Next Article in Journal
Acute Glucose Shift Induces the Activation of the NLRP3 Inflammasome in THP-1 Cells
Previous Article in Journal
Downregulated TNF-α Levels after Cryo-Thermal Therapy Drive Tregs Fragility to Promote Long-Term Antitumor Immunity
Previous Article in Special Issue
Expression of p53 Protein Associates with Anti-PD-L1 Treatment Response on Human-Derived Xenograft Model of GATA3/CR5/6-Negative Recurrent Nonmuscular Invasive Bladder Urothelial Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Targeting of the Gas6/Axl Signaling Pathway in Cancer

Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(18), 9953; https://doi.org/10.3390/ijms22189953
Submission received: 24 August 2021 / Revised: 9 September 2021 / Accepted: 12 September 2021 / Published: 15 September 2021
(This article belongs to the Special Issue Cell Signaling in Cancer Therapy)

Abstract

:
Many signaling pathways are dysregulated in cancer cells and the host tumor microenvironment. Aberrant receptor tyrosine kinase (RTK) pathways promote cancer development, progression, and metastasis. Hence, numerous therapeutic interventions targeting RTKs have been actively pursued. Axl is an RTK that belongs to the Tyro3, Axl, MerTK (TAM) subfamily. Axl binds to a high affinity ligand growth arrest specific 6 (Gas6) that belongs to the vitamin K-dependent family of proteins. The Gas6/Axl signaling pathway has been implicated to promote progression, metastasis, immune evasion, and therapeutic resistance in many cancer types. Therapeutic agents targeting Gas6 and Axl have been developed, and promising results have been observed in both preclinical and clinical settings when such agents are used alone or in combination therapy. This review examines the current state of therapeutics targeting the Gas6/Axl pathway in cancer and discusses Gas6- and Axl-targeting agents that have been evaluated preclinically and clinically.

1. Introduction

Receptor tyrosine kinases (RTKs) are cell surface receptors that mediate a number of physiological responses and homeostasis. However, gene amplification, overexpression, and activating mutations of RTKs are often associated with cancer development, progression, and metastasis and have served as pharmacological targets in cancer treatment. Axl belongs to the TAM (Tyro3, Axl, MerTK) subfamily of the RTKs. Physiologically, the Gas6/Axl pathway plays an important role in platelet aggregation and vessel integrity [1,2,3]. Axl knockout in germ cells does not result in embryonic lethality [4]. The growth arrest specific 6 (Gas6) protein belongs to the vitamin K-dependent family of proteins and is a high affinity ligand for Axl. Overexpression and activation of Axl are widely observed in various cancer types and have been implicated in multiple steps of cancer pathogenesis. In addition, high Axl expression and activation are associated with poor prognosis, outcome, and resistance to therapy in cancer patients [5,6,7,8,9,10,11]. As such, the Gas6/Axl pathway has gained attention as a promising therapeutic target for drug development in multiple tumor types. In this review, we present an overview of the Gas6/Axl signaling pathway and highlight the Gas6 and Axl inhibitors that are being investigated preclinically and clinically.

2. Gas6/Axl Pathway in Cancer

Axl was first isolated from chronic myelogenous leukemia cells in 1988 [12]. Structurally, Axl contains two immunoglobulin-like (IgL) domains, two fibronectin III (FNIII) domains, a transmembrane domain, and an intracellular kinase domain [13]. The Axl pathway can be activated by its ligand Gas6, which belongs to the vitamin K-dependent family of proteins. Gas6 contains the γ-carboxyglutamic acid (Gla) domain, four epidermal growth factor (EGF)-like domains, and two laminin G-like (LG) domains [14,15]. The N-terminus Gla domain mediates binding to phosphatidylserine expressed on cell membranes, apoptotic cells, and debris [16,17]. Gas6 and Axl interact via the first LG-like domain of Gas6 and the two IgL domains of the Axl ectodomain [18]. Gas6 and Axl bind at a one-to-one receptor-to-ligand ratio and then dimerize with another Gas6–Axl complex to initiate a downstream signaling cascade.
The role of Gas6 and Axl in cancer has been reviewed previously [19,20,21,22]. Briefly, Axl is overexpressed in many cancer types and is associated with therapeutic resistance, poor clinical prognosis, and worse outcome [23,24,25,26]. Axl also mediates key components of the metastatic cascade, including, but not limited to, epithelial-to-mesenchymal transition, migration and invasion, proliferation, survival, stemness, angiogenesis, and immune evasion [19,20,22,27] (Figure 1). A study by Zdzalik-Bielecka and colleagues demonstrated that the Gas6/Axl pathway mediates actin cytoskeleton remodeling for cell migration and invasion via formations of peripheral ruffles and circular dorsal ruffles [28]. Such Gas6-induced Axl activation contributes to focal adhesion turnover, cell spreading, and elongation through the activation of PI3K and RAC1 [28,29]. Another study similarly demonstrated that Axl mediates cell invasion through the regulation of lysosome peripheral distribution [30].
In the tumor immune microenvironment, Axl signaling in tumor cells polarizes tumor-associated macrophages toward the M2 phenotype via the Axl/Pi3K/Akt/NF-kB pathway [31]. Axl inhibition elicits a favorable immune response, characterized by a decreased infiltration of CD206+ macrophages and neutrophils and an increased infiltration of CD45+ immune cells, CD8+ T cells, CD4+ T cells, and I-A/I-E+ macrophages [32].
Additionally, it is becoming increasingly clear that the Gas6/Axl signaling axis also impacts non-neoplastic cell populations, which may be of particular relevance when viewed in the context of the tumor microenvironment. For example, pericyte FAK expression is a negative regulator of tumor angiogenesis and tumor growth [33]. In FAK-depleted pericytes, activated Pyk2 increases Gas6 transcript levels and signals via the Gas6/Axl/Akt/Cyr61 pathway to promote tumor cell proliferation and angiogenesis [34,35]. Axl is also expressed by bone marrow-derived cells, dendritic cells, monocytes, natural killer cells, and platelets [36,37,38,39,40,41,42]. A study by Tirado-Gonzalez and colleagues also demonstrated that Axl-expressing leukemia-associated macrophages contribute to immune suppression and impair the functions of NK- and T-cell-mediated tumor cell killing [43].

3. Therapeutic Targeting of Gas6

3.1. Warfarin

Warfarin is a vitamin K antagonist that inhibits the enzyme vitamin K epoxide reductase to recycle inactive vitamin K epoxide to its active form. Warfarin blocks vitamin K-dependent gamma-carboxylation of the γ-carboxyglutamic acid-rich (Gla) domain of Gas6 [44], which prevents the Gas6 interaction with externalized phosphatidylserine on the surface of apoptotic cells and debris (Table 1). Preclinically, the ability of warfarin to impair Gas6 function has been investigated in pancreatic cancer, lung cancer, melanoma, and breast cancer models [42,45,46,47]. Zweemer and colleagues demonstrated that warfarin disrupts the phosphatidyl serine (PS)–Gas6 interaction and consequently impairs tumor cell migration [47]. In addition, Paolino and colleagues demonstrated that warfarin inhibits Gas6-mediated activation of TAM receptors on NK cells and exerts anti-metastatic activity [42]. While an early Phase I study to evaluate the effect of warfarin on markers of the Axl pathway was initiated in 2019, the study was withdrawn in 2021 due to lack of accrual (Clinical Trial Identification #: NCT03536208).

3.2. Soluble Receptors

3.2.1. MYD1/MYD1-72

MYD1 is an Axl decoy receptor with a high affinity for Gas6 (Table 1) [48]. MYD1 contains the major site of Axl Ig1 with four mutations to improve binding to Gas6 [48]. MYD1 inhibited Gas6-mediated Axl-, Akt-, and Erk-phosphorylation and decreased the number of peritoneal metastases in an ovarian cancer model [48].
MYD1-72 is a second-generation Axl decoy receptor that has a higher binding affinity than that of MYD1 (KD: 420 fM vs. 93 fM, respectively). In preclinical studies, MYD1-72 decreased Axl phosphorylation and impaired tumor growth and metastatic burden in vivo. A combination of MYD1-72 with gemcitabine significantly improved the survival of mice with pancreatic cancer to 57 days compared to 17 days in the control and MYD1-72 alone groups and 35 days in the gemcitabine group. A clinical evaluation of this compound has not been reported.

3.2.2. AVB-500

AVB-500 is an Axl decoy receptor with an affinity of 150 fM for Gas6 (Table 1). AVB-500 has been preclinically investigated in renal cell carcinoma and ovarian cancer. The results showed that treatment with AVB-500 decreases Gas6-induced Axl and Src phosphorylation, tumor vessel density, tumor growth, and metastatic burden [49,50,51].
AVB-500 is undergoing Phase I/II clinical trials for patients with platinum-resistant or recurrent ovarian, fallopian tube, or peritoneal cancers as a combination therapy (Clinical Trial Identification #s: NCT03639246, NCT04019288). Three other studies are currently recruiting patients for ovarian cancer, renal cell carcinoma, and pancreatic adenocarcinoma to evaluate its efficacy as a combination therapy with paclitaxel, cabozantinib, and nab-paclitaxel/gemcitabine, respectively (Clinical Trial Identification #s: NCT04729608, NCT04300140, NCT04983407).

4. Therapeutic Targeting of Axl

4.1. Type I Kinase Inhibitors

Small molecular kinase inhibitors can be classified based on the structure of the protein–inhibitor complex [52]. The N-terminal Asp-Phe-Gly (DFG) motif of the activation loop is conserved and regulates kinase activity. Type I protein kinase inhibitors bind to the active protein kinase conformation or DFG-in conformation.

4.1.1. Bemcentinib (BGB324, R428)

Bemcentinib is a highly specific and selective Axl inhibitor with an IC50 of 14 nM in cellular assays (Table 2) [53,54]. This agent has been widely studied in the laboratory in a variety of cancer models, including breast, prostate, lung, pancreatic, and ovarian cancer, and has been shown to decrease tumor cell migration, invasion, and colony formation in vitro and impair primary tumor growth, immune cell infiltration, and metastasis in vivo [55,56,57,58,59,60,61,62]. Axl inhibition alters the expression patterns of EMT markers, upregulating epithelial markers, such as E-cadherin, and downregulating mesenchymal markers, such as N-cadherin, ZEB1, Snail, Slug, Twist, and MMP9 [63,64,65]. Furthermore, several studies have demonstrated that Axl inhibition reverses the therapeutic resistance of certain chemotherapies. Axl has also been associated with immune evasion, and the systematic treatment of tumor-bearing mice with bemcentinib has led to a reduction in tumor-infiltrating host cells, most notably cells of the myeloid lineage [66].
Bemcentinib entered clinical trials as the first Axl-specific inhibitor and is undergoing Phase I/II clinical trials for melanoma, non-small-cell lung cancer (NSCLC), mesothelioma, acute myeloid leukemia, glioblastoma, and pancreatic adenocarcinoma (Clinical Trial Identification Numbers: NCT02872259; NCT02922777; NCT03184571; NCT03654833; NCT03824080; NCT03965494; NCT03649321). A Phase II clinical trial of bemcentinib in combination with pembrolizumab for patients with triple-negative breast cancer has been completed, but the results are not yet available (Clinical Trial Identification #: NCT03184558).

4.1.2. Amuvatinib (MP-470)

Amuvatinib is a c-Kit/Axl kinase inhibitor (Table 2), with higher selectivity against c-Kit than against Axl (IC50 < 1 μM in cellular assays). Amuvatinib has been studied in models of gastrointestinal stromal tumors, lung cancer, and prostate cancer in the context of overcoming therapeutic resistance [60,67,68]. In the gastrointestinal stromal tumor (GIST) model, Mahadevan and colleagues showed that an imatinib-resistant GIST cell line overexpressed Axl compared to its parental cell line [68]. Treatment with amuvatinib showed synergistic, cytotoxic effects with docetaxel [68]. In an erlotinib-resistant lung cancer model, Axl inhibition by amuvatinib restored sensitivity to erlotinib [67].
Clinically, amuvatinib is undergoing Phase I/II trials for small-cell lung cancer (SCLC) and other unresectable or metastatic solid tumors (Clinical Trial Identification Numbers: NCT01357395; NCT00894894; NCT00881166). In a Phase I, dose-escalation study of patients with advanced solid tumors, amuvatinib was well tolerated up to 1500 mg/day with no report of dose-limiting toxicities; indeed, the study did not reach the maximum tolerated dose [69]. In a Phase Ib study of amuvatinib in combination with standard of care therapies for adults with advanced solid tumors, 12% demonstrated partial response to amuvatinib combined with paclitaxel/carboplatin or carboplatin/etoposide in neuroendocrine, NSCLC, and SCLC tumors [70].

4.1.3. Dubermatinib (TP-0903)

Dubermatinib is an Axl inhibitor that has additional activity against Aurora-A and -B and Janus kinase 2 (JAK2). Dubermatinib has been studied in breast cancer, acute myeloid leukemia, neuroblastoma, and chronic lymphocytic leukemia models (Table 2) [71,72,73,74]. Dubermatinib impairs tumor cell migration, invasion, survival, proliferation, and colony formation and can affect chemotherapeutic agent sensitivity [75,76,77,78]. At the molecular level, it inhibits Axl signaling and Aurora B activation to induce G2/M cell cycle arrest [77]. In CLL B cells, dubermatinib induced apoptosis through the downregulation of Mcl-1, Bcl-2, and XIAP and the upregulation of BIM [72]. In a neuroblastoma model, a phospho-kinase array of dubermatinib-treated cells showed greater than 50% increases in p53 S392 and Chk-2 T68 phosphorylation, in line with a corresponding increase in histone H2A.X phosphorylation [73].
Dubermatinib has been studied as monotherapy and combination therapy. Preclinically, combination strategies with all-trans retinoic acid (ATRA), cisplatin, and VP16 demonstrated synergistic, cytotoxic effects [73]. Aveic and colleagues showed that a dubermatinib/ATRA combination in neuroblastoma cell lines was particularly effective against CD133+ cells, a marker associated with chemo- and radio-resistance [79,80,81]. Phase I/II studies are ongoing for patients with advanced metastatic or progressive solid tumors and AML to determine the dose, safety, pharmacokinetics, and pharmacodynamics of dubermatinib (Clinical Trial Identification Numbers: NCT02729298, NCT04518345, NCT03013998). A Phase I/II clinical trial investigating dubermatinib monotherapy and combination therapy with ibrutinib for patients with chronic lymphocytic leukemia was terminated (Clinical Trial Identification #: NCT03572634).

