reduces cancer stem cell activity. NPJ Breast Cancer. 2021;7:65.PubMedPubMedCentral
99.
Zurück zum Zitat Tancioni I, Miller NLG, Uryu S, Lawson C, Jean C, Chen XL, et al. FAK activity protects nucleostemin in facilitating breast cancer spheroid and tumor growth. Breast Cancer Res. 2015;17:47.PubMedPubMedCentral Tancioni I, Miller NLG, Uryu S, Lawson C, Jean C, Chen XL, et al. FAK activity protects nucleostemin in facilitating breast cancer spheroid and tumor growth. Breast Cancer Res. 2015;17:47.PubMedPubMedCentral
100.
Zurück zum Zitat Luo M, Fan H, Nagy T, Wei H, Wang C, Liu S, et al. Mammary epithelial-specific ablation of the focal adhesion kinase suppresses mammary tumorigenesis by affecting mammary cancer stem/progenitor cells. Cancer Res. 2009;69:466–74.PubMedPubMedCentral Luo M, Fan H, Nagy T, Wei H, Wang C, Liu S, et al. Mammary epithelial-specific ablation of the focal adhesion kinase suppresses mammary tumorigenesis by affecting mammary cancer stem/progenitor cells. Cancer Res. 2009;69:466–74.PubMedPubMedCentral
101.
Zurück zum Zitat Thiagarajan PS, Sinyuk M, Turaga SM, Mulkearns-Hubert EE, Hale JS, Rao V, et al. Cx26 drives self-renewal in triple-negative breast cancer via interaction with NANOG and focal adhesion kinase. Nat Commun. 2018;9:578.PubMedPubMedCentral Thiagarajan PS, Sinyuk M, Turaga SM, Mulkearns-Hubert EE, Hale JS, Rao V, et al. Cx26 drives self-renewal in triple-negative breast cancer via interaction with NANOG and focal adhesion kinase. Nat Commun. 2018;9:578.PubMedPubMedCentral
102.
Zurück zum Zitat Fan H, Zhao X, Sun S, Luo M, Guan J-L. Function of focal adhesion kinase scaffolding to mediate endophilin A2 phosphorylation promotes epithelial-mesenchymal transition and mammary cancer stem cell activities in vivo. J Biol Chem. 2013;288:3322–33.PubMed Fan H, Zhao X, Sun S, Luo M, Guan J-L. Function of focal adhesion kinase scaffolding to mediate endophilin A2 phosphorylation promotes epithelial-mesenchymal transition and mammary cancer stem cell activities in vivo. J Biol Chem. 2013;288:3322–33.PubMed
103.
Zurück zum Zitat Kolev VN, Tam WF, Wright QG, McDermott SP, Vidal CM, Shapiro IM, et al. Inhibition of FAK kinase activity preferentially targets cancer stem cells. Oncotarget. 2017;8:51733–47.PubMedPubMedCentral Kolev VN, Tam WF, Wright QG, McDermott SP, Vidal CM, Shapiro IM, et al. Inhibition of FAK kinase activity preferentially targets cancer stem cells. Oncotarget. 2017;8:51733–47.PubMedPubMedCentral
104.
Zurück zum Zitat Moon JH, Rho YS, Lee SH, Koo BS, Lee HJ, Do SI, et al. Role of integrin β1 as a biomarker of stemness in head and neck squamous cell carcinoma. Oral Oncol. 2019;96:34–41.PubMed Moon JH, Rho YS, Lee SH, Koo BS, Lee HJ, Do SI, et al. Role of integrin β1 as a biomarker of stemness in head and neck squamous cell carcinoma. Oral Oncol. 2019;96:34–41.PubMed
105.
Zurück zum Zitat Ming X-Y, Fu L, Zhang L-Y, Qin Y-R, Cao T-T, Chan KW, et al. Integrin α7 is a functional cancer stem cell surface marker in oesophageal squamous cell carcinoma. Nat Commun. 2016;7:13568.PubMedPubMedCentral Ming X-Y, Fu L, Zhang L-Y, Qin Y-R, Cao T-T, Chan KW, et al. Integrin α7 is a functional cancer stem cell surface marker in oesophageal squamous cell carcinoma. Nat Commun. 2016;7:13568.PubMedPubMedCentral
106.
Zurück zum Zitat Wang L, He T, Liu J, Tai J, Wang B, Chen Z, et al. Pan-cancer analysis reveals tumor-associated macrophage communication in the tumor microenvironment. Exp Hematol Oncol. 2021;10:31.PubMedPubMedCentral Wang L, He T, Liu J, Tai J, Wang B, Chen Z, et al. Pan-cancer analysis reveals tumor-associated macrophage communication in the tumor microenvironment. Exp Hematol Oncol. 2021;10:31.PubMedPubMedCentral
107.
Zurück zum Zitat Zhu S, Yi M, Wu Y, Dong B, Wu K. Roles of tumor-associated macrophages in tumor progression: implications on therapeutic strategies. Exp Hematol Oncol. 2021;10:60.PubMedPubMedCentral Zhu S, Yi M, Wu Y, Dong B, Wu K. Roles of tumor-associated macrophages in tumor progression: implications on therapeutic strategies. Exp Hematol Oncol. 2021;10:60.PubMedPubMedCentral
108.
Zurück zum Zitat Yan Y, Huang L, Liu Y, Yi M, Chu Q, Jiao D, et al. Metabolic profiles of regulatory T cells and their adaptations to the tumor microenvironment: implications for antitumor immunity. J Hematol Oncol. 2022;15:104.PubMedPubMedCentral Yan Y, Huang L, Liu Y, Yi M, Chu Q, Jiao D, et al. Metabolic profiles of regulatory T cells and their adaptations to the tumor microenvironment: implications for antitumor immunity. J Hematol Oncol. 2022;15:104.PubMedPubMedCentral
109.
Zurück zum Zitat Lei X, Lei Y, Li J-K, Du W-X, Li R-G, Yang J, et al. Immune cells within the tumor microenvironment: biological functions and roles in cancer immunotherapy. Cancer Lett. 2020;470:126–33.PubMed Lei X, Lei Y, Li J-K, Du W-X, Li R-G, Yang J, et al. Immune cells within the tumor microenvironment: biological functions and roles in cancer immunotherapy. Cancer Lett. 2020;470:126–33.PubMed
110.
Zurück zum Zitat Wendt MK, Schiemann WP. Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-beta signaling and metastasis. Breast Cancer Res. 2009;11:R68.PubMedPubMedCentral Wendt MK, Schiemann WP. Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-beta signaling and metastasis. Breast Cancer Res. 2009;11:R68.PubMedPubMedCentral
111.
Zurück zum Zitat Walsh C, Tanjoni I, Uryu S, Tomar A, Nam J-O, Luo H, et al. Oral delivery of PND-1186 FAK inhibitor decreases tumor growth and spontaneous breast to lung metastasis in pre-clinical models. Cancer Biol Ther. 2010;9:778–90.PubMed Walsh C, Tanjoni I, Uryu S, Tomar A, Nam J-O, Luo H, et al. Oral delivery of PND-1186 FAK inhibitor decreases tumor growth and spontaneous breast to lung metastasis in pre-clinical models. Cancer Biol Ther. 2010;9:778–90.PubMed
112.
Zurück zum Zitat Stokes JB, Adair SJ, Slack-Davis JK, Walters DM, Tilghman RW, Hershey ED, 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;10:2135–45.PubMedPubMedCentral Stokes JB, Adair SJ, Slack-Davis JK, Walters DM, Tilghman RW, Hershey ED, 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;10:2135–45.PubMedPubMedCentral
113.