4.1.4. Crizotinib (XALKORI®, PF-02341066)

Crizotinib is a multitargeted, small molecule tyrosine kinase inhibitor that inhibits c-Met, ALK, Ron, and Axl (IC50-Axl = 294 nM in cell-free assay) (Table 2). Crizotinib has been studied in a variety of cancer types, including NSCLC, gastric carcinoma, glioblastoma, anaplastic large-cell lymphoma, and leiomyosarcoma [82,83,84,85]. With respect to the role of crizotinib on the Axl signaling pathway, Dantas-Barbosa and colleagues showed that crizotinib impaired Axl phosphorylation, cell growth, cell cycle, and colony formation in leiomyosarcoma cells [82].
Crizotinib has been approved by the FDA to treat patients with ALK- or ROS1-positive NSCLC and pediatric patients with relapsed or refractory ALK-positive systemic anaplastic large-cell lymphoma (ALCL) [86,87]. There are 151 registered studies associated with crizotinib in the ClinicalTrials.gov database, and 44.4% (67 studies) are currently recruiting, enrolling by invitation, or active/not recruiting for monotherapy and combination therapy strategies. Acquisition of crizotinib resistance is common [88,89], typically developing within a few years for anaplastic lymphoma kinase-rearranged NSCLC [88]. Crizotinib-resistant NSCLC cells showed overexpression of Axl, epithelial-to-mesenchymal transition, and the acquisition of cancer stem cell-like properties [88,90,91].

4.1.5. Bosutinib (BOSULIF®, SKI-606)

Bosutinib is a second-generation, multitargeted, small molecule tyrosine kinase inhibitor that targets Src, Abl, TGFβ, and Axl (Table 2). Bosutinib has been studied in BCR-ABL-dependent diseases, breast cancer, colorectal cancer, and NSCLC [92]. Bosutinib decreases Gas6-mediated Axl phosphorylation, migration, and invasion [93,94,95]. An immunoblot of bosutinib-treated cells showed decreased phosphorylation of Src pY419, FAK Y576/577/925, Pyk2 Y580, and p130Cas Y410 [93]; expression of vimentin and Slug; and increased expression of E-cadherin [96]. In a preclinical thyroid cancer model using ThrbPV/PPten+/- mice, bosutinib treatment significantly prolonged survival and impaired the development of lung metastases [96].
Bosutinib was approved by the FDA to treat patients with chronic, accelerated, or blast-phase Philadelphia chromosome-positive (Ph+) chronic myelogenous leukemia [97]. Bosutinib monotherapy and combination therapy with chemotherapy and other small molecule inhibitors are also being investigated in patients with breast cancer, glioblastoma, and other advanced solid cancers (Clinical Trial identification #s: NCT00319254, NCT01331291, NCT01001936).

4.1.6. S49076

S49076 is a potent inhibitor of c-Met, as well as Axl, MerTK, and FGFR (Table 2). S49076 has been studied preclinically in gastric, lung, and liver cancer models [98,99] and has been shown to decrease the phosphorylation of Axl in a dose-dependent manner. S49076 impairs tumor cell viability, migration, and colony formation in vitro and tumor growth in vivo [98]. Clémenson and colleagues demonstrated that the mechanism of action of S49076 is dependent upon c-Met signaling dependency [100,101,102], where S49076 targets Aurora B kinase in c-Met-independent cells [99].
In a Phase I open-label study in patients with advanced solid tumors, a recommended once-daily dose of 600 mg was defined [103]. While 81.4% of patients had drug-related adverse events, the majority of the adverse events were grade I–II [103]. In a Phase I/II study in combination with bevacizumab in patients with recurrent glioblastoma, the combination did not improve patient survival outcomes [104]. The recommended Phase II dose was confirmed at 600 mg once daily, and no additional dose-limiting toxicity was observed [105,106].

4.1.7. Sunitinib (SU111248)

Sunitinib is an oral, multitargeted, small molecule receptor tyrosine kinase inhibitor that targets VEGFR2, PDGFRβ, c-Kit, and Axl (Table 2). Sunitinib is approved by the FDA to treat patients with gastrointestinal stromal cancer, advanced RCC, and progressive, well-differentiated pancreatic neuroendocrine tumors [107,108]. However, Axl overexpression is associated with sunitinib resistance in renal cell carcinoma cells [109,110]. In patients with renal cell carcinoma, elevated Axl expression is associated with shorter overall survival [109]. Genetic and pharmacologic Axl inhibition in sunitinib-resistant cell lines demonstrate decreased tumor cell migration, invasion, EMT, and angiogenesis [109].

4.1.8. SNS-314

SNS-314 is an aurora kinase inhibitor that also targets Axl, with an IC50 of 84 nM in a cellular assay (Table 2). SNS-314 impairs colony formation, histone H3 phosphorylation, and tumor growth in vivo [111,112]. In 2007, SNS-314 entered a Phase I study for patients with advanced solid tumors to evaluate safety and tolerability (Clinical Trial Identification Number: NCT00519662). The maximum tolerated dose was not established, and no responses were observed [113].

4.1.9. Other Type I Axl Kinase Inhibitors Undergoing Preclinical Evaluation

NA80x1 is a 3-quinolinecarbonitrile that inhibits Axl phosphorylation, tumor cell migration, and invasion [94]. The IC50 of NA80x1 against Axl is 4.11 μM in a cellular assay and demonstrates inhibitory activity against Src kinase [94,114]. SGI-7079 is a selective Axl inhibitor that reduces Gas6-induced Axl phosphorylation and impairs the growth of mesenchymal NSCLC xenograft tumors [115]. In addition, Axl inhibitor DP-3975 [116,117], a TAM kinase inhibitor UNC569 [118], and MerTK/Flt3 dual inhibitor UNC2025 [119] are also type I Axl inhibitors with promising preclinical results.

4.2. Type II Kinase Inhibitors

Unlike type I protein kinase inhibitors, type II protein kinase inhibitors prefer to bind the inactive, DFG-out conformation.

4.2.1. Cabozantinib (BMS-907351)

Cabozantinib is an oral, multitargeted, small molecule tyrosine kinase inhibitor that targets VEGFR, c-Met, and Axl (Table 3). Cabozantinib has been studied in RCC, esophageal squamous cell carcinoma, and lung cancer. Preclinically, cabozantinib treatment decreased Axl phosphorylation and TGF-β-induced E-cadherin expression, cell viability, migration, and tumor growth [120,121].
Cabozantinib has been approved by the FDA for patients with advanced renal cell carcinoma and sorafenib-resistant hepatocellular carcinoma. In January 2021, combination therapy of cabozantinib with nivolumab was approved as first-line treatment for patients with advanced renal cell carcinoma. The efficacy of the combination therapy versus sunitinib alone was evaluated in CHECKMATE-9ER, a randomized, open-label trial in patients with previously untreated advanced renal cell carcinoma [122] (Clinical Trial Identification: NCT03141177). The trial demonstrated significant improvement in progression-free survival, overall survival, and confirmed overall response rate for patients in the combination therapy arm compared with those who received sunitinib monotherapy [122].

4.2.2. Foretinib (GSK1363089)

Foretinib is an oral, small molecule kinase inhibitor of c-Met, VEGFR, and Axl. Foretinib has been studied in esophageal squamous cell carcinoma, breast cancer, leiomyosarcoma, head and neck cancer, and gastric cancer (Table 3) [11,82,123,124,125]. Foretinib decreases Axl and Akt phosphorylation and impairs tumor cell viability and xenograft tumor growth [11]. In a lapatinib-resistant, HER2-positive, ER-positive breast cancer model, the foretinib-mediated downregulation of Axl restored lapatinib sensitivity [126].
Clinically, foretinib has been studied as a monotherapy and a combination therapy. In a Phase I dose-escalation study in patients with advanced solid tumors, the recommended maximum tolerated dose of foretinib was identified [127] (Clinical Trial Identification #: NCT00742131). Stable disease and partial responses were identified in 22 and 3 cases, respectively [127]. In a Phase I study of foretinib in combination with lapatinib in patients with HER2-positive metastatic breast cancer, no responses or improvement in progression-free survival were observed [123].

4.2.3. Sitravatinib (MGCD516)

Sitravatinib is a multitargeted, small molecule receptor tyrosine kinase inhibitor that inhibits c-Kit, PDGFRα/β, c-Met, and Axl (Table 3). Sitravatinib has been shown to decrease Axl phosphorylation, colony formation, angiogenesis, and proliferation in sunitinib- or axitinib-resistant cells, as well as impairing tumor growth inhibition in vivo [128,129,130]. Du and colleagues demonstrated that sitravatinib alters the immune landscape of tumors by reducing the number of tumor-associated immunosuppressive myeloid cells and increasing the number of CD4+ T cells and proliferating CD8+ T cells [130]. Combination treatment of sitravatinib and PD-1 blockade demonstrated complete remission in 2 out of 14 mice with E0771 tumors [130].
In the clinic, sitravatinib is being investigated for use as monotherapy and combination therapy. A Phase I study is currently ongoing for patients with advanced cancer to evaluate its safety, pharmacokinetic, metabolism, pharmacodynamic, and clinical activity profiles (Clinical Trial Identification #: NCT02219711). In addition, several Phase I/II studies are underway to evaluate sitravatinib in combination with immune checkpoint inhibitors (Clinical Trial Identification #s: NCT03015740, NCT04734262, NCT03666143, NCT02954991).

4.2.4. Glesatinib (MGCD265)

Glesatinib is an oral, multitargeted tyrosine kinase inhibitor that targets c-Met, VEGFR, Ron, Tie-2, and Axl (Table 3). Glesatinib impairs tumor angiogenesis and upregulates the expression of PD-L1 [130]. Du and colleagues demonstrated that combination therapy of glesatinib with anti-PD1 therapy delayed tumor growth [130]. As such, a Phase II study of glesatinib in combination with nivolumab in patients with non-small-cell lung cancer is ongoing (Clinical Trial Identification #: NCT02954991).

4.2.5. Rebastinib (DCC-2036)

Rebastinib is a Bcr-Abl inhibitor that also impairs Src family proteins, c-Met, Tie-2, and Axl (Table 3). In a preclinical study utilizing breast cancer models, rebastinib impaired Axl phosphorylation, colony formation, cell cycle distribution, migration, invasion, and tumor growth and pulmonary metastasis in vivo [131]. Phase I/II studies are ongoing for patients with advanced metastatic or progressive solid tumors, breast cancer, and chronic myeloid leukemia to evaluate its safety, pharmacokinetics, and pharmacodynamics as monotherapy (Clinical Trial Identification Number: NCT00827138) or combination therapy with paclitaxel (NCT03601897, NCT02824575), eribulin (NCT02824575), and carboplatin (NCT03717415).

4.2.6. Merestinib (LY2801653)

Merestinib is an oral, multitarget kinase inhibitor that impairs c-Met, Axl, Ron, and Flt3 (Table 3) [132]. Merestinib impairs cell proliferation, angiogenesis, and tumor growth in vivo [132,133]. Merestinib is being investigated clinically in multiple indications, including advanced or metastatic solid tumor, biliary tract cancer, non-small-cell lung cancer, and chronic myeloid leukemia, as a monotherapy or combination therapy (Clinical Trial Identification #s: NCT02711553, NCT02920996, NCT03125239). A Phase I study of merestinib with or without cetuximab, cisplatin, or gemcitabine identified the recommended dose at 120 mg once daily [134] (Clinical Trial Identification #: NCT01285037). One complete response and three partial responses were observed in patients with cholangiosarcoma [134].

4.2.7. BMS-777607

BMS-777607 is a small molecule kinase inhibitor designed against c-Met, but it also has activity against Axl, Ron, and Tyro3 (Table 3). BMS-777607 has been studied in multiple indications, including prostate cancer, breast cancer, brain cancer (glioma/GBM), and fibrosarcoma [135,136,137,138,139,140]. BMS-777607 inhibits HGF-induced Met-autophosphorylation, colony formation, migration, and invasion [137,138]. In vivo, BMS-777607 impaired KHT sarcoma pulmonary metastases [135]; however, this was not the case for DU-145 and MDA-MB-231-4175-LM2 models [140]. Phase I/II clinical trials have been completed for patients with advanced or metastatic solid tumors, but the results are not yet available (Clinical Trial Identification #s: NCT01721148, NCT00605618).

4.2.8. RXDX-106

RXDX-106 is an oral, small molecule inhibitor targeting the TAM receptors and c-Met (Table 3). RXDX-106 has been characterized in colorectal and breast cancer models and has been shown to impair Axl, Tyro3, MerTK, Akt, and Erk phosphorylation; tumor growth in vivo; and infiltration of leukocytes, macrophages, NK cells, and CD8+ T cells [141]. While an early Phase I study to evaluate RXDX-106 safety, tolerability, pharmacokinetics, pharmacodynamics, and clinical activity was planned, the study was terminated in 2018 (Clinical Trial Identification #: NCT03454243).

4.2.9. Other Type II Axl Kinase Inhibitors Undergoing Preclinical Evaluation

LDC1267 is a selective TAM kinase inhibitor that provides anti-metastatic potential by enhancing NK cell activity in vivo [42]. In addition, NPS-1034 is a c-Met/Axl inhibitor that is efficacious against EGFR-TKI resistant non-small-cell lung cancer cells with c-Met and Axl activation [142].

4.3. Monoclonal Antibodies

4.3.1. 20G7-D9

20G7-D9 has been studied in pancreatic cancer and triple-negative breast cancer models (Table 1) [143,144]. In pancreatic cancer cell lines Panc-1 and BxPC-3, 20G7-D9 reduced Gas6-induced Axl and Akt phosphorylation and impaired tumor cell migration, viability, and tumor growth in vivo [143]. Similarly, in a triple-negative breast cancer model, 20G7-D9 decreased tumor growth in vivo and silenced the Gas6-induced upregulation of EMT markers, including Snail, Slug, Twist, and Zeb1/2 [144].

4.3.2. Axl-107-MMAE

Axl-107-MMAE is an antibody drug conjugate, containing human Axl antibody linked to the microtubule-disrupting agent monomethyl auristatin E (Table 1) [145]. Axl-107-MMAE induces cytotoxicity in vitro and impairs tumor growth in cervical cancer, lung cancer, and melanoma models [145]. In addition, combination treatment with MAPK pathway inhibitors cooperatively inhibited melanoma patient-derived xenograft growth [145]. The clinical safety and efficacy of Axl-107-MMAE are currently being evaluated in a Phase I/II clinical study for patients with advanced solid tumors (Clinical Trial Identification #: NCT02988817).

4.3.3. BA3011

BA3011 is an anti-Axl humanized monoclonal antibody conjugated to monomethyl auristatin E using a cleavable linker (CAB-Axl-ADC) (Table 1). BA3011 is capable of reversibly binding to recombinant Axl and Axl-expressing cells in the tumor microenvironment but not in normal tissues. Preclinical studies demonstrate that BA3011 induces cytotoxicity in vitro and impairs tumor growth in lung, prostate, and pancreatic xenograft models [146]. The clinical safety and efficacy of BA3011 are currently being evaluated in Phase I/II and II clinical studies for patients with solid tumors, NSCLC, and ovarian cancers (Clinical Trial Identification #s: NCT03425279, NCT04681131, NCT04918186).

4.3.4. Other Monoclonal Axl Antibodies Undergoing Preclinical Evaluation

YW327.6S2 is a phage-derived monoclonal antibody that recognizes both human and murine Axl [147]. In NSCLC and breast cancer models, YW327.6S2 impaired xenograft tumor growth, reduced breast cancer metastasis, and enhanced the efficacy of erlotinib as well as chemotherapy when used in combination [147]. Mab173 has been demonstrated to induce Axl degradation and impair tumor cell migration, invasion, apoptosis, and xenograft tumor growth in a Kaposi sarcoma model [148].

4.4. Axl-Specific Chimeric Antigen Receptor (CAR) T Cells

T cells with a chimeric antigen receptor (CAR) consisting of a single-chain variable fragment against Axl have been developed and evaluated preclinically. In a triple-negative breast cancer model, Axl-CAR-T cells inhibited tumor growth and showed association with increased secretion of antitumor cytokines, including TNFα, IFNγ, IL-2, IL-6, and IL-17A (Table 1) [149,150]. Axl-CAR.C7R (Axl-targeting CAR-T cells) that co-express constitutively activated the IL-7 receptor (C7R), showed enhanced activation of Axl-CAR-T cells in vitro, and impaired xenograft tumor growth (Table 1) [151].
In the clinical landscape, CCT301 CAR-T is undergoing a Phase I/II study for patients with renal cell carcinoma to assess safety, tolerability, and anti-tumor activity (Table 1) (Clinical Trial Identification #: NCT03393936). A Phase I study for Axl-CAR-T cell therapy for patients with lung cancer is also being initiated to evaluate safety, tolerability, and preliminary efficacy (Clinical Trial Identification #: NCT03198052).

4.5. Natural Products

4.5.1. Viscum album

Mistletoe extract prepared from Viscum album extract (VAE) has demonstrated anti-Axl activities (Table 1). VAE treatment decreased Axl expression and phosphorylation and impaired colony formation and tumor cell proliferation of naïve cells as well as cisplatin- and erlotinib-resistant cells [152].

4.5.2. Celastrol

Celastrol is a pentacyclic triterpenoid extracted from Tripterygium wilfordii Hook F and Celastrus regelii, L., which has anti-Axl activities in non-small-cell lung cancer cells (Table 1). Celastrol reduces Axl expression and impairs tumor cell migration and proliferation in both naïve and gefitinib-resistant non-small-cell lung cancer cells [153].

4.5.3. Yuanhuadine

Yuanhuadine is a daphnane diterpenoid from the flowers of Daphne genkwa, which has previously shown anti-proliferative effects in human lung cancer cells (Table 1) [154]. Yuanhuadine decreases Axl expression in both naïve and gefitinib- and osimertinib-resistant non-small-cell lung cancer cells [155]. Yuanhuadine suppresses tumor growth in vivo [155] and synergistically inhibits the growth of EGFR-TKI-resistant cells in vitro and in vivo [156].

5. Conclusions

Receptor tyrosine kinases have served as pharmacological targets in cancer treatment due to their frequent dysregulations, including gene amplifications, overexpression, and activating mutations, which promote tumor development, progression, and metastasis. The Gas6/Axl signaling pathway has gained significant attention as a therapeutic target due to its implications in cancer progression, metastasis, and therapeutic resistance [19,20,21]. In addition, studies demonstrate that the Gas6/Axl signaling pathway also modulates the tumor microenvironment [22]. Gas6 is secreted by both cancer and stromal cells [76,157,158,159,160,161,162,163], and studies demonstrate that Axl is also expressed by cancer and stromal cells [22]. Therefore, targeting the Gas6/Axl pathway offers an attractive and promising approach to impair tumor progression and dissemination.
In addition, Axl overexpression is associated with resistance to conventional anticancer treatments, such as chemotherapy and radiotherapy, as well as targeted therapies [60,67,77,109,110,115,126,155,164,165,166,167,168,169,170]. Studies demonstrate that the Axl targeting of resistant tumors can restore sensitivity. Hence, combination therapy with Axl inhibitors offers a rational treatment strategy to overcome therapeutic resistant tumors. Furthermore, Axl inhibition has been shown to modulate the tumor immune microenvironment by multiple mechanisms [22]. Chimeric antigen receptor T cells against Axl have been developed and studied preclinically [149,150,151], and a number of clinical studies are ongoing to evaluate the combination therapy of small molecule Axl inhibitors with immune checkpoint inhibitors. Since Axl is involved in multiple facets of the hallmarks of cancer, results from the ongoing clinical trials will highlight its implications in therapeutic resistance, immune evasion, and metastasis while also demonstrating its safety, tolerability, and efficacy.
Overall, a number of strategies have been employed to target Gas6 and Axl, including small molecule inhibitors, soluble receptors, monoclonal antibodies, CAR-T cell therapy, and natural compounds. These inhibitors have been tested as a monotherapy as well as combination therapies. While targeting the Gas6/Axl pathway has shown promising preclinical and clinical efficacy, targeting Gas6 or Axl as a monotherapy raises a concern about a possible narrow therapeutic index due to its expression and regulation of tumor growth in various cell types of the tumor. The Gas6/Axl pathway has been implicated in drug resistance and immune evasion, and preclinical and clinical studies have demonstrated that combination therapy may exhibit synergistic antitumor activity and drug selectivity. Hence, rational combination approaches and the selection of an appropriate patient population remain a necessity.
Although Axl overexpression is associated with poor clinical prognosis and outcome in multiple cancer types [20], the histological confirmation of Axl expression or activation is often absent in clinical study inclusion criteria. Furthermore, as Gas6 mRNA expression may not predict breast cancer outcomes [171], the assessment of Gas6 protein expression or serum levels, as a potential biomarker for patient response and outcome, is also warranted. Since small molecule tyrosine kinase inhibitors often have multiple targets, results from clinical trials utilizing a monoclonal antibody against Axl (i.e., Axl-107-MMAE and BA301), a chimeric antigen receptor T (CAR-T) cell against Axl (CCT301 CAR-T), and a soluble receptor against Gas6 (ABV-500) are highly anticipated and will offer efficacy and toxicity profiles and guidance for the future development of treatment strategies. Still, preclinical and clinical findings for various Gas6 and Axl inhibitors are favorable and make targeting this axis an attractive and promising approach to impair tumor progression and dissemination.