Zurück zum Zitat Huehn J, Hamann A. Homing to suppress: address codes for Treg migration. Trends Immunol. 2005;26:632–6.PubMed Huehn J, Hamann A. Homing to suppress: address codes for Treg migration. Trends Immunol. 2005;26:632–6.PubMed
114.
Zurück zum Zitat Serrels A, Lund T, Serrels B, Byron A, McPherson RC, von Kriegsheim A, et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell. 2015;163:160–73.PubMedPubMedCentral Serrels A, Lund T, Serrels B, Byron A, McPherson RC, von Kriegsheim A, et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell. 2015;163:160–73.PubMedPubMedCentral
115.
Zurück zum Zitat Griffith BGC, Upstill-Goddard R, Brunton H, Grimes GR, Biankin AV, Serrels B, et al. FAK regulates IL-33 expression by controlling chromatin accessibility at c-Jun motifs. Sci Rep. 2021;11:229.PubMedPubMedCentral Griffith BGC, Upstill-Goddard R, Brunton H, Grimes GR, Biankin AV, Serrels B, et al. FAK regulates IL-33 expression by controlling chromatin accessibility at c-Jun motifs. Sci Rep. 2021;11:229.PubMedPubMedCentral
116.
Zurück zum Zitat Schiering C, Krausgruber T, Chomka A, Fröhlich A, Adelmann K, Wohlfert EA, et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature. 2014;513:564–8.PubMedPubMedCentral Schiering C, Krausgruber T, Chomka A, Fröhlich A, Adelmann K, Wohlfert EA, et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature. 2014;513:564–8.PubMedPubMedCentral
117.
Zurück zum Zitat Borst J, Ahrends T, Bąbała N, Melief CJM, Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18:635–47.PubMed Borst J, Ahrends T, Bąbała N, Melief CJM, Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18:635–47.PubMed
118.
Zurück zum Zitat Canel M, Taggart D, Sims AH, Lonergan DW, Waizenegger IC, Serrels A. T-cell co-stimulation in combination with targeting FAK drives enhanced anti-tumor immunity. Elife. 2020;9: e48092.PubMedPubMedCentral Canel M, Taggart D, Sims AH, Lonergan DW, Waizenegger IC, Serrels A. T-cell co-stimulation in combination with targeting FAK drives enhanced anti-tumor immunity. Elife. 2020;9: e48092.PubMedPubMedCentral
119.
Zurück zum Zitat Llewellyn RA, Gutknecht MF, Thomas KS, Conaway MR, Bouton AH. Focal adhesion kinase (FAK) deficiency in mononuclear phagocytes alters murine breast tumor progression. Am J Cancer Res. 2018;8:675–87.PubMedPubMedCentral Llewellyn RA, Gutknecht MF, Thomas KS, Conaway MR, Bouton AH. Focal adhesion kinase (FAK) deficiency in mononuclear phagocytes alters murine breast tumor progression. Am J Cancer Res. 2018;8:675–87.PubMedPubMedCentral
120.
Zurück zum Zitat Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18:99–115.PubMed Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18:99–115.PubMed
121.
Zurück zum Zitat Wang X, Zhou Q, Yu Z, Wu X, Chen X, Li J, et al. Cancer-associated fibroblast-derived Lumican promotes gastric cancer progression via the integrin β1-FAK signaling pathway. Int J Cancer. 2017;141:998–1010.PubMed Wang X, Zhou Q, Yu Z, Wu X, Chen X, Li J, et al. Cancer-associated fibroblast-derived Lumican promotes gastric cancer progression via the integrin β1-FAK signaling pathway. Int J Cancer. 2017;141:998–1010.PubMed
122.
Zurück zum Zitat Begum A, McMillan RH, Chang Y-T, Penchev VR, Rajeshkumar NV, Maitra A, et al. Direct interactions with cancer-associated fibroblasts lead to enhanced pancreatic cancer stem cell function. Pancreas. 2019;48:329–34.PubMedPubMedCentral Begum A, McMillan RH, Chang Y-T, Penchev VR, Rajeshkumar NV, Maitra A, et al. Direct interactions with cancer-associated fibroblasts lead to enhanced pancreatic cancer stem cell function. Pancreas. 2019;48:329–34.PubMedPubMedCentral
123.
Zurück zum Zitat Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22:851–60.PubMedPubMedCentral Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22:851–60.PubMedPubMedCentral
124.
Zurück zum Zitat Begum A, Ewachiw T, Jung C, Huang A, Norberg KJ, Marchionni L, et al. The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PLoS ONE. 2017;12: e0180181.PubMedPubMedCentral Begum A, Ewachiw T, Jung C, Huang A, Norberg KJ, Marchionni L, et al. The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PLoS ONE. 2017;12: e0180181.PubMedPubMedCentral
125.
Zurück zum Zitat Wu H-J, Hao M, Yeo SK, Guan J-L. FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene. 2020;39:2539–49.PubMedPubMedCentral Wu H-J, Hao M, Yeo SK, Guan J-L. FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene. 2020;39:2539–49.PubMedPubMedCentral
126.
Zurück zum Zitat Demircioglu F, Wang J, Candido J, Costa ASH, Casado P, de Luxan DB, et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun. 2020;11:1290.PubMedPubMedCentral Demircioglu F, Wang J, Candido J, Costa ASH, Casado P, de Luxan DB, et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun. 2020;11:1290.PubMedPubMedCentral
127.
Zurück zum Zitat Rizzo MT. Focal adhesion kinase and angiogenesis. Where do we go from here? Cardiovasc Res. 2004;64:377–8.PubMed Rizzo MT. Focal adhesion kinase and angiogenesis. Where do we go from here? Cardiovasc Res. 2004;64:377–8.PubMed
128.
Zurück zum Zitat Peng X, Ueda H, Zhou H, Stokol T, Shen T-L, Alcaraz A, et al. Overexpression of focal adhesion kinase in vascular endothelial cells promotes angiogenesis in transgenic mice. Cardiovasc Res. 2004;64:421–30.PubMed Peng X, Ueda H, Zhou H, Stokol T, Shen T-L, Alcaraz A, et al. Overexpression of focal adhesion kinase in vascular endothelial cells promotes angiogenesis in transgenic mice. Cardiovasc Res. 2004;64:421–30.PubMed
129.
Zurück zum Zitat Tavora B, Batista S, Reynolds LE, Jadeja S, Robinson S, Kostourou V, et al. Endothelial FAK is required for tumour angiogenesis. EMBO Mol Med. 2016;8:1229.PubMedPubMedCentral Tavora B, Batista S, Reynolds LE, Jadeja S, Robinson S, Kostourou V, et al. Endothelial FAK is required for tumour angiogenesis. EMBO Mol Med. 2016;8:1229.PubMedPubMedCentral
130.
Zurück zum Zitat Kurio N, Shimo T, Fukazawa T, Okui T, Hassan NMM, Honami T, et al. Anti-tumor effect of a novel FAK inhibitor TAE226 against human oral squamous cell carcinoma. Oral Oncol. 2012;48:1159–70.PubMed Kurio N, Shimo T, Fukazawa T, Okui T, Hassan NMM, Honami T, et al. Anti-tumor effect of a novel FAK inhibitor TAE226 against human oral squamous cell carcinoma. Oral Oncol. 2012;48:1159–70.PubMed
131.
Zurück zum Zitat Rothhut B, Ghoneim C, Antonicelli F, Soula-Rothhut M. Epidermal growth factor stimulates matrix metalloproteinase-9 expression and invasion in human follicular thyroid carcinoma cells through Focal adhesion kinase. Biochimie. 2007;89:613–24.PubMed Rothhut B, Ghoneim C, Antonicelli F, Soula-Rothhut M. Epidermal growth factor stimulates matrix metalloproteinase-9 expression and invasion in human follicular thyroid carcinoma cells through Focal adhesion kinase. Biochimie. 2007;89:613–24.PubMed
132.
Zurück zum Zitat Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol. 2022;15:143.PubMedPubMedCentral Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol. 2022;15:143.PubMedPubMedCentral
133.
Zurück zum Zitat Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol. 2021;14:45.PubMedPubMedCentral Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol. 2021;14:45.PubMedPubMedCentral
134.
Zurück zum Zitat Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y, et al. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 2022;15:24.PubMedPubMedCentral Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y, et al. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 2022;15:24.PubMedPubMedCentral
135.
Zurück zum Zitat Wu H, Gong Y, Ji P, Xie Y, Jiang Y-Z, Liu G. Targeting nucleotide metabolism: a promising approach to enhance cancer immunotherapy. J Hematol Oncol. 2022;15:45.PubMedPubMedCentral Wu H, Gong Y, Ji P, Xie Y, Jiang Y-Z, Liu G. Targeting nucleotide metabolism: a promising approach to enhance cancer immunotherapy. J Hematol Oncol. 2022;15:45.PubMedPubMedCentral
136.
Zurück zum Zitat Dai M, Liu M, Yang H, Küçük C, You H. New insights into epigenetic regulation of resistance to PD-1/PD-L1 blockade cancer immunotherapy: mechanisms and therapeutic opportunities. Exp Hematol Oncol. 2022;11:101.PubMedPubMedCentral Dai M, Liu M, Yang H, Küçük C, You H. New insights into epigenetic regulation of resistance to PD-1/PD-L1 blockade cancer immunotherapy: mechanisms and therapeutic opportunities. Exp Hematol Oncol. 2022;11:101.PubMedPubMedCentral
137.
Zurück zum Zitat Zhu S, Wu Y, Song B, Yi M, Yan Y, Mei Q, et al. Recent advances in targeted strategies for triple-negative breast cancer. J Hematol Oncol. 2023;16:100.PubMedPubMedCentral Zhu S, Wu Y, Song B, Yi M, Yan Y, Mei Q, et al. Recent advances in targeted strategies for triple-negative breast cancer. J Hematol Oncol. 2023;16:100.PubMedPubMedCentral
138.
Zurück zum Zitat Mohan N, Hosain S, Zhao J, Shen Y, Luo X, Jiang J, et al. Atezolizumab potentiates Tcell-mediated cytotoxicity and coordinates with FAK to suppress cell invasion and motility in PD-L1+ triple negative breast cancer cells. Oncoimmunology. 2019;8: e1624128.PubMedPubMedCentral Mohan N, Hosain S, Zhao J, Shen Y, Luo X, Jiang J, et al. Atezolizumab potentiates Tcell-mediated cytotoxicity and coordinates with FAK to suppress cell invasion and motility in PD-L1+ triple negative breast cancer cells. Oncoimmunology. 2019;8: e1624128.PubMedPubMedCentral
139.
Zurück zum Zitat Gao X, Mi Y, Guo N, Xu H, Xu L, Gou X, et al. Cytokine-induced killer cells as pharmacological tools for cancer immunotherapy. Front Immunol. 2017;8:774.PubMedPubMedCentral Gao X, Mi Y, Guo N, Xu H, Xu L, Gou X, et al. Cytokine-induced killer cells as pharmacological tools for cancer immunotherapy. Front Immunol. 2017;8:774.PubMedPubMedCentral
140.
Zurück zum Zitat Pan M-R, Wu C-C, Kan J-Y, Li Q-L, Chang S-J, Wu C-C, et al. Impact of FAK expression on the cytotoxic effects of CIK therapy in triple-negative breast cancer. Cancers. 2019;12:94.PubMedPubMedCentral Pan M-R, Wu C-C, Kan J-Y, Li Q-L, Chang S-J, Wu C-C, et al. Impact of FAK expression on the cytotoxic effects of CIK therapy in triple-negative breast cancer. Cancers. 2019;12:94.PubMedPubMedCentral
141.
Zurück zum Zitat Wei Y, Wang Y, Liu N, Qi R, Xu Y, Li K, et al. A FAK inhibitor boosts anti-PD1 immunotherapy in a hepatocellular carcinoma mouse model. Front Pharmacol. 2021;12: 820446.PubMed Wei Y, Wang Y, Liu N, Qi R, Xu Y, Li K, et al. A FAK inhibitor boosts anti-PD1 immunotherapy in a hepatocellular carcinoma mouse model. Front Pharmacol. 2021;12: 820446.PubMed
142.
Zurück zum Zitat Ozmadenci D, Shankara Narayanan JS, Andrew J, Ojalill M, Barrie AM, Jiang S, et al. Tumor FAK orchestrates immunosuppression in ovarian cancer via the CD155/TIGIT axis. Proc Natl Acad Sci USA. 2022;119: e2117065119.PubMedPubMedCentral Ozmadenci D, Shankara Narayanan JS, Andrew J, Ojalill M, Barrie AM, Jiang S, et al. Tumor FAK orchestrates immunosuppression in ovarian cancer via the CD155/TIGIT axis. Proc Natl Acad Sci USA. 2022;119: e2117065119.PubMedPubMedCentral
143.
Zurück zum Zitat Shan S, Fang B, Zhang Y, Wang C, Zhou J, Niu C, et al. Mechanical stretch promotes tumoricidal M1 polarization via the FAK/NF-κB signaling pathway. FASEB J. 2019;33:13254–66.PubMed Shan S, Fang B, Zhang Y, Wang C, Zhou J, Niu C, et al. Mechanical stretch promotes tumoricidal M1 polarization via the FAK/NF-κB signaling pathway. FASEB J. 2019;33:13254–66.PubMed
144.
Zurück zum Zitat Dragoj M, Milosevic Z, Bankovic J, Tanic N, Pesic M, Stankovic T. Targeting CXCR4 and FAK reverses doxorubicin resistance and suppresses invasion in non-small cell lung carcinoma. Cell Oncol. 2017;40:47–62. Dragoj M, Milosevic Z, Bankovic J, Tanic N, Pesic M, Stankovic T. Targeting CXCR4 and FAK reverses doxorubicin resistance and suppresses invasion in non-small cell lung carcinoma. Cell Oncol. 2017;40:47–62.
145.
Zurück zum Zitat Zhang B, Zhang Y, Zhang J, Liu P, Jiao B, Wang Z, et al. Focal adhesion kinase (FAK) inhibition synergizes with KRAS G12C inhibitors in treating cancer through the regulation of the FAK-YAP signaling. Adv Sci. 2021;8: e2100250. Zhang B, Zhang Y, Zhang J, Liu P, Jiao B, Wang Z, et al. Focal adhesion kinase (FAK) inhibition synergizes with KRAS G12C inhibitors in treating cancer through the regulation of the FAK-YAP signaling. Adv Sci. 2021;8: e2100250.
146.