Author Contributions

Conceptualization, M.T.; investigation, M.T.; resources, D.W.S.; data curation, M.T.; writing—original draft preparation, M.T.; writing—review and editing, D.W.S.; supervision, D.W.S.; funding acquisition, D.W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was in part funded by a grant from the National Institutes of Health (1R01CA197477).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Burstyn-Cohen, T.; Heeb, M.J.; Lemke, G. Lack of protein S in mice causes embryonic lethal coagulopathy and vascular dysgenesis. J. Clin. Investig. 2009, 119, 2942–2953. [Google Scholar] [CrossRef] [Green Version]
  2. Angelillo-Scherrer, A.; de Frutos, P.; Aparicio, C.; Melis, E.; Savi, P.; Lupu, F.; Arnout, J.; Dewerchin, M.; Hoylaerts, M.; Herbert, J.; et al. Deficiency or inhibition of Gas6 causes platelet dysfunction and protects mice against thrombosis. Nat. Med. 2001, 7, 215–221. [Google Scholar] [CrossRef] [Green Version]
  3. Angelillo-Scherrer, A.; Burnier, L.; Flores, N.; Savi, P.; DeMol, M.; Schaeffer, P.; Herbert, J.M.; Lemke, G.; Goff, S.P.; Matsushima, G.K.; et al. Role of Gas6 receptors in platelet signaling during thrombus stabilization and implications for antithrombotic therapy. J. Clin. Investig. 2005, 115, 237–246. [Google Scholar] [CrossRef] [Green Version]
  4. Lu, Q.; Gore, M.; Zhang, Q.; Camenisch, T.; Boast, S.; Casagranda, F.; Lai, C.; Skinner, M.K.; Klein, R.; Matsushima, G.K.; et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 1999, 398, 723–728. [Google Scholar] [CrossRef]
  5. Shikawa, M.; Sonobe, M.; Nakayama, E.; Kobayashi, M.; Kikuchi, R.; Kitamura, J.; Imamura, N.; Date, H. Higher expression of receptor tyrosine kinase Axl, and differential expression of its ligand, Gas6, predict poor survival in lung adenocarcinoma patients. Ann. Surg. Oncol. 2013, 20 (Suppl. S3), S467–S476. [Google Scholar] [CrossRef] [Green Version]
  6. Tanaka, K.; Tokunaga, E.; Inoue, Y.; Yamashita, N.; Saeki, H.; Okano, S.; Kitao, H.; Oki, E.; Oda, Y.; Maehara, Y. Impact of Expression of Vimentin and Axl in Breast Cancer. Clin. Breast Cancer 2016, 16, 520–526.e2. [Google Scholar] [CrossRef]
  7. Lozneanu, L.; Pinciroli, P.; Ciobanu, D.A.; Carcangiu, M.L.; Canevari, S.; Tomassetti, A.; Caruntu, I.D. Computational and Immunohistochemical Analyses Highlight AXL as a Potential Prognostic Marker for Ovarian Cancer Patients. Anticancer Res. 2016, 36, 4155–4163. [Google Scholar] [PubMed]
  8. Cardone, C.; Blauensteiner, B.; Moreno-Viedma, V.; Martini, G.; Simeon, V.; Vitiello, P.P.; Ciardiello, D.; Belli, V.; Matrone, N.; Troiani, T.; et al. AXL is a predictor of poor survival and of resistance to anti-EGFR therapy in RAS wild-type metastatic colorectal cancer. Eur. J. Cancer 2020, 138, 1–10. [Google Scholar] [CrossRef] [PubMed]
  9. Liu, J.; Wang, K.; Yan, Z.; Xia, Y.; Li, J.; Shi, L.; Zou, Q.; Wan, X.; Jiao, B.; Wang, H.; et al. Axl Expression Stratifies Patients with Poor Prognosis after Hepatectomy for Hepatocellular Carcinoma. PLoS ONE 2016, 11, e0154767. [Google Scholar] [CrossRef] [PubMed]
  10. Bottai, G.; Raschioni, C.; Szekely, B.; Di Tommaso, L.; Szasz, A.M.; Losurdo, A.; Gyorffy, B.; Acs, B.; Torrisi, R.; Karachaliou, N.; et al. AXL-associated tumor inflammation as a poor prognostic signature in chemotherapy-treated triple-negative breast cancer patients. NPJ Breast Cancer 2016, 2, 16033. [Google Scholar] [CrossRef] [PubMed]
  11. Hsieh, M.S.; Yang, P.W.; Wong, L.F.; Lee, J.M. The AXL receptor tyrosine kinase is associated with adverse prognosis and distant metastasis in esophageal squamous cell carcinoma. Oncotarget 2016, 7, 36956–36970. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, E.; Hjelle, B.; Bishop, J.M. Transforming genes in chronic myelogenous leukemia. Proc. Natl. Acad. Sci. USA 1988, 85, 1952–1956. [Google Scholar] [CrossRef] [Green Version]
  13. O’Bryan, J.P.; Frye, R.A.; Cogswell, P.C.; Neubauer, A.; Kitch, B.; Prokop, C.; Espinosa, R., 3rd; Le Beau, M.M.; Earp, H.S.; Liu, E.T. Axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase. Mol. Cell. Biol. 1991, 11, 5016–5031. [Google Scholar] [CrossRef] [Green Version]
  14. Bellido-Martin, L.; de Frutos, P.G. Vitamin K-dependent actions of Gas6. Vitam. Horm. 2008, 78, 185–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Manfioletti, G.; Brancolini, C.; Avanzi, G.; Schneider, C. The protein encoded by a growth arrest-specific gene (gas6) is a new member of the vitamin K-dependent proteins related to protein S, a negative coregulator in the blood coagulation cascade. Mol. Cell. Biol. 1993, 13, 4976–4985. [Google Scholar] [CrossRef] [Green Version]
  16. Huang, M.; Rigby, A.C.; Morelli, X.; Grant, M.A.; Huang, G.; Furie, B.; Seaton, B.; Furie, B.C. Structural basis of membrane binding by Gla domains of vitamin K-dependent proteins. Nat. Struct. Biol. 2003, 10, 751–756. [Google Scholar] [CrossRef]
  17. Mark, M.R.; Chen, J.; Hammonds, R.G.; Sadick, M.; Godowsk, P.J. Characterization of Gas6, a member of the superfamily of G domain-containing proteins, as a ligand for Rse and Axl. J. Biol. Chem. 1996, 271, 9785–9789. [Google Scholar] [CrossRef] [Green Version]
  18. Sasaki, T.; Knyazev, P.G.; Clout, N.J.; Cheburkin, Y.; Gohring, W.; Ullrich, A.; Timpl, R.; Hohenester, E. Structural basis for Gas6-Axl signalling. EMBO J. 2006, 25, 80–87. [Google Scholar] [CrossRef] [Green Version]
  19. Axelrod, H.; Pienta, K.J. Axl as a mediator of cellular growth and survival. Oncotarget 2014, 5, 8818–8852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Rankin, E.B.; Giaccia, A.J. The Receptor Tyrosine Kinase AXL in Cancer Progression. Cancers 2016, 8, 103. [Google Scholar] [CrossRef]
  21. Gay, C.M.; Balaji, K.; Byers, L.A. Giving AXL the axe: Targeting AXL in human malignancy. Br. J. Cancer 2017, 116, 415–423. [Google Scholar] [CrossRef] [PubMed]
  22. Tanaka, M.; Siemann, D.W. Gas6/Axl Signaling Pathway in the Tumor Immune Microenvironment. Cancers 2020, 12, 1850. [Google Scholar] [CrossRef] [PubMed]
  23. Hutterer, M.; Knyazev, P.; Abate, A.; Reschke, M.; Maier, H.; Stefanova, N.; Knyazeva, T.; Barbieri, V.; Reindl, M.; Muigg, A.; et al. Axl and growth arrest-specific gene 6 are frequently overexpressed in human gliomas and predict poor prognosis in patients with glioblastoma multiforme. Clin. Cancer Res. 2008, 14, 130–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Shinh, Y.-S.; Lai, C.-Y.; Kao, Y.-R.; Shiah, S.-G.; Chu, Y.-W.; Lee, H.-S.; Wu, C.-W. Expression of Axl in Lung Adenocarcinoma and Correlation with Tumor Progression. Neoplasia 2005, 7, 1058–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gjerdrum, C.; Tiron, C.; Høiby, T.; Stefansson, I.; Haugen, H.; Sandal, T.; Collett, K.; Li, S.; McCormack, E.; Gjertsen, B.T.; et al. Axl is an essential epithelial-to-mesenchymal transition-induced regulator of breast cancer metastasis and patient survival. Proc. Natl. Acad. Sci. USA 2010, 107, 1124–1129. [Google Scholar] [CrossRef] [Green Version]
  26. Gustafsson, A.; Martuszewska, D.; Johansson, M.; Ekman, C.; Hafizi, S.; Ljungberg, B.; Dahlbäck, B. Differential expression of Axl and Gas6 in renal cell carcinoma reflecting tumor advancement and survival. Clin. Cancer Res. 2009, 15, 4742–4749. [Google Scholar] [CrossRef] [Green Version]
  27. Tanaka, M.; Siemann, D.W. Axl signaling is an important mediator of tumor angiogenesis. Oncotarget 2019, 10, 2887–2898. [Google Scholar] [CrossRef] [Green Version]
  28. Zdzalik-Bielecka, D.; Poswiata, A.; Kozik, K.; Jastrzebski, K.; Schink, K.O.; Brewinska-Olchowik, M.; Piwocka, K.; Stenmark, H.; Miaczynska, M. The GAS6-AXL signaling pathway triggers actin remodeling that drives membrane ruffling, macropinocytosis, and cancer-cell invasion. Proc. Natl. Acad. Sci. USA 2021, 118, e2024596118. [Google Scholar] [CrossRef]
  29. Abu-Thuraia, A.; Goyette, M.A.; Boulais, J.; Delliaux, C.; Apcher, C.; Schott, C.; Chidiac, R.; Bagci, H.; Thibault, M.P.; Davidson, D.; et al. AXL confers cell migration and invasion by hijacking a PEAK1-regulated focal adhesion protein network. Nat. Commun. 2020, 11, 3586. [Google Scholar] [CrossRef]
  30. Maacha, S.; Hong, J.; von Lersner, A.; Zijlstra, A.; Belkhiri, A. AXL Mediates Esophageal Adenocarcinoma Cell Invasion through Regulation of Extracellular Acidification and Lysosome Trafficking. Neoplasia 2018, 20, 1008–1022. [Google Scholar] [CrossRef]
  31. Chiu, K.C.; Lee, C.H.; Liu, S.Y.; Chou, Y.T.; Huang, R.Y.; Huang, S.M.; Shieh, Y.S. Polarization of tumor-associated macrophages and Gas6/Axl signaling in oral squamous cell carcinoma. Oral Oncol. 2015, 51, 683–689. [Google Scholar] [CrossRef]
  32. Goyette, M.A.; Elkholi, I.E.; Apcher, C.; Kuasne, H.; Rothlin, C.V.; Muller, W.J.; Richard, D.E.; Park, M.; Gratton, J.P.; Cote, J.F. Targeting Axl favors an antitumorigenic microenvironment that enhances immunotherapy responses by decreasing Hif-1alpha levels. Proc. Natl. Acad. Sci. USA 2021, 118, e2023868118. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, Z.; Xu, X.H.; Hu, J. Role of pericytes in angiogenesis: Focus on cancer angiogenesis and anti-angiogenic therapy. Neoplasma 2016, 63, 173–182. [Google Scholar] [CrossRef] [PubMed]
  34. Lechertier, T.; Reynolds, L.E.; Kim, H.; Pedrosa, A.R.; Gomez-Escudero, J.; Munoz-Felix, J.M.; Batista, S.; Dukinfield, M.; Demircioglu, F.; Wong, P.P.; et al. Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth. Nat. Commun. 2020, 11, 2810. [Google Scholar] [CrossRef]
  35. Collett, G.; Wood, A.; Alexander, M.Y.; Varnum, B.C.; Boot-Handford, R.P.; Ohanian, V.; Ohanian, J.; Fridell, Y.W.; Canfield, A.E. Receptor tyrosine kinase Axl modulates the osteogenic differentiation of pericytes. Circ. Res. 2003, 92, 1123–1129. [Google Scholar] [CrossRef] [Green Version]
  36. Kasikara, C.; Davra, V.; Calianese, D.; Geng, K.; Spires, T.E.; Quigley, M.; Wichroski, M.; Sriram, G.; Suarez-Lopez, L.; Yaffe, M.B.; et al. Pan-TAM Tyrosine Kinase Inhibitor BMS-777607 Enhances Anti-PD-1 mAb Efficacy in a Murine Model of Triple-Negative Breast Cancer. Cancer Res. 2019, 79, 2669–2683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Satomura, K.; Derubeis, A.R.; Fedarko, N.S.; Ibaraki-O’Connor, K.; Kuznetsov, S.A.; Rowe, D.W.; Young, M.F.; Gehron Robey, P. Receptor tyrosine kinase expression in human bone marrow stromal cells. J. Cell. Physiol. 1998, 177, 426–438. [Google Scholar] [CrossRef]
  38. Seitz, H.M.; Camenisch, T.D.; Lemke, G.; Earp, H.S.; Matsushima, G.K. Macrophages and dendritic cells use different Axl/Mertk/Tyro3 receptors in clearance of apoptotic cells. J. Immunol. 2007, 178, 5635–5642. [Google Scholar] [CrossRef]
  39. Subramanian, M.; Hayes, C.D.; Thome, J.J.; Thorp, E.; Matsushima, G.K.; Herz, J.; Farber, D.L.; Liu, K.; Lakshmana, M.; Tabas, I. An AXL/LRP-1/RANBP9 complex mediates DC efferocytosis and antigen cross-presentation in vivo. J. Clin. Investig. 2014, 124, 1296–1308. [Google Scholar] [CrossRef] [Green Version]
  40. Deng, T.; Zhang, Y.; Chen, Q.; Yan, K.; Han, D. Toll-like receptor-mediated inhibition of Gas6 and ProS expression facilitates inflammatory cytokine production in mouse macrophages. Immunology 2012, 135, 40–50. [Google Scholar] [CrossRef]
  41. Neubauer, A.; Fiebeler, A.; Graham, D.K.; O’Bryan, J.P.; Schmidt, C.A.; Barckow, P.; Serke, S.; Siegert, W.; Snodgrass, H.R.; Huhn, D.; et al. Expression of axl, a transforming receptor tyrosine kinase, in normal and malignant hematopoiesis. Blood 1994, 84, 1931–1941. [Google Scholar] [CrossRef] [Green Version]
  42. Paolino, M.; Choidas, A.; Wallner, S.; Pranjic, B.; Uribesalgo, I.; Loeser, S.; Jamieson, A.M.; Langdon, W.Y.; Ikeda, F.; Fededa, J.P.; et al. The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells. Nature 2014, 507, 508–512. [Google Scholar] [CrossRef] [PubMed]
  43. Tirado-Gonzalez, I.; Descot, A.; Soetopo, D.; Nevmerzhitskaya, A.; Schaffer, A.; Kur, I.M.; Czlonka, E.; Wachtel, C.; Tsoukala, I.; Muller, L.; et al. AXL inhibition in macrophages stimulates host-versus-leukemia immunity and eradicates naive and treatment resistant leukemia. Cancer Discov. 2021. [Google Scholar] [CrossRef]
  44. Lew, E.D.; Oh, J.; Burrola, P.G.; Lax, I.; Zagorska, A.; Traves, P.G.; Schlessinger, J.; Lemke, G. Differential TAM receptor-ligand-phospholipid interactions delimit differential TAM bioactivities. Elife 2014, 3, e03385. [Google Scholar] [CrossRef]
  45. Kirane, A.; Ludwig, K.F.; Sorrelle, N.; Haaland, G.; Sandal, T.; Ranaweera, R.; Toombs, J.E.; Wang, M.; Dineen, S.P.; Micklem, D.; et al. Warfarin Blocks Gas6-Mediated Axl Activation Required for Pancreatic Cancer Epithelial Plasticity and Metastasis. Cancer Res. 2015, 75, 3699–3705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Tsou, W.I.; Nguyen, K.Q.; Calarese, D.A.; Garforth, S.J.; Antes, A.L.; Smirnov, S.V.; Almo, S.C.; Birge, R.B.; Kotenko, S.V. Receptor tyrosine kinases, TYRO3, AXL, and MER, demonstrate distinct patterns and complex regulation of ligand-induced activation. J. Biol. Chem. 2014, 289, 25750–25763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zweemer, A.J.M.; French, C.B.; Mesfin, J.; Gordonov, S.; Meyer, A.S.; Lauffenburger, D.A. Apoptotic Bodies Elicit Gas6-Mediated Migration of AXL-Expressing Tumor Cells. Mol. Cancer Res. 2017, 15, 1656–1666. [Google Scholar] [CrossRef] [Green Version]
  48. Kariolis, M.S.; Miao, Y.R.; Jones, D.S., 2nd; Kapur, S.; Mathews, I.I.; Giaccia, A.J.; Cochran, J.R. An engineered Axl ‘decoy receptor’ effectively silences the Gas6-Axl signaling axis. Nat. Chem. Biol. 2014, 10, 977–983. [Google Scholar] [CrossRef]
  49. Rankin, E.B.; Fuh, K.C.; Taylor, T.E.; Krieg, A.J.; Musser, M.; Yuan, J.; Wei, K.; Kuo, C.J.; Longacre, T.A.; Giaccia, A.J. AXL is an essential factor and therapeutic target for metastatic ovarian cancer. Cancer Res. 2010, 70, 7570–7579. [Google Scholar] [CrossRef] [Green Version]
  50. Rankin, E.B.; Fuh, K.C.; Castellini, L.; Viswanathan, K.; Finger, E.C.; Diep, A.N.; LaGory, E.L.; Kariolis, M.S.; Chan, A.; Lindgren, D.; et al. Direct regulation of GAS6/AXL signaling by HIF promotes renal metastasis through SRC and MET. Proc. Natl. Acad. Sci. USA 2014, 111, 13373–13378. [Google Scholar] [CrossRef] [Green Version]
  51. Xiao, Y.; Zhao, H.; Tian, L.; Nolley, R.; Diep, A.N.; Ernst, A.; Fuh, K.C.; Miao, Y.R.; von Eyben, R.; Leppert, J.T.; et al. S100A10 Is a Critical Mediator of GAS6/AXL-Induced Angiogenesis in Renal Cell Carcinoma. Cancer Res. 2019, 79, 5758–5768. [Google Scholar] [CrossRef] [Green Version]
  52. Roskoski, R., Jr. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol. Res. 2016, 103, 26–48. [Google Scholar] [CrossRef] [PubMed]
  53. Holland, S.J.; Pan, A.; Franci, C.; Hu, Y.; Chang, B.; Li, W.; Duan, M.; Torneros, A.; Yu, J.; Heckrodt, T.J.; et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res. 2010, 70, 1544–1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Vouri, M.; An, Q.; Birt, M.; Pilkington, G.J.; Hafizi, S. Small molecule inhibition of Axl receptor tyrosine kinase potently suppresses multiple malignant properties of glioma cells. Oncotarget 2015, 6, 16183–16197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Goyette, M.A.; Duhamel, S.; Aubert, L.; Pelletier, A.; Savage, P.; Thibault, M.P.; Johnson, R.M.; Carmeliet, P.; Basik, M.; Gaboury, L.; et al. The Receptor Tyrosine Kinase AXL Is Required at Multiple Steps of the Metastatic Cascade during HER2-Positive Breast Cancer Progression. Cell Rep. 2018, 23, 1476–1490. [Google Scholar] [CrossRef]
  56. Wang, C.; Jin, H.; Wang, N.; Fan, S.; Wang, Y.; Zhang, Y.; Wei, L.; Tao, X.; Gu, D.; Zhao, F.; et al. Gas6/Axl Axis Contributes to Chemoresistance and Metastasis in Breast Cancer through Akt/GSK-3beta/beta-catenin Signaling. Theranostics 2016, 6, 1205–1219. [Google Scholar] [CrossRef]
  57. Ludwig, K.F.; Du, W.; Sorrelle, N.B.; Wnuk-Lipinska, K.; Topalovski, M.; Toombs, J.E.; Cruz, V.H.; Yabuuchi, S.; Rajeshkumar, N.V.; Maitra, A.; et al. Small-Molecule Inhibition of Axl Targets Tumor Immune Suppression and Enhances Chemotherapy in Pancreatic Cancer. Cancer Res. 2018, 78, 246–255. [Google Scholar] [CrossRef] [Green Version]
  58. D’Errico, G.; Alonso-Nocelo, M.; Vallespinos, M.; Hermann, P.C.; Alcala, S.; Garcia, C.P.; Martin-Hijano, L.; Valle, S.; Earl, J.; Cassiano, C.; et al. Tumor-associated macrophage-secreted 14-3-3zeta signals via AXL to promote pancreatic cancer chemoresistance. Oncogene 2019, 38, 5469–5485. [Google Scholar] [CrossRef]
  59. Antony, J.; Zanini, E.; Kelly, Z.; Tan, T.Z.; Karali, E.; Alomary, M.; Jung, Y.; Nixon, K.; Cunnea, P.; Fotopoulou, C.; et al. The tumour suppressor OPCML promotes AXL inactivation by the phosphatase PTPRG in ovarian cancer. EMBO Rep. 2018, 19, e45670. [Google Scholar] [CrossRef]
  60. Lin, J.Z.; Wang, Z.J.; De, W.; Zheng, M.; Xu, W.Z.; Wu, H.F.; Armstrong, A.; Zhu, J.G. Targeting AXL overcomes resistance to docetaxel therapy in advanced prostate cancer. Oncotarget 2017, 8, 41064–41077. [Google Scholar] [CrossRef]
  61. Axelrod, H.D.; Valkenburg, K.C.; Amend, S.R.; Hicks, J.L.; Parsana, P.; Torga, G.; DeMarzo, A.M.; Pienta, K.J. AXL Is a Putative Tumor Suppressor and Dormancy Regulator in Prostate Cancer. Mol. Cancer Res. 2019, 17, 356–369. [Google Scholar] [CrossRef] [Green Version]
  62. Tanaka, M.; Dykes, S.S.; Siemann, D.W. Inhibition of the Axl pathway impairs breast and prostate cancer metastasis to the bones and bone remodeling. Clin. Exp. Metastasis 2021, 38, 321–335. [Google Scholar] [CrossRef]
  63. Asiedu, M.K.; Beauchamp-Perez, F.D.; Ingle, J.N.; Behrens, M.D.; Radisky, D.C.; Knutson, K.L. AXL induces epithelial-to-mesenchymal transition and regulates the function of breast cancer stem cells. Oncogene 2014, 33, 1316–1324. [Google Scholar] [CrossRef] [Green Version]
  64. Cichon, M.A.; Szentpetery, Z.; Caley, M.P.; Papadakis, E.S.; Mackenzie, I.C.; Brennan, C.H.; O’Toole, E.A. The receptor tyrosine kinase Axl regulates cell-cell adhesion and stemness in cutaneous squamous cell carcinoma. Oncogene 2014, 33, 4185–4192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Koorstra, J.B.; Karikari, C.A.; Feldmann, G.; Bisht, S.; Rojas, P.L.; Offerhaus, G.J.; Alvarez, H.; Maitra, A. The Axl receptor tyrosine kinase confers an adverse prognostic influence in pancreatic cancer and represents a new therapeutic target. Cancer Biol. Ther. 2009, 8, 618–626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Guo, Z.; Li, Y.; Zhang, D.; Ma, J. Axl inhibition induces the antitumor immune response which can be further potentiated by PD-1 blockade in the mouse cancer models. Oncotarget 2017, 8, 89761–89774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; Abdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef]
  68. Mahadevan, D.; Cooke, L.; Riley, C.; Swart, R.; Simons, B.; Della Croce, K.; Wisner, L.; Iorio, M.; Shakalya, K.; Garewal, H.; et al. A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors. Oncogene 2007, 26, 3909–3919. [Google Scholar] [CrossRef] [Green Version]
  69. Tibes, R.; Fine, G.; Choy, G.; Redkar, S.; Taverna, P.; Oganesian, A.; Sahai, A.; Azab, M.; Tolcher, A.W. A phase I, first-in-human dose-escalation study of amuvatinib, a multi-targeted tyrosine kinase inhibitor, in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2013, 71, 463–471. [Google Scholar] [CrossRef]
  70. Mita, M.; Gordon, M.; Rosen, L.; Kapoor, N.; Choy, G.; Redkar, S.; Taverna, P.; Oganesian, A.; Sahai, A.; Azab, M.; et al. Phase 1B study of amuvatinib in combination with five standard cancer therapies in adults with advanced solid tumors. Cancer Chemother. Pharmacol. 2014, 74, 195–204. [Google Scholar] [CrossRef]
  71. Patel, V.; Keating, M.J.; Wierda, W.G.; Gandhi, V. Preclinical combination of TP-0903, an AXL inhibitor and B-PAC-1, a procaspase-activating compound with ibrutinib in chronic lymphocytic leukemia. Leuk. Lymphoma 2016, 57, 1494–1497. [Google Scholar] [CrossRef]
  72. Sinha, S.; Boysen, J.; Nelson, M.; Secreto, C.; Warner, S.L.; Bearss, D.J.; Lesnick, C.; Shanafelt, T.D.; Kay, N.E.; Ghosh, A.K. Targeted Axl Inhibition Primes Chronic Lymphocytic Leukemia B Cells to Apoptosis and Shows Synergistic/Additive Effects in Combination with BTK Inhibitors. Clin. Cancer Res. 2015, 21, 2115–2126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Aveic, S.; Corallo, D.; Porcu, E.; Pantile, M.; Boso, D.; Zanon, C.; Viola, G.; Sidarovich, V.; Mariotto, E.; Quattrone, A.; et al. TP-0903 inhibits neuroblastoma cell growth and enhances the sensitivity to conventional chemotherapy. Eur. J. Pharmacol. 2018, 818, 435–448. [Google Scholar] [CrossRef] [PubMed]
  74. Jeon, J.Y.; Buelow, D.R.; Garrison, D.A.; Niu, M.; Eisenmann, E.D.; Huang, K.M.; Zavorka Thomas, M.E.; Weber, R.H.; Whatcott, C.J.; Warner, S.L.; et al. TP-0903 is active in models of drug-resistant acute myeloid leukemia. JCI Insight 2020, 5, e140169. [Google Scholar] [CrossRef] [PubMed]
  75. Sen, T.; Tong, P.; Diao, L.; Li, L.; Fan, Y.; Hoff, J.; Heymach, J.V.; Wang, J.; Byers, L.A. Targeting AXL and mTOR Pathway Overcomes Primary and Acquired Resistance to WEE1 Inhibition in Small-Cell Lung Cancer. Clin. Cancer Res. 2017, 23, 6239–6253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kanzaki, R.; Naito, H.; Kise, K.; Takara, K.; Eino, D.; Minami, M.; Shintani, Y.; Funaki, S.; Kawamura, T.; Kimura, T.; et al. Gas6 derived from cancer-associated fibroblasts promotes migration of Axl-expressing lung cancer cells during chemotherapy. Sci. Rep. 2017, 7, 10613. [Google Scholar] [CrossRef] [Green Version]
  77. Balaji, K.; Vijayaraghavan, S.; Diao, L.; Tong, P.; Fan, Y.; Carey, J.P.; Bui, T.N.; Warner, S.; Heymach, J.V.; Hunt, K.K.; et al. AXL Inhibition Suppresses the DNA Damage Response and Sensitizes Cells to PARP Inhibition in Multiple Cancers. Mol. Cancer Res. 2017, 15, 45–58. [Google Scholar] [CrossRef] [Green Version]
  78. Park, I.K.; Mundy-Bosse, B.; Whitman, S.P.; Zhang, X.; Warner, S.L.; Bearss, D.J.; Blum, W.; Marcucci, G.; Caligiuri, M.A. Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia. Leukemia 2015, 29, 2382–2389. [Google Scholar] [CrossRef]
  79. Chen, K.H.; Hsu, C.C.; Song, W.S.; Huang, C.S.; Tsai, C.C.; Kuo, C.D.; Hsu, H.S.; Tsai, T.H.; Tsai, C.Y.; Woung, L.C.; et al. Celecoxib enhances radiosensitivity in medulloblastoma-derived CD133-positive cells. Childs Nerv. Syst. 2010, 26, 1605–1612. [Google Scholar] [CrossRef]
  80. Blazek, E.R.; Foutch, J.L.; Maki, G. Daoy medulloblastoma cells that express CD133 are radioresistant relative to CD133- cells, and the CD133+ sector is enlarged by hypoxia. Int. J. Radiat. Oncol. Biol. Phys. 2007, 67, 1–5. [Google Scholar] [CrossRef]
  81. Lin, J.; Zhang, X.M.; Yang, J.C.; Ye, Y.B.; Luo, S.Q. gamma-secretase inhibitor-I enhances radiosensitivity of glioblastoma cell lines by depleting CD133+ tumor cells. Arch. Med. Res. 2010, 41, 519–529. [Google Scholar] [CrossRef] [PubMed]
  82. Dantas-Barbosa, C.; Lesluyes, T.; Loarer, F.L.; Chibon, F.; Treilleux, I.; Coindre, J.M.; Meeus, P.; Brahmi, M.; Bally, O.; Ray-Coquard, I.; et al. Expression and role of TYRO3 and AXL as potential therapeutical targets in leiomyosarcoma. Br. J. Cancer 2017, 117, 1787–1797. [Google Scholar] [CrossRef] [Green Version]