Zurück zum Zitat Paradis JS, Acosta M, Saddawi-Konefka R, Kishore A, Gomes F, Arang N, et al. Synthetic lethal screens reveal cotargeting FAK and MEK as a multimodal precision therapy for GNAQ-driven uveal melanoma. Clin Cancer Res. 2021;27:3190–200.PubMedPubMedCentral Paradis JS, Acosta M, Saddawi-Konefka R, Kishore A, Gomes F, Arang N, et al. Synthetic lethal screens reveal cotargeting FAK and MEK as a multimodal precision therapy for GNAQ-driven uveal melanoma. Clin Cancer Res. 2021;27:3190–200.PubMedPubMedCentral
147.
Zurück zum Zitat Chen G, Gao C, Gao X, Zhang DH, Kuan S-F, Burns TF, et al. Wnt/β-catenin pathway activation mediates adaptive resistance to BRAF inhibition in colorectal cancer. Mol Cancer Ther. 2018;17:806–13.PubMed Chen G, Gao C, Gao X, Zhang DH, Kuan S-F, Burns TF, et al. Wnt/β-catenin pathway activation mediates adaptive resistance to BRAF inhibition in colorectal cancer. Mol Cancer Ther. 2018;17:806–13.PubMed
148.
Zurück zum Zitat Taylor KN, Schlaepfer DD. Adaptive resistance to chemotherapy, a multi-FAK-torial linkage. Mol Cancer Ther. 2018;17:719–23.PubMedPubMedCentral Taylor KN, Schlaepfer DD. Adaptive resistance to chemotherapy, a multi-FAK-torial linkage. Mol Cancer Ther. 2018;17:719–23.PubMedPubMedCentral
149.
Zurück zum Zitat Romito I, Porru M, Braghini MR, Pompili L, Panera N, Crudele A, et al. Focal adhesion kinase inhibitor TAE226 combined with Sorafenib slows down hepatocellular carcinoma by multiple epigenetic effects. J Exp Clin Cancer Res. 2021;40:364.PubMedPubMedCentral Romito I, Porru M, Braghini MR, Pompili L, Panera N, Crudele A, et al. Focal adhesion kinase inhibitor TAE226 combined with Sorafenib slows down hepatocellular carcinoma by multiple epigenetic effects. J Exp Clin Cancer Res. 2021;40:364.PubMedPubMedCentral
150.
Zurück zum Zitat Xu B, Lefringhouse J, Liu Z, West D, Baldwin LA, Ou C, et al. Inhibition of the integrin/FAK signaling axis and c-Myc synergistically disrupts ovarian cancer malignancy. Oncogenesis. 2017;6: e295.PubMedPubMedCentral Xu B, Lefringhouse J, Liu Z, West D, Baldwin LA, Ou C, et al. Inhibition of the integrin/FAK signaling axis and c-Myc synergistically disrupts ovarian cancer malignancy. Oncogenesis. 2017;6: e295.PubMedPubMedCentral
151.
Zurück zum Zitat Dawson JC, Serrels B, Byron A, Muir MT, Makda A, García-Muñoz A, et al. A synergistic anticancer FAK and HDAC inhibitor combination discovered by a novel chemical-genetic high-content phenotypic screen. Mol Cancer Ther. 2020;19:637–49.PubMed Dawson JC, Serrels B, Byron A, Muir MT, Makda A, García-Muñoz A, et al. A synergistic anticancer FAK and HDAC inhibitor combination discovered by a novel chemical-genetic high-content phenotypic screen. Mol Cancer Ther. 2020;19:637–49.PubMed
152.
Zurück zum Zitat Ichihara E, Westover D, Meador CB, Yan Y, Bauer JA, Lu P, et al. SFK/FAK signaling attenuates osimertinib efficacy in both drug-sensitive and drug-resistant models of EGFR-mutant lung cancer. Cancer Res. 2017;77:2990–3000.PubMedPubMedCentral Ichihara E, Westover D, Meador CB, Yan Y, Bauer JA, Lu P, et al. SFK/FAK signaling attenuates osimertinib efficacy in both drug-sensitive and drug-resistant models of EGFR-mutant lung cancer. Cancer Res. 2017;77:2990–3000.PubMedPubMedCentral
153.
Zurück zum Zitat Jiang H, Liu X, Knolhoff BL, Hegde S, Lee KB, Jiang H, et al. Development of resistance to FAK inhibition in pancreatic cancer is linked to stromal depletion. Gut. 2020;69:122–32.PubMed Jiang H, Liu X, Knolhoff BL, Hegde S, Lee KB, Jiang H, et al. Development of resistance to FAK inhibition in pancreatic cancer is linked to stromal depletion. Gut. 2020;69:122–32.PubMed
154.
Zurück zum Zitat Byeon Y, Lee J-W, Choi WS, Won JE, Kim GH, Kim MG, et al. CD44-targeting PLGA nanoparticles incorporating paclitaxel and FAK siRNA overcome chemoresistance in epithelial ovarian cancer. Cancer Res. 2018;78:6247–56.PubMed Byeon Y, Lee J-W, Choi WS, Won JE, Kim GH, Kim MG, et al. CD44-targeting PLGA nanoparticles incorporating paclitaxel and FAK siRNA overcome chemoresistance in epithelial ovarian cancer. Cancer Res. 2018;78:6247–56.PubMed
155.
Zurück zum Zitat Hou J, Tan Y, Su C, Wang T, Gao Z, Song D, et al. Inhibition of protein FAK enhances 5-FU chemosensitivity to gastric carcinoma via p53 signaling pathways. Comput Struct Biotechnol J. 2020;18:125–36.PubMed Hou J, Tan Y, Su C, Wang T, Gao Z, Song D, et al. Inhibition of protein FAK enhances 5-FU chemosensitivity to gastric carcinoma via p53 signaling pathways. Comput Struct Biotechnol J. 2020;18:125–36.PubMed
156.
Zurück zum Zitat Le Large TYS, Bijlsma MF, El Hassouni B, Mantini G, Lagerweij T, Henneman AA, et al. Focal adhesion kinase inhibition synergizes with nab-paclitaxel to target pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. 2021;40:91.PubMedPubMedCentral Le Large TYS, Bijlsma MF, El Hassouni B, Mantini G, Lagerweij T, Henneman AA, et al. Focal adhesion kinase inhibition synergizes with nab-paclitaxel to target pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. 2021;40:91.PubMedPubMedCentral
157.
Zurück zum Zitat Roy-Luzarraga M, Reynolds LE, de Luxán-Delgado B, Maiques O, Wisniewski L, Newport E, et al. Suppression of endothelial Cell FAK expression reduces pancreatic ductal adenocarcinoma metastasis after gemcitabine treatment. Cancer Res. 2022;82:1909–25.PubMedPubMedCentral Roy-Luzarraga M, Reynolds LE, de Luxán-Delgado B, Maiques O, Wisniewski L, Newport E, et al. Suppression of endothelial Cell FAK expression reduces pancreatic ductal adenocarcinoma metastasis after gemcitabine treatment. Cancer Res. 2022;82:1909–25.PubMedPubMedCentral
158.
Zurück zum Zitat Tavora B, Reynolds LE, Batista S, Demircioglu F, Fernandez I, Lechertier T, et al. Endothelial–cell FAK targeting sensitizes tumours to DNA–damaging therapy. Nature. 2014;514:112–6.PubMedPubMedCentral Tavora B, Reynolds LE, Batista S, Demircioglu F, Fernandez I, Lechertier T, et al. Endothelial–cell FAK targeting sensitizes tumours to DNA–damaging therapy. Nature. 2014;514:112–6.PubMedPubMedCentral
159.