  83. Christensen, J.G.; Zou, H.Y.; Arango, M.E.; Li, Q.; Lee, J.H.; McDonnell, S.R.; Yamazaki, S.; Alton, G.R.; Mroczkowski, B.; Los, G. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic large-cell lymphoma. Mol. Cancer Ther. 2007, 6, 3314–3322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Cullinane, C.; Dorow, D.S.; Jackson, S.; Solomon, B.; Bogatyreva, E.; Binns, D.; Young, R.; Arango, M.E.; Christensen, J.G.; McArthur, G.A.; et al. Differential (18)F-FDG and 3’-deoxy-3’-(18)F-fluorothymidine PET responses to pharmacologic inhibition of the c-MET receptor in preclinical tumor models. J. Nucl. Med. 2011, 52, 1261–1267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Zou, H.Y.; Li, Q.; Lee, J.H.; Arango, M.E.; McDonnell, S.R.; Yamazaki, S.; Koudriakova, T.B.; Alton, G.; Cui, J.J.; Kung, P.P.; et al. An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007, 67, 4408–4417. [Google Scholar] [CrossRef] [Green Version]
  86. FDA Approves Crizotinib for Children and Young Adults with Relapsed or Refractory, Systemic Anaplastic Large Cell Lymphoma. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-crizotinib-children-and-young-adults-relapsed-or-refractory-systemic-anaplastic-large (accessed on 18 August 2021).
  87. Highlights of Prescribing Information: XALKORI. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202570s021lbl.pdf (accessed on 18 August 2021).
  88. Dagogo-Jack, I.; Shaw, A.T. Crizotinib resistance: Implications for therapeutic strategies. Ann. Oncol. 2016, 27 (Suppl. S3), iii42–iii50. [Google Scholar] [CrossRef]
  89. Casaluce, F.; Sgambato, A.; Sacco, P.C.; Palazzolo, G.; Maione, P.; Rossi, A.; Ciardiello, F.; Gridelli, C. Resistance to Crizotinib in Advanced Non-Small Cell Lung Cancer (NSCLC) with ALK Rearrangement: Mechanisms, Treatment Strategies and New Targeted Therapies. Curr. Clin. Pharmacol. 2016, 11, 77–87. [Google Scholar] [CrossRef]
  90. Nakamichi, S.; Seike, M.; Miyanaga, A.; Chiba, M.; Zou, F.; Takahashi, A.; Ishikawa, A.; Kunugi, S.; Noro, R.; Kubota, K.; et al. Overcoming drug-tolerant cancer cell subpopulations showing AXL activation and epithelial-mesenchymal transition is critical in conquering ALK-positive lung cancer. Oncotarget 2018, 9, 27242–27255. [Google Scholar] [CrossRef] [Green Version]
  91. Kato, Y.; Ninomiya, K.; Ohashi, K.; Tomida, S.; Makimoto, G.; Watanabe, H.; Kudo, K.; Matsumoto, S.; Umemura, S.; Goto, K.; et al. Combined effect of cabozantinib and gefitinib in crizotinib-resistant lung tumors harboring ROS1 fusions. Cancer Sci. 2018, 109, 3149–3158. [Google Scholar] [CrossRef] [Green Version]
  92. Isfort, S.; Keller-v Amsberg, G.; Schafhausen, P.; Koschmieder, S.; Brummendorf, T.H. Bosutinib: A novel second-generation tyrosine kinase inhibitor. Recent Results Cancer Res. 2014, 201, 81–97. [Google Scholar] [CrossRef]
  93. Vultur, A.; Buettner, R.; Kowolik, C.; Liang, W.; Smith, D.; Boschelli, F.; Jove, R. SKI-606 (bosutinib), a novel Src kinase inhibitor, suppresses migration and invasion of human breast cancer cells. Mol. Cancer Ther. 2008, 7, 1185–1194. [Google Scholar] [CrossRef] [Green Version]
  94. Zhang, Y.X.; Knyazev, P.G.; Cheburkin, Y.V.; Sharma, K.; Knyazev, Y.P.; Orfi, L.; Szabadkai, I.; Daub, H.; Keri, G.; Ullrich, A. AXL is a potential target for therapeutic intervention in breast cancer progression. Cancer Res. 2008, 68, 1905–1915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Tan, D.S.; Haaland, B.; Gan, J.M.; Tham, S.C.; Sinha, I.; Tan, E.H.; Lim, K.H.; Takano, A.; Krisna, S.S.; Thu, M.M.; et al. Bosutinib inhibits migration and invasion via ACK1 in KRAS mutant non-small cell lung cancer. Mol. Cancer 2014, 13, 13. [Google Scholar] [CrossRef] [Green Version]
  96. Kim, W.G.; Guigon, C.J.; Fozzatti, L.; Park, J.W.; Lu, C.; Willingham, M.C.; Cheng, S.Y. SKI-606, an Src inhibitor, reduces tumor growth, invasion, and distant metastasis in a mouse model of thyroid cancer. Clin. Cancer Res. 2012, 18, 1281–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Hochhaus, A.; Gambacorti-Passerini, C.; Abboud, C.; Gjertsen, B.T.; Brummendorf, T.H.; Smith, B.D.; Ernst, T.; Giraldo-Castellano, P.; Olsson-Stromberg, U.; Saussele, S.; et al. Bosutinib for pretreated patients with chronic phase chronic myeloid leukemia: Primary results of the phase 4 BYOND study. Leukemia 2020, 34, 2125–2137. [Google Scholar] [CrossRef]
  98. Burbridge, M.F.; Bossard, C.J.; Saunier, C.; Fejes, I.; Bruno, A.; Leonce, S.; Ferry, G.; Da Violante, G.; Bouzom, F.; Cattan, V.; et al. S49076 is a novel kinase inhibitor of MET, AXL, and FGFR with strong preclinical activity alone and in association with bevacizumab. Mol. Cancer Ther. 2013, 12, 1749–1762. [Google Scholar] [CrossRef] [Green Version]
  99. Clemenson, C.; Chargari, C.; Liu, W.; Mondini, M.; Ferte, C.; Burbridge, M.F.; Cattan, V.; Jacquet-Bescond, A.; Deutsch, E. The MET/AXL/FGFR Inhibitor S49076 Impairs Aurora B Activity and Improves the Antitumor Efficacy of Radiotherapy. Mol. Cancer Ther. 2017, 16, 2107–2119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Pennacchietti, S.; Cazzanti, M.; Bertotti, A.; Rideout, W.M., 3rd; Han, M.; Gyuris, J.; Perera, T.; Comoglio, P.M.; Trusolino, L.; Michieli, P. Microenvironment-derived HGF overcomes genetically determined sensitivity to anti-MET drugs. Cancer Res. 2014, 74, 6598–6609. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, Y.; Xia, M.; Jin, K.; Wang, S.; Wei, H.; Fan, C.; Wu, Y.; Li, X.; Li, X.; Li, G.; et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol. Cancer 2018, 17, 45. [Google Scholar] [CrossRef]
  102. Sharma, S.V.; Settleman, J. Oncogene addiction: Setting the stage for molecularly targeted cancer therapy. Genes Dev. 2007, 21, 3214–3231. [Google Scholar] [CrossRef] [Green Version]
  103. Rodon, J.; Postel-Vinay, S.; Hollebecque, A.; Nuciforo, P.; Azaro, A.; Cattan, V.; Marfai, L.; Sudey, I.; Brendel, K.; Delmas, A.; et al. First-in-human phase I study of oral S49076, a unique MET/AXL/FGFR inhibitor, in advanced solid tumours. Eur. J. Cancer 2017, 81, 142–150. [Google Scholar] [CrossRef]
  104. Hoang-Xuan, K.; Hottinger, A.; Royer-Perron, L.; Alentorn, A.; Savatovsky, J.; Micheli, R.D.; Homicsko, K.; Banquet, S.; Pauly, J.; Sudey, I.; et al. Phase I/II study of S49076, a multi-target inhibitor of c-MET, AXL, FGFR in combination with bevacizumab in patients with recurrent glioblastoma. J. Clin. Oncol. 2016, 34, 2033. [Google Scholar] [CrossRef]
  105. Anonymized Synopsis for CL1-49076-002. Available online: https://clinicaltrials.servier.com/wp-content/uploads/CL1-49076-002-anonymised-synopsis.pdf (accessed on 12 August 2021).
  106. Study That Tested the Combination of 2 Medicinesfor People with Glioblastoma, a Kind of Brain Cancer: A New Drug Called S49076 with a Marketed Drug Called Bevacizumab. Available online: https://clinicaltrials.servier.com/wp-content/uploads/CL1-49076-002-lay-summary.pdf (accessed on 12 August 2021).
  107. Blumenthal, G.M.; Cortazar, P.; Zhang, J.J.; Tang, S.; Sridhara, R.; Murgo, A.; Justice, R.; Pazdur, R. FDA approval summary: Sunitinib for the treatment of progressive well-differentiated locally advanced or metastatic pancreatic neuroendocrine tumors. Oncologist 2012, 17, 1108–1113. [Google Scholar] [CrossRef] [Green Version]
  108. Gyawali, B.; Goldstein, D.A. The US Food and Drug Administration’s Approval of Adjuvant Sunitinib for Renal Cell Cancer: A Case of Regulatory Capture? JAMA Oncol. 2018, 4, 623–624. [Google Scholar] [CrossRef]
  109. Zhou, L.; Liu, X.D.; Sun, M.; Zhang, X.; German, P.; Bai, S.; Ding, Z.; Tannir, N.; Wood, C.G.; Matin, S.F.; et al. Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Oncogene 2016, 35, 2687–2697. [Google Scholar] [CrossRef]
  110. Van der Mijn, J.C.; Broxterman, H.J.; Knol, J.C.; Piersma, S.R.; De Haas, R.R.; Dekker, H.; Pham, T.V.; Van Beusechem, V.W.; Halmos, B.; Mier, J.W.; et al. Sunitinib activates Axl signaling in renal cell cancer. Int. J. Cancer 2016, 138, 3002–3010. [Google Scholar] [CrossRef] [Green Version]
  111. Oslob, J.D.; Romanowski, M.J.; Allen, D.A.; Baskaran, S.; Bui, M.; Elling, R.A.; Flanagan, W.M.; Fung, A.D.; Hanan, E.J.; Harris, S.; et al. Discovery of a potent and selective aurora kinase inhibitor. Bioorg. Med. Chem. Lett. 2008, 18, 4880–4884. [Google Scholar] [CrossRef] [PubMed]
  112. Arbitrario, J.P.; Belmont, B.J.; Evanchik, M.J.; Flanagan, W.M.; Fucini, R.V.; Hansen, S.K.; Harris, S.O.; Hashash, A.; Hoch, U.; Hogan, J.N.; et al. SNS-314, a pan-Aurora kinase inhibitor, shows potent anti-tumor activity and dosing flexibility in vivo. Cancer Chemother. Pharmacol. 2010, 65, 707–717. [Google Scholar] [CrossRef] [PubMed]
  113. Viracta Therapeutics, Inc. FORM 10-K. 31 March 2010. Available online: http://www.getfilings.com/sec-filings/100331/SUNESIS-PHARMACEUTICALS-INC_10-K/#ixzz74TL3YKdu (accessed on 14 August 2021).
  114. Boschelli, D.H.; Wang, Y.D.; Ye, F.; Wu, B.; Zhang, N.; Dutia, M.; Powell, D.W.; Wissner, A.; Arndt, K.; Weber, J.M.; et al. Synthesis and Src kinase inhibitory activity of a series of 4-phenylamino-3-quinolinecarbonitriles. J. Med. Chem. 2001, 44, 822–833. [Google Scholar] [CrossRef] [PubMed]
  115. Byers, L.A.; Diao, L.; Wang, J.; Saintigny, P.; Girard, L.; Peyton, M.; Shen, L.; Fan, Y.; Giri, U.; Tumula, P.K.; et al. An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin. Cancer Res. 2013, 19, 279–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Ou, W.B.; Hubert, C.; Corson, J.M.; Bueno, R.; Flynn, D.L.; Sugarbaker, D.J.; Fletcher, J.A. Targeted inhibition of multiple receptor tyrosine kinases in mesothelioma. Neoplasia 2011, 13, 12–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Ou, W.B.; Corson, J.M.; Flynn, D.L.; Lu, W.P.; Wise, S.C.; Bueno, R.; Sugarbaker, D.J.; Fletcher, J.A. AXL regulates mesothelioma proliferation and invasiveness. Oncogene 2011, 30, 1643–1652. [Google Scholar] [CrossRef] [Green Version]
  118. Christoph, S.; Deryckere, D.; Schlegel, J.; Frazer, J.K.; Batchelor, L.A.; Trakhimets, A.Y.; Sather, S.; Hunter, D.M.; Cummings, C.T.; Liu, J.; et al. UNC569, a novel small-molecule mer inhibitor with efficacy against acute lymphoblastic leukemia in vitro and in vivo. Mol. Cancer Ther. 2013, 12, 2367–2377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Zhang, W.; DeRyckere, D.; Hunter, D.; Liu, J.; Stashko, M.A.; Minson, K.A.; Cummings, C.T.; Lee, M.; Glaros, T.G.; Newton, D.L.; et al. UNC2025, a potent and orally bioavailable MER/FLT3 dual inhibitor. J. Med. Chem. 2014, 57, 7031–7041. [Google Scholar] [CrossRef] [Green Version]
  120. Zhao, Z.; Zhu, X.; Cui, K.; Mancuso, J.; Federley, R.; Fischer, K.; Teng, G.; Mittal, V.; Gao, D.; Zhao, H.; et al. In Vivo Visualization and Characterization of Epithelial-Mesenchymal Transition in Breast Tumors. Cancer Res. 2016, 76, 2094–2104. [Google Scholar] [CrossRef] [Green Version]
  121. Yang, P.W.; Liu, Y.C.; Chang, Y.H.; Lin, C.C.; Huang, P.M.; Hua, K.T.; Lee, J.M.; Hsieh, M.S. Cabozantinib (XL184) and R428 (BGB324) Inhibit the Growth of Esophageal Squamous Cell Carcinoma (ESCC). Front. Oncol. 2019, 9, 1138. [Google Scholar] [CrossRef]
  122. Choueiri, T.K.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Zurawski, B.; Oyervides Juarez, V.M.; Hsieh, J.J.; Basso, U.; Shah, A.Y.; et al. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2021, 384, 829–841. [Google Scholar] [CrossRef] [PubMed]
  123. Chia, S.K.; Ellard, S.L.; Mates, M.; Welch, S.; Mihalcioiu, C.; Miller, W.H., Jr.; Gelmon, K.; Lohrisch, C.; Kumar, V.; Taylor, S.; et al. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res. 2017, 19, 54. [Google Scholar] [CrossRef]
  124. Seiwert, T.; Sarantopoulos, J.; Kallender, H.; McCallum, S.; Keer, H.N.; Blumenschein, G., Jr. Phase II trial of single-agent foretinib (GSK1363089) in patients with recurrent or metastatic squamous cell carcinoma of the head and neck. Investig. New Drugs 2013, 31, 417–424. [Google Scholar] [CrossRef] [Green Version]