Zurück zum Zitat Roy-Luzarraga M, Abdel-Fatah T, Reynolds LE, Clear A, Taylor JG, Gribben JG, et al. Association of low tumor endothelial cell pY397-focal adhesion kinase expression with survival in patients with neoadjuvant-treated locally advanced breast cancer. JAMA Netw Open. 2020;3: e2019304.PubMedPubMedCentral Roy-Luzarraga M, Abdel-Fatah T, Reynolds LE, Clear A, Taylor JG, Gribben JG, et al. Association of low tumor endothelial cell pY397-focal adhesion kinase expression with survival in patients with neoadjuvant-treated locally advanced breast cancer. JAMA Netw Open. 2020;3: e2019304.PubMedPubMedCentral
160.
Zurück zum Zitat Chen H, Tu W, Lu Y, Zhang Y, Xu Y, Chen X, et al. Low-dose X-ray irradiation combined with FAK inhibitors improves the immune microenvironment and confers sensitivity to radiotherapy in pancreatic cancer. Biomed Pharmacother. 2022;151: 113114.PubMed Chen H, Tu W, Lu Y, Zhang Y, Xu Y, Chen X, et al. Low-dose X-ray irradiation combined with FAK inhibitors improves the immune microenvironment and confers sensitivity to radiotherapy in pancreatic cancer. Biomed Pharmacother. 2022;151: 113114.PubMed
161.
Zurück zum Zitat Mohamed AA, Thomsen A, Follo M, Zamboglou C, Bronsert P, Mostafa H, et al. FAK inhibition radiosensitizes pancreatic ductal adenocarcinoma cells in vitro. Strahlenther Onkol. 2021;197:27–38.PubMed Mohamed AA, Thomsen A, Follo M, Zamboglou C, Bronsert P, Mostafa H, et al. FAK inhibition radiosensitizes pancreatic ductal adenocarcinoma cells in vitro. Strahlenther Onkol. 2021;197:27–38.PubMed
162.
Zurück zum Zitat Osipov A, Blair AB, Liberto J, Wang J, Li K, Herbst B, et al. Inhibition of focal adhesion kinase enhances antitumor response of radiation therapy in pancreatic cancer through CD8+ T cells. Cancer Biol Med. 2021;18:206–14.PubMedPubMedCentral Osipov A, Blair AB, Liberto J, Wang J, Li K, Herbst B, et al. Inhibition of focal adhesion kinase enhances antitumor response of radiation therapy in pancreatic cancer through CD8+ T cells. Cancer Biol Med. 2021;18:206–14.PubMedPubMedCentral
163.
Zurück zum Zitat Williams KE, Bundred NJ, Landberg G, Clarke RB, Farnie G. Focal adhesion kinase and Wnt signaling regulate human ductal carcinoma in situ stem cell activity and response to radiotherapy. Stem Cells. 2015;33:327–41.PubMed Williams KE, Bundred NJ, Landberg G, Clarke RB, Farnie G. Focal adhesion kinase and Wnt signaling regulate human ductal carcinoma in situ stem cell activity and response to radiotherapy. Stem Cells. 2015;33:327–41.PubMed
164.
Zurück zum Zitat NguemgoKouam P, Bühler H, Hero T, Adamietz IA. The increased adhesion of tumor cells to endothelial cells after irradiation can be reduced by FAK-inhibition. Radiat Oncol. 2019;14:25. NguemgoKouam P, Bühler H, Hero T, Adamietz IA. The increased adhesion of tumor cells to endothelial cells after irradiation can be reduced by FAK-inhibition. Radiat Oncol. 2019;14:25.
165.
Zurück zum Zitat Si Q, Ye Q, Bing Z, Fan R, Hu X, Liu B, et al. Carbon ion irradiation enhances the anti-tumor efficiency in tongue squamous cell carcinoma via modulating the FAK signaling. Front Public Health. 2021;9: 631118.PubMedPubMedCentral Si Q, Ye Q, Bing Z, Fan R, Hu X, Liu B, et al. Carbon ion irradiation enhances the anti-tumor efficiency in tongue squamous cell carcinoma via modulating the FAK signaling. Front Public Health. 2021;9: 631118.PubMedPubMedCentral
166.
Zurück zum Zitat Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: mechanistic findings and clinical applications. Nat Rev Cancer. 2014;14:598–610.PubMedPubMedCentral Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: mechanistic findings and clinical applications. Nat Rev Cancer. 2014;14:598–610.PubMedPubMedCentral
167.
Zurück zum Zitat Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol. 2006;18:516–23.PubMed Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol. 2006;18:516–23.PubMed
168.
Zurück zum Zitat Diaz Osterman CJ, Ozmadenci D, Kleinschmidt EG, Taylor KN, Barrie AM, Jiang S, et al. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy. Elife. 2019;8: e47327.PubMedPubMedCentral Diaz Osterman CJ, Ozmadenci D, Kleinschmidt EG, Taylor KN, Barrie AM, Jiang S, et al. FAK activity sustains intrinsic and acquired ovarian cancer resistance to platinum chemotherapy. Elife. 2019;8: e47327.PubMedPubMedCentral
169.
Zurück zum Zitat Qin Z, Zhou C. HOXA13 promotes gastric cancer progression partially via the FN1-mediated FAK/Src axis. Exp Hematol Oncol. 2022;11:7.PubMedPubMedCentral Qin Z, Zhou C. HOXA13 promotes gastric cancer progression partially via the FN1-mediated FAK/Src axis. Exp Hematol Oncol. 2022;11:7.PubMedPubMedCentral
170.
Zurück zum Zitat Dawson JC, Serrels A, Stupack DG, Schlaepfer DD, Frame MC. Targeting FAK in anticancer combination therapies. Nat Rev Cancer. 2021;21:313–24.PubMedPubMedCentral Dawson JC, Serrels A, Stupack DG, Schlaepfer DD, Frame MC. Targeting FAK in anticancer combination therapies. Nat Rev Cancer. 2021;21:313–24.PubMedPubMedCentral
171.
Zurück zum Zitat Xiong J, Yan L, Zou C, Wang K, Chen M, Xu B, et al. Integrins regulate stemness in solid tumor: an emerging therapeutic target. J Hematol Oncol. 2021;14:177.PubMedPubMedCentral Xiong J, Yan L, Zou C, Wang K, Chen M, Xu B, et al. Integrins regulate stemness in solid tumor: an emerging therapeutic target. J Hematol Oncol. 2021;14:177.PubMedPubMedCentral
172.
Zurück zum Zitat Lin T-H, Tan T-W, Tsai T-H, Chen C-C, Hsieh T-F, Lee S-S, et al. d-Pinitol inhibits prostate cancer metastasis through inhibition of αVβ3 integrin by modulating FAK, c-Src and NF-κB pathways. Int J Mol Sci. 2013;14:9790–802.PubMedPubMedCentral Lin T-H, Tan T-W, Tsai T-H, Chen C-C, Hsieh T-F, Lee S-S, et al. d-Pinitol inhibits prostate cancer metastasis through inhibition of αVβ3 integrin by modulating FAK, c-Src and NF-κB pathways. Int J Mol Sci. 2013;14:9790–802.PubMedPubMedCentral
173.
Zurück zum Zitat Cromm PM, Samarasinghe KTG, Hines J, Crews CM. Addressing kinase-independent functions of Fak via PROTAC-mediated degradation. J Am Chem Soc. 2018;140:17019–26.PubMed Cromm PM, Samarasinghe KTG, Hines J, Crews CM. Addressing kinase-independent functions of Fak via PROTAC-mediated degradation. J Am Chem Soc. 2018;140:17019–26.PubMed
174.
Zurück zum Zitat Li H, Dong J, Cai M, Xu Z, Cheng X-D, Qin J-J. Protein degradation technology: a strategic paradigm shift in drug discovery. J Hematol Oncol. 2021;14:138.PubMedPubMedCentral Li H, Dong J, Cai M, Xu Z, Cheng X-D, Qin J-J. Protein degradation technology: a strategic paradigm shift in drug discovery. J Hematol Oncol. 2021;14:138.PubMedPubMedCentral
175.
Zurück zum Zitat Roberts WG, Ung E, Whalen P, Cooper B, Hulford C, Autry C, et al. Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Can Res. 2008;68:1935–44. Roberts WG, Ung E, Whalen P, Cooper B, Hulford C, Autry C, et al. Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Can Res. 2008;68:1935–44.
176.
Zurück zum Zitat Sun H, Pisle S, Gardner ER, Figg WD. Bioluminescent imaging study: FAK inhibitor, PF-562,271, preclinical study in PC3M-luc-C6 local implant and metastasis xenograft models. Cancer Biol Ther. 2010;10:38–43.PubMedPubMedCentral Sun H, Pisle S, Gardner ER, Figg WD. Bioluminescent imaging study: FAK inhibitor, PF-562,271, preclinical study in PC3M-luc-C6 local implant and metastasis xenograft models. Cancer Biol Ther. 2010;10:38–43.PubMedPubMedCentral
177.
Zurück zum Zitat Slack-Davis JK, Hershey ED, Theodorescu D, Frierson HF, Parsons JT. Differential requirement for focal adhesion kinase signaling in cancer progression in the transgenic adenocarcinoma of mouse prostate model. Mol Cancer Ther. 2009;8:2470–7.PubMedPubMedCentral Slack-Davis JK, Hershey ED, Theodorescu D, Frierson HF, Parsons JT. Differential requirement for focal adhesion kinase signaling in cancer progression in the transgenic adenocarcinoma of mouse prostate model. Mol Cancer Ther. 2009;8:2470–7.PubMedPubMedCentral
178.
Zurück zum Zitat Serrels A, McLeod K, Canel M, Kinnaird A, Graham K, Frame MC, et al. The role of focal adhesion kinase catalytic activity on the proliferation and migration of squamous cell carcinoma cells. Int J Cancer. 2012;131:287–97.PubMed Serrels A, McLeod K, Canel M, Kinnaird A, Graham K, Frame MC, et al. The role of focal adhesion kinase catalytic activity on the proliferation and migration of squamous cell carcinoma cells. Int J Cancer. 2012;131:287–97.PubMed
179.
Zurück zum Zitat Bagi CM, Christensen J, Cohen DP, Roberts WG, Wilkie D, Swanson T, et al. 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;8:856–65.PubMed Bagi CM, Christensen J, Cohen DP, Roberts WG, Wilkie D, Swanson T, et al. 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;8:856–65.PubMed
180.
Zurück zum Zitat Kessler BE, Sharma V, Zhou Q, Jing X, Pike LA, Kerege AA, et al. FAK expression, not kinase activity, is a key mediator of thyroid tumorigenesis and protumorigenic processes. Mol Cancer Res. 2016;14:869–82.PubMedPubMedCentral Kessler BE, Sharma V, Zhou Q, Jing X, Pike LA, Kerege AA, et al. FAK expression, not kinase activity, is a key mediator of thyroid tumorigenesis and protumorigenic processes. Mol Cancer Res. 2016;14:869–82.PubMedPubMedCentral
181.
Zurück zum Zitat Newport E, Pedrosa AR, Lees D, Dukinfield M, Carter E, Gomez-Escudero J, et al. Elucidating the role of the kinase activity of endothelial cell focal adhesion kinase in angiocrine signalling and tumour growth. J Pathol. 2022;256:235–47.PubMed Newport E, Pedrosa AR, Lees D, Dukinfield M, Carter E, Gomez-Escudero J, et al. Elucidating the role of the kinase activity of endothelial cell focal adhesion kinase in angiocrine signalling and tumour growth. J Pathol. 2022;256:235–47.PubMed
182.
Zurück zum Zitat Stone RL, Baggerly KA, Armaiz-Pena GN, Kang Y, Sanguino AM, Thanapprapasr D, et al. Focal adhesion kinase: an alternative focus for anti-angiogenesis therapy in ovarian cancer. Cancer Biol Ther. 2014;15:919–29.PubMedPubMedCentral Stone RL, Baggerly KA, Armaiz-Pena GN, Kang Y, Sanguino AM, Thanapprapasr D, et al. Focal adhesion kinase: an alternative focus for anti-angiogenesis therapy in ovarian cancer. Cancer Biol Ther. 2014;15:919–29.PubMedPubMedCentral
183.
Zurück zum Zitat Wang S, Hwang EE, Guha R, O’Neill AF, Melong N, Veinotte CJ, et al. High-throughput chemical screening identifies focal adhesion kinase and aurora kinase B inhibition as a synergistic treatment combination in Ewing sarcoma. Clin Cancer Res. 2019;25:4552–66.PubMedPubMedCentral Wang S, Hwang EE, Guha R, O’Neill AF, Melong N, Veinotte CJ, et al. High-throughput chemical screening identifies focal adhesion kinase and aurora kinase B inhibition as a synergistic treatment combination in Ewing sarcoma. Clin Cancer Res. 2019;25:4552–66.PubMedPubMedCentral
184.
Zurück zum Zitat Lander VE, Belle JI, Kingstonl NL, Herndon JM, Hogg GD, Liu X, et al. Stromal reprogramming by FAK inhibition overcomes radiation resistance to allow for immune priming and response to checkpoint blockade. Cancer Discov. 2022;12:2774–99.PubMedPubMedCentral Lander VE, Belle JI, Kingstonl NL, Herndon JM, Hogg GD, Liu X, et al. Stromal reprogramming by FAK inhibition overcomes radiation resistance to allow for immune priming and response to checkpoint blockade. Cancer Discov. 2022;12:2774–99.PubMedPubMedCentral
185.
Zurück zum Zitat Fu Y, Zhang Y, Lei Z, Liu T, Cai T, Wang A, et al. Abnormally activated OPN/integrin αVβ3/FAK signalling is responsible for EGFR-TKI resistance in EGFR mutant non-small-cell lung cancer. J Hematol Oncol. 2020;13:169.PubMedPubMedCentral Fu Y, Zhang Y, Lei Z, Liu T, Cai T, Wang A, et al. Abnormally activated OPN/integrin αVβ3/FAK signalling is responsible for EGFR-TKI resistance in EGFR mutant non-small-cell lung cancer. J Hematol Oncol. 2020;13:169.PubMedPubMedCentral
186.
Zurück zum Zitat Zhang Y, Cheng K, Xu B, Shi J, Qiang J, Shi S, et al. Epigenetic input dictates the threshold of targeting of the integrin-dependent pathway in non-small cell lung cancer. Front Cell Dev Biol. 2020;8:652.PubMedPubMedCentral Zhang Y, Cheng K, Xu B, Shi J, Qiang J, Shi S, et al. Epigenetic input dictates the threshold of targeting of the integrin-dependent pathway in non-small cell lung cancer. Front Cell Dev Biol. 2020;8:652.PubMedPubMedCentral
187.