  125. Kataoka, Y.; Mukohara, T.; Tomioka, H.; Funakoshi, Y.; Kiyota, N.; Fujiwara, Y.; Yashiro, M.; Hirakawa, K.; Hirai, M.; Minami, H. Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks. Investig. New Drugs 2012, 30, 1352–1360. [Google Scholar] [CrossRef]
  126. Liu, L.; Greger, J.; Shi, H.; Liu, Y.; Greshock, J.; Annan, R.; Halsey, W.; Sathe, G.M.; Martin, A.M.; Gilmer, T.M. Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: Activation of AXL. Cancer Res. 2009, 69, 6871–6878. [Google Scholar] [CrossRef] [Green Version]
  127. Eder, J.P.; Shapiro, G.I.; Appleman, L.J.; Zhu, A.X.; Miles, D.; Keer, H.; Cancilla, B.; Chu, F.; Hitchcock-Bryan, S.; Sherman, L.; et al. A phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2. Clin. Cancer Res. 2010, 16, 3507–3516. [Google Scholar] [CrossRef] [Green Version]
  128. Dolan, M.; Mastri, M.; Tracz, A.; Christensen, J.G.; Chatta, G.; Ebos, J.M.L. Enhanced efficacy of sitravatinib in metastatic models of antiangiogenic therapy resistance. PLoS ONE 2019, 14, e0220101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Patwardhan, P.P.; Ivy, K.S.; Musi, E.; de Stanchina, E.; Schwartz, G.K. Significant blockade of multiple receptor tyrosine kinases by MGCD516 (Sitravatinib), a novel small molecule inhibitor, shows potent anti-tumor activity in preclinical models of sarcoma. Oncotarget 2016, 7, 4093–4109. [Google Scholar] [CrossRef] [Green Version]
  130. Du, W.; Huang, H.; Sorrelle, N.; Brekken, R.A. Sitravatinib potentiates immune checkpoint blockade in refractory cancer models. JCI Insight 2018, 3, e124184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Shen, Y.; Zhang, W.; Liu, J.; He, J.; Cao, R.; Chen, X.; Peng, X.; Xu, H.; Zhao, Q.; Zhong, J.; et al. Therapeutic activity of DCC-2036, a novel tyrosine kinase inhibitor, against triple-negative breast cancer patient-derived xenografts by targeting AXL/MET. Int. J. Cancer 2019, 144, 651–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Yan, S.B.; Peek, V.L.; Ajamie, R.; Buchanan, S.G.; Graff, J.R.; Heidler, S.A.; Hui, Y.H.; Huss, K.L.; Konicek, B.W.; Manro, J.R.; et al. LY2801653 is an orally bioavailable multi-kinase inhibitor with potent activity against MET, MST1R, and other oncoproteins, and displays anti-tumor activities in mouse xenograft models. Investig. New Drugs 2013, 31, 833–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Barat, S.; Bozko, P.; Chen, X.; Scholta, T.; Hanert, F.; Gotze, J.; Malek, N.P.; Wilkens, L.; Plentz, R.R. Targeting c-MET by LY2801653 for treatment of cholangiocarcinoma. Mol. Carcinog. 2016, 55, 2037–2050. [Google Scholar] [CrossRef]
  134. He, A.R.; Cohen, R.B.; Denlinger, C.S.; Sama, A.; Birnbaum, A.; Hwang, J.; Sato, T.; Lewis, N.; Mynderse, M.; Niland, M.; et al. First-in-Human Phase I Study of Merestinib, an Oral Multikinase Inhibitor, in Patients with Advanced Cancer. Oncologist 2019, 24, e930–e942. [Google Scholar] [CrossRef] [Green Version]
  135. Dai, Y.; Bae, K.; Pampo, C.; Siemann, D.W. Impact of the small molecule Met inhibitor BMS-777607 on the metastatic process in a rodent tumor model with constitutive c-Met activation. Clin. Exp. Metastasis 2012, 29, 253–261. [Google Scholar] [CrossRef]
  136. Schroeder, G.M.; An, Y.; Cai, Z.W.; Chen, X.T.; Clark, C.; Cornelius, L.A.; Dai, J.; Gullo-Brown, J.; Gupta, A.; Henley, B.; et al. Discovery of N-(4-(2-amino-3-chloropyridin-4-yloxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl)-2-oxo-1,2-dihydropyridine-3-carboxamide (BMS-777607), a selective and orally efficacious inhibitor of the Met kinase superfamily. J. Med. Chem. 2009, 52, 1251–1254. [Google Scholar] [CrossRef] [PubMed]
  137. Onken, J.; Torka, R.; Korsing, S.; Radke, J.; Krementeskaia, I.; Nieminen, M.; Bai, X.; Ullrich, A.; Heppner, F.; Vajkoczy, P. Inhibiting receptor tyrosine kinase AXL with small molecule inhibitor BMS-777607 reduces glioblastoma growth, migration, and invasion in vitro and in vivo. Oncotarget 2016, 7, 9876–9889. [Google Scholar] [CrossRef] [Green Version]
  138. Dai, Y.; Siemann, D.W. BMS-777607, a small-molecule met kinase inhibitor, suppresses hepatocyte growth factor-stimulated prostate cancer metastatic phenotype in vitro. Mol. Cancer Ther. 2010, 9, 1554–1561. [Google Scholar] [CrossRef] [Green Version]
  139. Sharma, S.; Zeng, J.Y.; Zhuang, C.M.; Zhou, Y.Q.; Yao, H.P.; Hu, X.; Zhang, R.; Wang, M.H. Small-molecule inhibitor BMS-777607 induces breast cancer cell polyploidy with increased resistance to cytotoxic chemotherapy agents. Mol. Cancer Ther. 2013, 12, 725–736. [Google Scholar] [CrossRef] [Green Version]
  140. Hughes, V.S.; Siemann, D.W. Failures in preclinical and clinical trials of c-Met inhibitors: Evaluation of pathway activity as a promising selection criterion. Oncotarget 2019, 10, 184–197. [Google Scholar] [CrossRef]
  141. Yokoyama, Y.; Lew, E.D.; Seelige, R.; Tindall, E.A.; Walsh, C.; Fagan, P.C.; Lee, J.Y.; Nevarez, R.; Oh, J.; Tucker, K.D.; et al. Immuno-oncological Efficacy of RXDX-106, a Novel TAM (TYRO3, AXL, MER) Family Small-Molecule Kinase Inhibitor. Cancer Res. 2019, 79, 1996–2008. [Google Scholar] [CrossRef] [Green Version]
  142. Rho, J.K.; Choi, Y.J.; Kim, S.Y.; Kim, T.W.; Choi, E.K.; Yoon, S.J.; Park, B.M.; Park, E.; Bae, J.H.; Choi, C.M.; et al. MET and AXL inhibitor NPS-1034 exerts efficacy against lung cancer cells resistant to EGFR kinase inhibitors because of MET or AXL activation. Cancer Res. 2014, 74, 253–262. [Google Scholar] [CrossRef] [Green Version]
  143. Leconet, W.; Larbouret, C.; Chardes, T.; Thomas, G.; Neiveyans, M.; Busson, M.; Jarlier, M.; Radosevic-Robin, N.; Pugniere, M.; Bernex, F.; et al. Preclinical validation of AXL receptor as a target for antibody-based pancreatic cancer immunotherapy. Oncogene 2014, 33, 5405–5414. [Google Scholar] [CrossRef] [Green Version]
  144. Leconet, W.; Chentouf, M.; du Manoir, S.; Chevalier, C.; Sirvent, A.; Ait-Arsa, I.; Busson, M.; Jarlier, M.; Radosevic-Robin, N.; Theillet, C.; et al. Therapeutic Activity of Anti-AXL Antibody against Triple-Negative Breast Cancer Patient-Derived Xenografts and Metastasis. Clin. Cancer Res. 2017, 23, 2806–2816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Boshuizen, J.; Koopman, L.A.; Krijgsman, O.; Shahrabi, A.; van den Heuvel, E.G.; Ligtenberg, M.A.; Vredevoogd, D.W.; Kemper, K.; Kuilman, T.; Song, J.Y.; et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat. Med. 2018, 24, 203–212. [Google Scholar] [CrossRef] [PubMed]
  146. Sharp, L.L.; Chang, C.; Frey, G.; Wang, J.; Liu, H.; Xing, C.; Yalcin, S.; Walls, M.; Ben, Y.; Boyle, W.J.; et al. Abstract 827: Anti-tumor efficacy of BA3011, a novel Conditionally Active Biologic (CAB) anti-AXL-ADC. Cancer Res. 2018, 78, 827. [Google Scholar] [CrossRef]
  147. Ye, X.; Li, Y.; Stawicki, S.; Couto, S.; Eastham-Anderson, J.; Kallop, D.; Weimer, R.; Wu, Y.; Pei, L. An anti-Axl monoclonal antibody attenuates xenograft tumor growth and enhances the effect of multiple anticancer therapies. Oncogene 2010, 29, 5254–5264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Liu, R.; Gong, M.; Li, X.; Zhou, Y.; Gao, W.; Tulpule, A.; Chaudhary, P.M.; Jung, J.; Gill, P.S. Induction, regulation, and biologic function of Axl receptor tyrosine kinase in Kaposi sarcoma. Blood 2010, 116, 297–305. [Google Scholar] [CrossRef]
  149. Reppert, S.; Koch, S.; Finotto, S. IL-17A is a central regulator of lung tumor growth. Oncoimmunology 2012, 1, 783–785. [Google Scholar] [CrossRef] [Green Version]
  150. Wei, J.; Sun, H.; Zhang, A.; Wu, X.; Li, Y.; Liu, J.; Duan, Y.; Xiao, F.; Wang, H.; Lv, M.; et al. A novel AXL chimeric antigen receptor endows T cells with anti-tumor effects against triple negative breast cancers. Cell. Immunol. 2018, 331, 49–58. [Google Scholar] [CrossRef]
  151. Zhao, Z.; Li, Y.; Liu, W.; Li, X. Engineered IL-7 Receptor Enhances the Therapeutic Effect of AXL-CAR-T Cells on Triple-Negative Breast Cancer. Biomed. Res. Int. 2020, 2020, 4795171. [Google Scholar] [CrossRef]
  152. Kim, S.; Kim, K.C.; Lee, C. Mistletoe (Viscum album) extract targets Axl to suppress cell proliferation and overcome cisplatin- and erlotinib-resistance in non-small cell lung cancer cells. Phytomedicine 2017, 36, 183–193. [Google Scholar] [CrossRef] [PubMed]
  153. Lee, Y.J.; Kim, S.Y.; Lee, C. Axl is a novel target of celastrol that inhibits cell proliferation and migration, and increases the cytotoxicity of gefitinib in EGFR mutant nonsmall cell lung cancer cells. Mol. Med. Rep. 2019, 19, 3230–3236. [Google Scholar] [CrossRef] [PubMed]
  154. Hong, J.Y.; Chung, H.J.; Lee, H.J.; Park, H.J.; Lee, S.K. Growth inhibition of human lung cancer cells via down-regulation of epidermal growth factor receptor signaling by yuanhuadine, a daphnane diterpene from Daphne genkwa. J. Nat. Prod. 2011, 74, 2102–2108. [Google Scholar] [CrossRef]
  155. Bae, S.Y.; Hong, J.Y.; Lee, H.J.; Park, H.J.; Lee, S.K. Targeting the degradation of AXL receptor tyrosine kinase to overcome resistance in gefitinib-resistant non-small cell lung cancer. Oncotarget 2015, 6, 10146–10160. [Google Scholar] [CrossRef] [Green Version]
  156. Kim, D.; Bach, D.H.; Fan, Y.H.; Luu, T.T.; Hong, J.Y.; Park, H.J.; Lee, S.K. AXL degradation in combination with EGFR-TKI can delay and overcome acquired resistance in human non-small cell lung cancer cells. Cell Death Dis. 2019, 10, 361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Mills, K.L.; Gomes, A.M.; Standlee, C.R.; Rojo, M.D.; Carmeliet, P.; Lin, Z.; Machado, H.L. Gas6 is dispensable for pubertal mammary gland development. PLoS ONE 2018, 13, e0208550. [Google Scholar] [CrossRef] [PubMed]
  158. Shiozawa, Y.; Pedersen, E.A.; Patel, L.R.; Ziegler, A.M.; Havens, A.M.; Jung, Y.; Wang, J.; Zalucha, S.; Loberg, R.D.; Pienta, K.J.; et al. GAS6/AXL axis regulates prostate cancer invasion, proliferation, and survival in the bone marrow niche. Neoplasia 2010, 12, 116–127. [Google Scholar] [CrossRef] [Green Version]
  159. Shiozawa, Y.; Pedersen, E.A.; Taichman, R.S. GAS6/Mer axis regulates the homing and survival of the E2A/PBX1-positive B-cell precursor acute lymphoblastic leukemia in the bone marrow niche. Exp. Hematol. 2010, 38, 132–140. [Google Scholar] [CrossRef] [Green Version]
  160. Khoo, W.H.; Ledergor, G.; Weiner, A.; Roden, D.L.; Terry, R.L.; McDonald, M.M.; Chai, R.C.; De Veirman, K.; Owen, K.L.; Opperman, K.S.; et al. A niche-dependent myeloid transcriptome signature defines dormant myeloma cells. Blood 2019, 134, 30–43. [Google Scholar] [CrossRef]
  161. Bae, C.A.; Ham, I.H.; Oh, H.J.; Lee, D.; Woo, J.; Son, S.Y.; Yoon, J.H.; Lorens, J.B.; Brekken, R.A.; Kim, T.M.; et al. Inhibiting the GAS6/AXL axis suppresses tumor progression by blocking the interaction between cancer-associated fibroblasts and cancer cells in gastric carcinoma. Gastric Cancer 2020, 23, 824–836. [Google Scholar] [CrossRef] [PubMed]
  162. Gomes, A.M.; Carron, E.C.; Mills, K.L.; Dow, A.M.; Gray, Z.; Fecca, C.R.; Lakey, M.A.; Carmeliet, P.; Kittrell, F.; Medina, D.; et al. Stromal Gas6 promotes the progression of premalignant mammary cells. Oncogene 2019, 38, 2437–2450. [Google Scholar] [CrossRef] [PubMed]
  163. Loges, S.; Schmidt, T.; Tjwa, M.; van Geyte, K.; Lievens, D.; Lutgens, E.; Vanhoutte, D.; Borgel, D.; Plaisance, S.; Hoylaerts, M.; et al. Malignant cells fuel tumor growth by educating infiltrating leukocytes to produce the mitogen Gas6. Blood 2010, 115, 2264–2273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Skinner, H.D.; Giri, U.; Yang, L.P.; Kumar, M.; Liu, Y.; Story, M.D.; Pickering, C.R.; Byers, L.A.; Williams, M.D.; Wang, J.; et al. Integrative Analysis Identifies a Novel AXL–PI3 Kinase–PD-L1 Signaling Axis Associated with Radiation Resistance in Head and Neck Cancer. Clin. Cancer Res. 2017, 23, 2713–2722. [Google Scholar] [CrossRef] [Green Version]