Zurück zum Zitat Tong X, Tanino R, Sun R, Tsubata Y, Okimoto T, Takechi M, et al. Protein tyrosine kinase 2: a novel therapeutic target to overcome acquired EGFR-TKI resistance in non-small cell lung cancer. Respir Res. 2019;20:270.PubMedPubMedCentral Tong X, Tanino R, Sun R, Tsubata Y, Okimoto T, Takechi M, et al. Protein tyrosine kinase 2: a novel therapeutic target to overcome acquired EGFR-TKI resistance in non-small cell lung cancer. Respir Res. 2019;20:270.PubMedPubMedCentral
188.
Zurück zum Zitat Lin H-M, Lee BY, Castillo L, Spielman C, Grogan J, Yeung NK, et al. Effect of FAK inhibitor VS-6063 (defactinib) on docetaxel efficacy in prostate cancer. Prostate. 2018;78:308–17.PubMed Lin H-M, Lee BY, Castillo L, Spielman C, Grogan J, Yeung NK, et al. Effect of FAK inhibitor VS-6063 (defactinib) on docetaxel efficacy in prostate cancer. Prostate. 2018;78:308–17.PubMed
189.
Zurück zum Zitat Kang Y, Hu W, Ivan C, Dalton HJ, Miyake T, Pecot CV, et al. Role of focal adhesion kinase in regulating YB-1-mediated paclitaxel resistance in ovarian cancer. J Natl Cancer Inst. 2013;105:1485–95.PubMedPubMedCentral Kang Y, Hu W, Ivan C, Dalton HJ, Miyake T, Pecot CV, et al. Role of focal adhesion kinase in regulating YB-1-mediated paclitaxel resistance in ovarian cancer. J Natl Cancer Inst. 2013;105:1485–95.PubMedPubMedCentral
190.
Zurück zum Zitat Huo X, Zhang W, Zhao G, Chen Z, Dong P, Watari H, et al. FAK PROTAC inhibits ovarian tumor growth and metastasis by disrupting kinase dependent and independent pathways. Front Oncol. 2022;12: 851065.PubMedPubMedCentral Huo X, Zhang W, Zhao G, Chen Z, Dong P, Watari H, et al. FAK PROTAC inhibits ovarian tumor growth and metastasis by disrupting kinase dependent and independent pathways. Front Oncol. 2022;12: 851065.PubMedPubMedCentral
191.
Zurück zum Zitat Zhang L, Zhao D, Wang Y, Zhang W, Zhang J, Fan J, et al. Focal adhesion kinase (FAK) inhibitor-defactinib suppresses the malignant progression of human esophageal squamous cell carcinoma (ESCC) cells via effective blockade of PI3K/AKT axis and downstream molecular network. Mol Carcinog. 2021;60:113–24.PubMed Zhang L, Zhao D, Wang Y, Zhang W, Zhang J, Fan J, et al. Focal adhesion kinase (FAK) inhibitor-defactinib suppresses the malignant progression of human esophageal squamous cell carcinoma (ESCC) cells via effective blockade of PI3K/AKT axis and downstream molecular network. Mol Carcinog. 2021;60:113–24.PubMed
192.
Zurück zum Zitat Lopez-Mejia IC, Pijuan J, Navaridas R, Santacana M, Gatius S, Velasco A, et al. Oxidative stress-induced FAK activation contributes to uterine serous carcinoma aggressiveness. Mol Oncol. 2023;17:98–118.PubMed Lopez-Mejia IC, Pijuan J, Navaridas R, Santacana M, Gatius S, Velasco A, et al. Oxidative stress-induced FAK activation contributes to uterine serous carcinoma aggressiveness. Mol Oncol. 2023;17:98–118.PubMed
193.
Zurück zum Zitat Tiede S, Meyer-Schaller N, Kalathur RKR, Ivanek R, Fagiani E, Schmassmann P, et al. The FAK inhibitor BI 853520 exerts anti-tumor effects in breast cancer. Oncogenesis. 2018;7:73.PubMedPubMedCentral Tiede S, Meyer-Schaller N, Kalathur RKR, Ivanek R, Fagiani E, Schmassmann P, et al. The FAK inhibitor BI 853520 exerts anti-tumor effects in breast cancer. Oncogenesis. 2018;7:73.PubMedPubMedCentral
194.
Zurück zum Zitat Li H, Gao Y, Ren C. Focal adhesion kinase inhibitor BI 853520 inhibits cell proliferation, migration and EMT process through PI3K/AKT/mTOR signaling pathway in ovarian cancer. Discov Oncol. 2021;12:29.PubMedPubMedCentral Li H, Gao Y, Ren C. Focal adhesion kinase inhibitor BI 853520 inhibits cell proliferation, migration and EMT process through PI3K/AKT/mTOR signaling pathway in ovarian cancer. Discov Oncol. 2021;12:29.PubMedPubMedCentral
195.
Zurück zum Zitat Hirt UA, Waizenegger IC, Schweifer N, Haslinger C, Gerlach D, Braunger J, et al. Efficacy of the highly selective focal adhesion kinase inhibitor BI 853520 in adenocarcinoma xenograft models is linked to a mesenchymal tumor phenotype. Oncogenesis. 2018;7:21.PubMedPubMedCentral Hirt UA, Waizenegger IC, Schweifer N, Haslinger C, Gerlach D, Braunger J, et al. Efficacy of the highly selective focal adhesion kinase inhibitor BI 853520 in adenocarcinoma xenograft models is linked to a mesenchymal tumor phenotype. Oncogenesis. 2018;7:21.PubMedPubMedCentral
196.
Zurück zum Zitat Kanteti R, Mirzapoiazova T, Riehm JJ, Dhanasingh I, Mambetsariev B, Wang J, et al. Focal adhesion kinase a potential therapeutic target for pancreatic cancer and malignant pleural mesothelioma. Cancer Biol Ther. 2018;19:316–27.PubMedPubMedCentral Kanteti R, Mirzapoiazova T, Riehm JJ, Dhanasingh I, Mambetsariev B, Wang J, et al. Focal adhesion kinase a potential therapeutic target for pancreatic cancer and malignant pleural mesothelioma. Cancer Biol Ther. 2018;19:316–27.PubMedPubMedCentral
197.
Zurück zum Zitat Chuang H-H, Wang P-H, Niu S-W, Zhen Y-Y, Huang M-S, Hsiao M, et al. Inhibition of FAK signaling elicits lamin A/C-associated nuclear deformity and cellular senescence. Front Oncol. 2019;9:22.PubMedPubMedCentral Chuang H-H, Wang P-H, Niu S-W, Zhen Y-Y, Huang M-S, Hsiao M, et al. Inhibition of FAK signaling elicits lamin A/C-associated nuclear deformity and cellular senescence. Front Oncol. 2019;9:22.PubMedPubMedCentral
198.
Zurück zum Zitat Golubovskaya VM, Virnig C, Cance WG. TAE226-Induced apoptosis in breast cancer cells with overexpressed Src or EGFR. Mol Carcinog. 2008;47:222–34.PubMed Golubovskaya VM, Virnig C, Cance WG. TAE226-Induced apoptosis in breast cancer cells with overexpressed Src or EGFR. Mol Carcinog. 2008;47:222–34.PubMed
199.
Zurück zum Zitat Kurio N, Shimo T, Fukazawa T, Takaoka M, Okui T, Hassan NMM, et al. Anti-tumor effect in human breast cancer by TAE226, a dual inhibitor for FAK and IGF-IR in vitro and in vivo. Exp Cell Res. 2011;317:1134–46.PubMed Kurio N, Shimo T, Fukazawa T, Takaoka M, Okui T, Hassan NMM, et al. Anti-tumor effect in human breast cancer by TAE226, a dual inhibitor for FAK and IGF-IR in vitro and in vivo. Exp Cell Res. 2011;317:1134–46.PubMed
200.
Zurück zum Zitat Liu T-J, LaFortune T, Honda T, Ohmori O, Hatakeyama S, Meyer T, et al. Inhibition of both focal adhesion kinase and insulin-like growth factor-I receptor kinase suppresses glioma proliferation in vitro and in vivo. Mol Cancer Ther. 2007;6:1357–67.PubMed Liu T-J, LaFortune T, Honda T, Ohmori O, Hatakeyama S, Meyer T, et al. Inhibition of both focal adhesion kinase and insulin-like growth factor-I receptor kinase suppresses glioma proliferation in vitro and in vivo. Mol Cancer Ther. 2007;6:1357–67.PubMed
201.
Zurück zum Zitat Storch K, Sagerer A, Cordes N. Cytotoxic and radiosensitizing effects of FAK targeting in human glioblastoma cells in vitro. Oncol Rep. 2015;33:2009–16.PubMed Storch K, Sagerer A, Cordes N. Cytotoxic and radiosensitizing effects of FAK targeting in human glioblastoma cells in vitro. Oncol Rep. 2015;33:2009–16.PubMed
202.
Zurück zum Zitat Wang ZG, Fukazawa T, Nishikawa T, Watanabe N, Sakurama K, Motoki 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;20:1473–7.PubMed Wang ZG, Fukazawa T, Nishikawa T, Watanabe N, Sakurama K, Motoki 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;20:1473–7.PubMed
203.
Zurück zum Zitat Watanabe N, Takaoka M, Sakurama K, Tomono Y, Hatakeyama S, Ohmori O, et al. Dual tyrosine kinase inhibitor for focal adhesion kinase and insulin-like growth factor-I receptor exhibits anticancer effect in esophageal adenocarcinoma in vitro and in vivo. Clin Cancer Res. 2008;14:4631–9.PubMed Watanabe N, Takaoka M, Sakurama K, Tomono Y, Hatakeyama S, Ohmori O, et al. Dual tyrosine kinase inhibitor for focal adhesion kinase and insulin-like growth factor-I receptor exhibits anticancer effect in esophageal adenocarcinoma in vitro and in vivo. Clin Cancer Res. 2008;14:4631–9.PubMed
204.
Zurück zum Zitat Golubovskaya VM, Figel S, Ho BT, Johnson CP, Yemma M, Huang G, et al. A small molecule focal adhesion kinase (FAK) inhibitor, targeting Y397 site: 1-(2-hydroxyethyl)-3, 5, 7-triaza-1-azoniatricyclo [3.3.1.1(3,7)]decane; bromide effectively inhibits FAK autophosphorylation activity and decreases cancer cell viability, clonogenicity and tumor growth in vivo. Carcinogenesis. 2012;33:1004–13.PubMedPubMedCentral Golubovskaya VM, Figel S, Ho BT, Johnson CP, Yemma M, Huang G, et al. A small molecule focal adhesion kinase (FAK) inhibitor, targeting Y397 site: 1-(2-hydroxyethyl)-3, 5, 7-triaza-1-azoniatricyclo [3.3.1.1(3,7)]decane; bromide effectively inhibits FAK autophosphorylation activity and decreases cancer cell viability, clonogenicity and tumor growth in vivo. Carcinogenesis. 2012;33:1004–13.PubMedPubMedCentral
205.
Zurück zum Zitat Haemmerle M, Bottsford-Miller J, Pradeep S, Taylor ML, Choi H-J, Hansen JM, et al. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal. J Clin Invest. 2016;126:1885–96.PubMedPubMedCentral Haemmerle M, Bottsford-Miller J, Pradeep S, Taylor ML, Choi H-J, Hansen JM, et al. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal. J Clin Invest. 2016;126:1885–96.PubMedPubMedCentral
206.
Zurück zum Zitat Tanjoni I, Walsh C, Uryu S, Tomar A, Nam J-O, Mielgo A, et al. PND-1186 FAK inhibitor selectively promotes tumor cell apoptosis in three-dimensional environments. Cancer Biol Ther. 2010;9:764–77.PubMed Tanjoni I, Walsh C, Uryu S, Tomar A, Nam J-O, Mielgo A, et al. PND-1186 FAK inhibitor selectively promotes tumor cell apoptosis in three-dimensional environments. Cancer Biol Ther. 2010;9:764–77.PubMed
207.
Zurück zum Zitat Golubovskaya VM, Nyberg C, Zheng M, Kweh F, Magis A, Ostrov D, et al. A small molecule inhibitor, 1,2,4,5-benzenetetraamine tetrahydrochloride, targeting the y397 site of focal adhesion kinase decreases tumor growth. J Med Chem. 2008;51:7405–16.PubMedPubMedCentral Golubovskaya VM, Nyberg C, Zheng M, Kweh F, Magis A, Ostrov D, et al. A small molecule inhibitor, 1,2,4,5-benzenetetraamine tetrahydrochloride, targeting the y397 site of focal adhesion kinase decreases tumor growth. J Med Chem. 2008;51:7405–16.PubMedPubMedCentral
208.
Zurück zum Zitat Zhang H, Shao H, Golubovskaya VM, Chen H, Cance W, Adjei AA, et al. Efficacy of focal adhesion kinase inhibition in non-small cell lung cancer with oncogenically activated MAPK pathways. Br J Cancer. 2016;115:203–11.PubMedPubMedCentral Zhang H, Shao H, Golubovskaya VM, Chen H, Cance W, Adjei AA, et al. Efficacy of focal adhesion kinase inhibition in non-small cell lung cancer with oncogenically activated MAPK pathways. Br J Cancer. 2016;115:203–11.PubMedPubMedCentral
209.
Zurück zum Zitat Kurenova EV, Hunt DL, He D, Magis AT, Ostrov DA, Cance WG. Small molecule chloropyramine hydrochloride (C4) targets the binding site of focal adhesion kinase and vascular endothelial growth factor receptor 3 and suppresses breast cancer growth in vivo. J Med Chem. 2009;52:4716–24.PubMedPubMedCentral Kurenova EV, Hunt DL, He D, Magis AT, Ostrov DA, Cance WG. Small molecule chloropyramine hydrochloride (C4) targets the binding site of focal adhesion kinase and vascular endothelial growth factor receptor 3 and suppresses breast cancer growth in vivo. J Med Chem. 2009;52:4716–24.PubMedPubMedCentral
210.
Zurück zum Zitat Golubovskaya VM, Ho B, Zheng M, Magis A, Ostrov D, Morrison C, et al. Disruption of focal adhesion kinase and p53 interaction with small molecule compound R2 reactivated p53 and blocked tumor growth. BMC Cancer. 2013;13:342.PubMedPubMedCentral Golubovskaya VM, Ho B, Zheng M, Magis A, Ostrov D, Morrison C, et al. Disruption of focal adhesion kinase and p53 interaction with small molecule compound R2 reactivated p53 and blocked tumor growth. BMC Cancer. 2013;13:342.PubMedPubMedCentral
Metadaten
Titel
Focal adhesion kinase: from biological functions to therapeutic strategies
verfasst von
Ximin Tan
Yuheng Yan
Bin Song
Shuangli Zhu
Qi Mei
Kongming Wu
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00446-7

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.