  165. Brand, T.M.; Iida, M.; Stein, A.P.; Corrigan, K.L.; Braverman, C.M.; Coan, J.P.; Pearson, H.E.; Bahrar, H.; Fowler, T.L.; Bednarz, B.P.; et al. AXL Is a Logical Molecular Target in Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2015, 21, 2601–2612. [Google Scholar] [CrossRef] [Green Version]
  166. Aguilera, T.A.; Rafat, M.; Castellini, L.; Shehade, H.; Kariolis, M.S.; Hui, A.B.; Stehr, H.; von Eyben, R.; Jiang, D.; Ellies, L.G.; et al. Reprogramming the immunological microenvironment through radiation and targeting Axl. Nat. Commun. 2016, 7, 13898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Hong, C.C.; Lay, J.D.; Huang, J.S.; Cheng, A.L.; Tang, J.L.; Lin, M.T.; Lai, G.M.; Chuang, S.E. Receptor tyrosine kinase AXL is induced by chemotherapy drugs and overexpression of AXL confers drug resistance in acute myeloid leukemia. Cancer Lett. 2008, 268, 314–324. [Google Scholar] [CrossRef]
  168. Kurokawa, M.; Ise, N.; Omi, K.; Goishi, K.; Higashiyama, S. Cisplatin influences acquisition of resistance to molecular-targeted agents through epithelial-mesenchymal transition-like changes. Cancer Sci. 2013, 104, 904–911. [Google Scholar] [CrossRef] [PubMed]
  169. Dufies, M.; Jacquel, A.; Belhacene, N.; Robert, G.; Cluzeau, T.; Luciano, F.; Cassuto, J.P.; Raynaud, S.; Auberger, P. Mechanisms of AXL overexpression and function in Imatinib-resistant chronic myeloid leukemia cells. Oncotarget 2011, 2, 874–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Meyer, A.S.; Miller, M.A.; Gertler, F.B.; Lauffenburger, D.A. The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci. Signal. 2013, 6, ra66. [Google Scholar] [CrossRef] [Green Version]
  171. Ibrahim, T.; Mercatali, L.; Amadori, D. A new emergency in oncology: Bone metastases in breast cancer patients (Review). Oncol. Lett. 2013, 6, 306–310. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The Gas6/Axl pathway mediates multiple steps of the metastatic cascade. Upon Axl binding to its ligand growth arrest specific 6 (Gas6) protein, Axl dimerizes and autophosphorylates its tyrosine residues in the kinase domain. Axl activation regulates downstream signaling, including the JAK/STAT pathway, Ras/MEK/Erk1/2 pathway, and PI3K/Akt pathway. In turn, the Gas6/Axl pathway upregulates pro-tumorigenic functions, such as immune evasion, survival, proliferation, drug resistance, angiogenesis, epithelial-to-mesenchymal transition (EMT), migration, and invasion. Gas6 and Axl are also expressed by stromal cells, including endothelial cells, pericytes, and subsets of immune cells, to promote tumor progression and metastasis.
Figure 1. The Gas6/Axl pathway mediates multiple steps of the metastatic cascade. Upon Axl binding to its ligand growth arrest specific 6 (Gas6) protein, Axl dimerizes and autophosphorylates its tyrosine residues in the kinase domain. Axl activation regulates downstream signaling, including the JAK/STAT pathway, Ras/MEK/Erk1/2 pathway, and PI3K/Akt pathway. In turn, the Gas6/Axl pathway upregulates pro-tumorigenic functions, such as immune evasion, survival, proliferation, drug resistance, angiogenesis, epithelial-to-mesenchymal transition (EMT), migration, and invasion. Gas6 and Axl are also expressed by stromal cells, including endothelial cells, pericytes, and subsets of immune cells, to promote tumor progression and metastasis.
Ijms 22 09953 g001
Table 1. Gas6 and Select Class of Axl Inhibitors.
Table 1. Gas6 and Select Class of Axl Inhibitors.
Inhibitor/DeveloperTargetTypeIndicationsPhaseStrategyStatus
WarfarinGas6Vitamin K agonistPancreatic cancer, lung cancer, melanoma, breast cancerPreclinical/
Phase I
MonotherapyWithdrawn
MYD1/MYD1-72
Stanford University
Gas6Soluble receptorOvarian cancer, pancreatic cancerPreclinicalMonotherapy/
Combination
-
AVB-500
Aravive/Stanford University
Gas6Soluble receptorOvarian, renal cell carcinoma, pancreatic adenocarcinomaPhase I/IIMonotherapy/
Combination
Active, not recruiting/
Recruiting
20G7-D9
Oribase Pharma/French Research Agency of Health/University of Montpellier/Regional Clinical Cancer Center
AxlMonoclonal antibodyPancreatic cancer, breast cancerPreclinicalMonotherapy-
Axl-107-MMAE
Genmab/Oncode Institute, The Netherlands Cancer Institute
AxlAntibody drug conjugateCervical cancer, lung cancer, melanomaPhase I/IIMonotherapyActive, not recruiting
BA3011
BioAtla
AxlAntibody drug conjugateLung cancer, prostate cancer, ovarian cancer, pancreatic cancerPhase I/IIMonotherapy/
Combination
Not yet recruiting/
Recruiting
YW327.6S2
Genentech
AxlMonoclonal antibodyNSCLC and breast cancerPreclinicalCombination-
Mab173
University of Southern California
AxlMonoclonal antibodyKaposi sarcomaPreclinicalMonotherapy-
Axl-CAR-T
Beijing Institute of Radiation Medicine
AxlCAR-T cellsTriple-negative breast cancerPreclinicalMonotherapy-
Axl-CAR.C7R
XinJiang Medical University
AxlCAR-T cellsTriple-negative breast cancerPreclinicalMonotherapy-
CCT301 CAR-T
PerHum Therapeutics
AxlCAR-T cellsRenal cell carcinomaPhase I/IIMonotherapyActive, not recruiting
Axl-CAR-T
Hunan Zhaotai Yongren Medical Innovation/Guangdong Zhaotai InVivo Biomedicine
AxlCAR-T cellsLung cancerPhase IMonotherapyRecruiting
Viscum albumAxlNatural productNSCLCPreclinicalMonotherapy-
CelastrolAxlNatural productNSCLCPreclinicalCombination-
YuanhuadineAxlNatural productNSCLCPreclinicalCombination-
Table 2. Type I kinase inhibitors against Axl.
Table 2. Type I kinase inhibitors against Axl.
Inhibitor/DeveloperTarget(s)IC50 for AxlIndications Approved by the FDA
Crizotinib (PF-02341066)
Pfizer
Axl, Alk,
c-Met, Ron
in vitro:
294 nM
Approved for patients with ALK- or ROS1-positive NSCLC and pediatric ALK-positive anaplastic large-cell lymphoma
Bosutinib (SKI-606)
Pfizer
Axl, Src kinases, Abl, TGF, BMPin vitro:
0.56 μM
cells: 1.65 μM
Approved for patients with chronic, accelerated, or blast phase Philadelphia chromosome chronic myelogenous leukemia
Sunitinib (SU11248)
Pfizer
Axl, Kit, Flt3, PDGFR, VEGFR2in vitro: 9 nMApproved for renal cell carcinoma and imatinib-resistant gastrointestinal stromal tumor and metastatic pancreatic neuroendocrine tumors
Inhibitor/DeveloperTarget(s)IC50 for AxlClinical Trial #PhaseStrategyStatusIndications
Bemcentinib (BGB324, R428)
Rigel Pharmaceuticals/BerGen BIO
Axlin vitro: 14 nM
cells: <30 nM
NCT03965494IMonotherapyRecruitingGlioblastoma
NCT02922777ICombinationRecruitingNSCLC
NCT03649321I/IICombinationRecruitingPancreatic adenocarcinoma
NCT02872259I/IICombinationRecruitingMelanoma
NCT03824080IIMonotherapyActive, not recruitingAcute myeloid leukemia (AML), Myelodysplastic syndrome
NCT03654833IICombinationRecruitingMesothelioma
NCT03184558IICombinationCompletedTNBC, TN-inflammatory breast cancer
NCT03184571IICombinationRecruitingNSCLC
Amuvatinib (MP-470)
Astex Pharmaceuticals
c-Kit, Axl, c-Met, PDGFRα, FLT3, c-Ret, RAD51cells: <1 μMNCT00881166ICombinationCompletedInvasive malignancy except non-melanoma skin cancers or cervical carcinoma in situ
NCT00894894IMonotherapyCompletedUnresectable or metastatic solid tumor
NCT01357395IIMonotherapyCompletedSmall-cell lung cancer (SCLC)
Dubermatinib (TP-0903)
Tolero Pharmaceuticals
Axl, Aurora A and B, JAK2, Alk, Abl, Merin vitro:
27 nM
cells: 222 nM
NCT02729298IMonotherapyActive, not recruitingAdvanced metastatic or progressive solid tumor (Phase 1b: EGFR positive NSCLC; BRAF-, KRAS-, or NRAS-mutated colorectal carcinoma; recurrent ovarian carcinoma; BRAF-mutated melanoma)
NCT03013998I/IIMonotherapy/
Combination
RecruitingAML
NCT04518345I/IIMonotherapyRecruitingAML
NCT03572634I/IICombinationTerminatedChronic lymphocytic leukemia, small lymphocytic lymphoma
S49076
Servier
Met mutants, Axl, Mer, FGFRsin vitro: 7 nM
cells: 56 nM
ISRCTN00759419IMonotherapyCompletedAdvanced solid tumors
ISRCTN11619481I/IICombinationCompletedGBM
SNS-314
Sunesis Pharmaceuticals
Aurora-A, -B, -C, Trk A/B, Flt4, Axl, c-Rafin vitro:
84 nM
NCT00519662IMonotherapyCompletedAdvanced solid tumors
NA80x1
Wyeth-Ayerst Research/
Max Planck Institute of Biochemistry
Axlin vitro:
12.8 μM
cells: 4.11 μM
Preclinical-Monotherapy--
SGI-7079
Northwestern University
Axl, Met, Mer, Yes, Ret, Flt3in vitro:
58 nM
cells: <1 μM
Preclinical-Monotherapy/
Combination
--
DP-3975
Deciphera Pharamaceuticals
Axlcells: 0.1 μMPreclinical-Monotherapy--
UNC569
University of North Carolina
Mer, Tyro3, Axlin vitro:
37 nM
Preclinical-Monotherapy--
UNC2025
University of North Carolina
Mer, Flt3, Axl, Tyro3in vitro:
1.6 nM
Preclinical-Monotherapy--
Bold: preferred target.
Table 3. Type II kinase inhibitors against Axl.
Table 3. Type II kinase inhibitors against Axl.
Inhibitor/
Developer
Target(s)IC50 for AxlIndications Approved by the FDA
Cabozantinib (BMS-907351)
Exelixis
VEGFR2, c-Met, Ret, Tie2, c-Kit, Axlin vitro: 7 nM
cells: 42 nM
Approved for patients with advanced renal cell carcinoma, sorafenib-resistant hepatocellular carcinoma, and combination therapy with nivolumab for advanced renal cell carcinoma
Inhibitor/
Developer
Target(s)IC50 for AxlClinical Trial #PhaseStrategyStatusIndications
Foretinib (GSK1363089)
GlaxoSmithKline
c-Met, VEGFR2, Ron, Axlin vitro: 11 nM
cells: <100 nM
NCT01138384I/IICombinationCompletedInvasive, HER2-positive breast cancer
NCT01147484IIMonotherapyCompletedRecurrent triple-negative breast cancer
NCT00726323IIMonotherapyCompletedRenal cell carcinoma
NCT00725712IIMonotherapyCompletedMetastatic gastric carcinoma
Sitravatinib (MGCD516)
Mirati Therapeutics
c-Kit, PDGFRα/β, Axl, c-Metin vitro:
1.5 nM
cells:
250–800 nM
NCT04123704IIMonotherapyRecruitingMetastatic TNBC
NCT03680521IICombinationActive,
not recruiting
ccRCC
Glesatinib (MGCD265)
Mirati Therapeutics
c-Met, RON, Axl, VEGFRΝ/ANCT02954991IICombinationActive,
not recruiting
NSCLC
Rebastinib (DCC-2036)
Deciphera Pharmaceuticals
Bcr-Abl, Axl, Flt3, VEGFR2, Srcin vitro:
42 nM
NCT02824575ICombinationRecruitingHER2-negative breast cancer
NCT03717415I/IICombinationRecruitingAdvanced or metastatic solid tumor
NCT03601897I/IICombinationRecruitingLocally advanced or metastatic solid tumor
NCT00827138IMonotherapyCompletedCML
Merestinib (LY2801653)
Eli Lilly and Company
c-Met, MST1R, Axl, ROS1, Flt3in vitro:
11 nM
cells: 2 nM
NCT02711553IICombinationActive,
not recruiting
Advanced or metastatic biliary tract cancer
NCT02920996IIMonotherapyActive,
not recruiting
NSCLC
NCT03125239ICombinationCompletedRelapsed or refractory AML
BMS-777607
Aslan Pharmaceuticals
Axl, Ron, c-Met, Tyro3in vitro:
1.1 nM
NCT01721148IMonotherapyCompletedAdvanced or metastatic solid tumor
NCT00605618I/IIMonotherapyCompletedAdvanced or metastatic solid tumor
RXDX-106 (CEP-40783)
Ignyta, Inc.
Axl, Tyro3, Mer, c-Metin vitro: 7 nMNCT03454243IMonotherapyTerminatedLocally advanced or metastatic solid tumor
NPS-1034
Neopharm
c-Met, Axlin vitro:
10 nM
Preclinical-Combination--
LDC1267
Lead Discovery Center
Axl, Tyro3, Mer, c-Met, Srcin vitro:
29 nM
Preclinical-Monotherapy/
Combination
--
Bold: preferred target.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tanaka, M.; Siemann, D.W. Therapeutic Targeting of the Gas6/Axl Signaling Pathway in Cancer. Int. J. Mol. Sci. 2021, 22, 9953. https://doi.org/10.3390/ijms22189953

AMA Style

Tanaka M, Siemann DW. Therapeutic Targeting of the Gas6/Axl Signaling Pathway in Cancer. International Journal of Molecular Sciences. 2021; 22(18):9953. https://doi.org/10.3390/ijms22189953

Chicago/Turabian Style

Tanaka, Mai, and Dietmar W. Siemann. 2021. "Therapeutic Targeting of the Gas6/Axl Signaling Pathway in Cancer" International Journal of Molecular Sciences 22, no. 18: 9953. https://doi.org/10.3390/ijms22189953

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop