Next Article in Journal
iMIL4PATH: A Semi-Supervised Interpretable Approach for Colorectal Whole-Slide Images
Previous Article in Journal
Comprehensive Analysis of Acquired Genetic Variants and Their Prognostic Impact in Systemic Mastocytosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Two-Front War on Cancer—Targeting TAM Receptors in Solid Tumour Therapy

1
Preclinical Development Department, Celon Pharma S.A., Research & Development Centre, Marymoncka 15, 05-152 Kazun Nowy, Poland
2
Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
3
Clinical Development Department, Celon Pharma S.A., Research & Development Centre, Marymoncka 15, 05-152 Kazun Nowy, Poland
*
Authors to whom correspondence should be addressed.
Cancers 2022, 14(10), 2488; https://doi.org/10.3390/cancers14102488
Submission received: 22 April 2022 / Revised: 14 May 2022 / Accepted: 17 May 2022 / Published: 18 May 2022

Abstract

:

Simple Summary

In recent years, many studies have shown the importance of TAM kinases in both normal and neoplastic cells. In this review, we present and discuss the role of the TAM family (AXL, MERTK, TYRO3) of receptor tyrosine kinases (RTKs) as a dual target in cancer, due to their intrinsic roles in tumour cell survival, migration, chemoresistance, and their immunosuppressive roles in the tumour microenvironment. This review presents the potential of TAMs as emerging therapeutic targets in cancer treatment, focusing on the distinct structures of TAM receptor tyrosine kinases. We analyse and compare different strategies of TAM inhibition, for a full perspective of current and future battlefields in the war with cancer.

Abstract

Receptor tyrosine kinases (RTKs) are transmembrane receptors that bind growth factors and cytokines and contain a regulated kinase activity within their cytoplasmic domain. RTKs play an important role in signal transduction in both normal and malignant cells, and their encoding genes belong to the most frequently affected genes in cancer cells. The TAM family proteins (TYRO3, AXL, and MERTK) are involved in diverse biological processes: immune regulation, clearance of apoptotic cells, platelet aggregation, cell proliferation, survival, and migration. Recent studies show that TAMs share overlapping functions in tumorigenesis and suppression of antitumour immunity. MERTK and AXL operate in innate immune cells to suppress inflammatory responses and promote an immunosuppressive tumour microenvironment, while AXL expression correlates with epithelial-to-mesenchymal transition, metastasis, and motility in tumours. Therefore, TAM RTKs represent a dual target in cancer due to their intrinsic roles in tumour cell survival, migration, chemoresistance, and their immunosuppressive roles in the tumour microenvironment (TME). In this review, we discuss the potential of TAMs as emerging therapeutic targets in cancer treatment. We critically assess and compare current approaches to target TAM RTKs in solid tumours and the development of new inhibitors for both extra- and intracellular domains of TAM receptor kinases.

1. Introduction

The TAM receptor family comprises three receptor tyrosine kinases (MERTK, AXL, TYRO3) that play important roles in diverse biological processes in normal cells [1,2,3,4]. The tyrosine kinases MERTK, AXL, and TYRO3 share a typical RTK structure with an extracellular domain (ECD) of two immunoglobulin-related domains (IgL), followed by two fibronectin type III (FNIII), a transmembrane domain, and a tyrosine kinase domain (TKD) on the cytoplasmic side of the membrane [5,6,7]. The human TAMs exhibit 31–36% identical (52–57% similar) amino acids (aa) within the extracellular region and the intracellular domains share 54–59% aa identity (72–75% similarity) within the tyrosine kinase domain [6].
TAM receptors are activated upon binding their extracellular ligands with IgL domains, subsequent receptor dimerisation, cross-autoactivation of TKD, and downstream signal transduction [8,9]. The first natural ligands of TAM receptors, i.e., Growth Arrest-Specific 6 (GAS6) and Protein S (PROS1), were identified in 1995 and to date, they remain the best-known ligands of the TAM family [7,10,11]. GAS6 and PROS1 require vitamin K-dependent γ-carboxylation for maximal activation. The ligands have a distinct affinity for the different TAM receptors: GAS6 binds all TAM receptors, with the highest affinity for AXL, while protein S binds to MERTK and TYRO3. GAS6 and PROS1 can bind the phosphatidylserine (PS), and TAMs become activated when PS is exposed on the apoptotic cells, aggregating platelets, exosomes, or virus envelopes [1,12,13,14].
The activated TAM receptors stimulate many downstream signalling pathways, including the phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) PI3K-Akt-mTOR, the MAPK/ERK kinase (MEK) 1/2-extracellular signal-regulated kinase (ERK) MEK-ERK, p38 MAPK, FAK (focal adhesion kinase), STAT (signal transducer and activator of transcription), and NFκB (nuclear factor-κB), impacting cell proliferation, migration, survival, and epithelial–mesenchymal transition (EMT) [15,16,17,18,19,20].
The TAM RTK are typically not mutated in cancer cells; however, their overexpression has been reported in numerous solid tumours and hematologic malignancies, including non-small cell lung cancer (NSCLC), glioblastoma, melanoma, prostate cancer, breast cancer, acute myeloid leukaemia, and others [21,22,23,24,25,26,27]. The expression of AXL corresponds with a metastatic propensity, invasiveness in vitro, and resistance to targeted therapies [28,29,30,31]. AXL and MERTK proteins have been increasingly implicated in drug resistance to both conventional and targeted therapies [17,32,33,34]. Moreover, the association of EMT with AXL kinase expression is well documented, and EMT in turn is highly correlated with drug resistance [35,36,37]. A growing number of findings show the involvement of the activated TAM kinases in cancer progression [15,21,24,35,38].
The immunoregulatory roles of MERTK RTK were demonstrated in single- or multiple TAM knockout mice where a lack of these receptors resulted in the development of a severe lymphoproliferative disorder along with autoimmunity [39]. The most-studied effects of TAM knockout have been in the immune system, where signalling from the receptors couples the clearance of cell debris with the negative regulation of the innate immune system [40,41,42]. One of the striking features of TAM single, double, and triple knockout mice were connected with autoimmunity and impaired apoptotic cell clearance. What is worth mentioning is that the inhibition of more than one TAM family kinase resulted in a more severe phenotype [39,43,44].
Both MERTK and AXL enhance the immunosuppressive nature of the tumour niche, as they are expressed on macrophages, NK cells, and dendritic cells [45,46,47,48]. It has been shown that the TAM receptors expressed in the tumour microenvironment play a role in the phagocytosis of apoptotic cells, differentiation of NK cells, the function of T cells, and the secretion of cytokines [24,46,49,50,51].
In recent years, TAM kinases have become an important therapeutic target in cancer therapy (Figure 1). Due to the similarities in the structure of the three TAM receptors, it is difficult to develop inhibitors specific to a single TAM RTK. Therefore, there are currently three main strategies implemented to inhibit TAM receptor activity and signal transduction in carcinogenesis: (1) inhibition of ligand–receptor complex formation; (2) decoy receptors in a soluble form designed to form inactive TAM complexes; and (3) small-molecule tyrosine kinase inhibitors (TKIs). In this review, we discuss how these strategies are applied to battle carcinogenesis, how successful they are, and provide support for the further drug development of TAM-targeting therapies.

2. TAM Family in Carcinogenesis

The classical activation of RTKs involves ligand binding to the extracellular domain of the protein [52]. Subsequently, ligand binding causes receptor dimerisation and the subsequent autophosphorylation of tyrosine residues within the cytoplasmic domain [53]. Recent studies demonstrated that each TAM receptor has a distinct pattern of activation by GAS6 and PROS1, and their interactions may be affected by the presence of apoptotic cells and PS-containing lipid vesicles [9]. Tsou et al. showed that the γ-carboxylation of ligands was required for the full activation of TAMs and soluble immunoglobulin-like TAM domains could act as specific ligand antagonists [9]. Although TYRO3, AXL, and MERTK share sequence similarity, they have distinct functions in the immunoregulation and the recognition/removal of apoptotic cells [9]. In two models of TAM-dependent homeostatic phagocytosis, MERTK played a dominant role, while AXL was dispensable; the activation of MERTK by PROS1 was sufficient to drive phagocytosis [54].
Ligand binding induces receptor dimerisation and subsequent trans-autophosphorylation of tyrosine residues within the cytoplasmic domain and activation of adaptor signalling proteins, which results in the initiation of the signalling cascade and phosphorylation of the downstream targets [55]. TAM family members share three conserved tyrosine residues within the activation loop in the kinase domain. Those sites are required for proper kinase enzymatic activity, and in the human, the conserved tyrosines are as follows: Tyr698, Tyr702, Tyr703 in AXL, Tyr749, Tyr753, and Tyr754 in MERTK, and Tyr681, Tyr685, and Tyr686 in TYRO3 [56,57]. Other phosphorylation sites of TAM proteins are positioned in the distal part of the cytoplasmic domain: Tyr779, Tyr821, and Tyr866 in AXL, Tyr847, Tyr872, and Tyr929 in MERTK, and Tyr762, Tyr804, and Tyr828 in TYRO3 [58,59].
In addition to ligand-dependent activation, the receptor-independent activation of TAM kinases also occurs and encompasses receptor-independent homodimerisation or aggregation of extracellular AXL domains on neighbouring cells [60,61,62]. Heterodimerisation between TAM family proteins has also been reported [63,64,65]. Moreover, some researchers have described heterodimerisation with non-TAM family receptors, such as EGFR or HER3, which activates AXL-associated signalling [28,66].
Various factors regulate the activity and expression of TAM kinases at different levels [67]. TAM kinases can be upregulated or downregulated at the transcriptional level, among others, through the action of cytokines. Post-transcriptional regulation also occurs via micro-RNAs, for example, miR-34a and miR-199a/b regulate AXL expression [68,69]. At the protein level, a metalloproteinase A disintegrin, and metalloprotease (ADAM)10 or ADAM17, may shed the extracellular domain of these proteins [70,71,72]. In addition, soluble forms of TAM receptors can inhibit the activity of these kinases by acting as a decoy receptor for ligands and preventing kinase stimulation by, for instance, GAS6 [73,74]. Although TAM family kinases are frequently overexpressed and activated in various types of cancer, genetic changes within their encoding genes are rather rare. Due to impaired phagocytosis, known relevant mutations in rodents and humans in MERTK genes could lead to retinitis pigmentosa [75,76]. In cancer, mutations, fusions, or amplifications in the TAM coding genes are not very common. However, the AXL aberrations have been identified in 3% or less of solid cancers (breast cancer, lung cancers, head, and neck cancer) and acute myeloid leukaemia [77,78].

3. Extracellular Domain: An Approach from the Outside

3.1. Ligand Binding/Dimerisation Inhibition

GAS6 and PROS1 are vitamin K-dependent proteins and share ~44% similar structural homology [79]. The general structure of GAS6 and PROS1 consists of a gamma-carboxy glutamic acid (Gla) domain at the N-terminus, then four epidermal growth factor-like (EGF-like) repeats, and at the C-terminus, a sex hormone-binding globulin (SHBG) domain made of two laminin G-like (LG) domains [80,81]. The Gla domain in the presence of vitamin K is γ-carboxylated and, in this form, recognises phosphatidylserine presented on the surface of apoptotic cells, which next forms a bridge between TAM receptors and an apoptotic cell [9]. The LG domains within the carboxy-terminal SHBG domains of the ligands are recognised and bound by IgL domains in TAM ECD. Upon ligand binding, the dimerisation of receptors occurs and is mediated by membrane-proximal fibronectin type III (FNIII) domains [8].
Although GAS6 and PROS1 are quite structurally similar, the functional differences and the distinct affinities of TAM receptors for these ligands are well established. GAS6 can bind to any of the TAM receptors, with the highest affinity for AXL, then TYRO3 and MERTK [9,54,82]. Interestingly, GAS6 has two binding sites reported—the major one is recognised exclusively by the AXL protein, whereas the minor GAS6 binding site is recognised by MERTK and TYRO3 [8]. This selectivity results from a β-sheet formation of charged and neutral residues within the major binding site of GAS6 to opposite faces of the newly formed β-sheet [8]. PROS1 presents a different binding profile, with a preference for TYRO3 and MERTK over AXL [83,84,85]. GAS6 and PROS1 could also be recognised by TAMs in dimeric forms, as there are reports of the formation of heterodimers GAS6-PROS1 and ligand multimerisation required for TAM receptor activation in certain scenarios [86,87].
A fine balance of ligand interaction with a tyrosine kinase receptor is necessary to maintain normal tyrosine kinase function without causing overactivation, which could result in human disease [88]. GAS6 is the most important ligand in anticancer therapy targeting TAM receptors. It is expressed in many human tissues and different types of cancers, and by binding to its three receptors (AXL, MERTK, TYRO3) plays a role in biological processes, i.e., proliferation, apoptosis, migration, and survival [89]. Notably, GAS6 expression within solid and non-solid tumours often correlates with poor prognosis [90,91,92,93]. Interestingly, this protein also plays a role in the TME [47,94,95]. References to overexpression of PROS1 in neoplasms are, so far, only single reports. PROS1 is known for its involvement in coagulation processes due to the thrombin-sensitive region within its structure, correlating with the blood coagulation cascade [96,97].
Antagonistic antibodies are a common strategy used to inhibit TAM signalling, due to their high specificity and versatility in blocking receptor–ligand complex formation. Monoclonal antibodies (mAbs) have been employed to stop not only simple ligand binding, but also TAM downstream activity. The formation of the antibody–receptor complex might also lead to the blockage of the receptor dimerisation, which may further result in receptor destabilisation and subsequent degradation. An immunological aspect of antibodies cannot be overlooked, as it may lead to the death of a cancer cell. Several mAbs have been designed for ECD of AXL, with proven activity against TAM receptors in vitro and in vivo. One, Tilvestamab, significantly inhibits AXL activation and tumour growth in mice [98] and is currently being tested in clinical trials (NCT04893551). Other antibodies did not reach this stage, but still present proven activity against TAM receptors in vitro and in vivo. Antibodies YW327.6S2, MAb173, D9/E8, 20G7-D9, 3G9/8B5/12A11/4F8 mAbs, and DAXL-88 block GAS6 binding to the receptors and reduce TAM transcription levels, with prominent inhibition of tumour growth, cell migration, and invasion in several cancer cell lines: SKOV3 (ovarian cancer), A549 (non-small cell lung cancer), and MDA-MB-231 (triple-negative breast cancer) [99,100,101,102,103]. Notably, there was an attempt to create a tetravalent bispecific IgG-scFv antibody format, combining anti-AXL CDX-0168 and anti-PD-L1 mAb (9H9) using an IgG-scFv format. In vitro results proved that this construct inhibited both PD-L1 and AXL signalling, as well as improved cytokine release and T-cell activation [104]. CDX-0168/9H9 was not further investigated and did not move to the clinical stage.
The antibody hTyro3-IgG against TYRO3 has been reported to induce drug sensitivity in primary colon cancer cell cultures and mouse xenografts [105]. RGX-019 humanised monoclonal antibody promotes MERTK receptor internalisation. MERTK signalling pathway inhibition reduced cancer cell viability and induced cytokine expression in the immune-suppressive M2 macrophages. The RGX-019 antibody also showed a good profile in toxicity studies, as it did not reveal retinal toxicity, a common undesirable effect of MERTK inhibition [106].
Although only one of these antibodies reached the clinical stage of development, antibody–drug conjugates (ADC) were more successful in this field and brought a definitive cytotoxic effect to highly specific anti-AXL antibodies. Two different ADCs are currently in clinical trials. Mecbotamab vedotin (CAB-AXL-ADC) is in the I/II clinical phase in patients with advanced solid tumours in phase 1 and BA3011 alone, and with a PD-1 inhibitor in phase II (clinical trials NCT03425279 and NCT04681131). Enapotamab vedotin (HuMax®-AXL-ADC) has shown some clinical activity in phase II for solid tumours, but was discontinued in 2020, as the data gathered during the trials did not meet the desired criteria (NCT02988817).

3.2. Receptor Cleavage and Decoy Receptors

RTKs in certain cellular conditions can be cleaved by ADAM10/17 to release the kinase domain into the cytoplasm [70,71,72]. TAM’s proteolytic cleavage is increased by lipopolysaccharide (LPS), phorbol 12-myristate 13-acetate (PMA), reactive oxygen species (ROS), and other environmental factors [71]. This mechanism can be exploited in cancer treatment, as TAM ECD in the soluble form is rendered dysfunctional, therefore it cannot transduce signal downstream; moreover, it may inhibit intact TAM receptor signalling by interacting with TAM ligands to limit their accessibility.
Interestingly, while soluble forms of TYRO3 and AXL effectively blocked both PROS1 and GAS6 signalling, respectively, soluble MERTK showed weak inhibitory activities against both ligands [9]. Still, the soluble form of MERTK (sMer) was reported to inhibit macrophage clearance of apoptotic cells and platelet aggregation [73], leading to the inhibition of apoptotic neutrophil clearance [107].
Targeting the AXL receptor domain directly is the leading strategy in this field, with a few engineered decoy receptors in development [108,109] and the most advanced Batiraxcept (formerly AVB-500) in the lead. Batiraxcept inhibits GAS6/AXL signalling in vivo and shows an 80-fold greater affinity to GAS6 than the natural receptor. Batiraxcept construct carries four point mutations (Asp87Gly, Val92Ala, Gly32Ser, and Gly127Arg), allowing the decoy receptor to trap the GAS6 ligand by both minor and major binding sites in a heterobivalent manner [110]. Moreover, Batiraxcept demonstrated a favourable safety profile in clinical trials and is now being tested for platinum-resistant ovarian cancer treatment in combination with paclitaxel in a phase III clinical trial (NCT04729608).

3.3. Low-Molecular-Weight Compounds Targeting ECD of TAMs

An alternative approach to inhibit TAMs with small non-biological compounds was also tested. RU-301 and RU-302 are low-molecular-weight (LMWs) compounds that bind to Ig1 domains of TAM extracellular domains, blocking ligand binding. Both compounds showed good inhibitory effects in low micromolar IC50s, with the activation of both TAMs being inhibited in cultured cancer cells and tumour growth in lung cancer xenograft models [111]. The inhibitory parameters might be lower than the tyrosine kinase inhibitors described in the next chapter, but RU-301 showed much higher specificity against TAM receptors. Kinase profiling revealed that RU-301 is much more specific and presents less off-target activities than R428, a strong AXL TKI with 14 nM IC50 against AXL [111]. AXL activity is also suppressed by a well-established anti-coagulant—warfarin. Low dosages of warfarin prevent the progression and spread of pancreatic cancer [112]. In this scenario, warfarin inhibits the activation of vitamin K by epoxide reductase complex 1 (VKORC1) and, by proxy, blocks the activation of GAS6, which is a vitamin K-dependent protein [113]. Consequently, an induced apoptosis of cancer cells was observed, and reduced migration, proliferation, and improved sensitivity to chemotherapy were revealed [112].

4. Kinase Domain: An Approach from the Inside

Tyrosine Kinase Inhibitors

The intracellular kinase domain of TAM receptors is considered a very promising therapeutic target in cancer therapy. Kinase domains are very conservative in their structure, which allows the wide use of bioinformatic approaches in developing small-molecule inhibitors. The intracellular domains of TAM receptors share 54–59% sequence identity and very high (72–75%) sequence similarity [6,114]. The homology is also represented in the overall structure of TAM kinase domains, with very similar topography and global conformation.
Crystal structures of MERTK/AXL/TYRO3 kinase domains show that all share a consensus KW(I/L)A(I/L)ES sequence; however, MERTK and AXL are structurally closer to each other than to TYRO3 [115]. AXL and MERTK form very similar pockets in ATP-binding sites, while the TYRO3 kinase domain has a bigger pocket that accommodates larger molecules [115]. Crucial amino acids for ATP binding have been identified in MERTK and are conserved within TYRO3 and AXL. These crucial residues are as follows: Leu593, Gly594, Val601, Ala617, Lys619, Leu671, Pro672, Phe673, Met674, Asp678, Arg727, Asn728, Met730, and Asp741 in MERTK. There are also two important substitutions reported: Ile650Ala in TYRO3 and Ile650Met in AXL [115]. It has been shown that Ile650Met introduced in MERTK results in the formation of a mimic AXL active site, whereas substitution Ile650Ala does not mimic the TYRO3 catalytic site due to larger structural differences in the TYRO3 catalytic pocket, where Ile650A forms a unique subpocket near the ATP binding site [115,116]. All of these structural differences mean that bigger inhibitors bind better to TYRO3, while smaller ones have a lower affinity towards TYRO3, with low selectivity between MERTK and AXL due to the high similarity of the two domains. This effect is visible in the IC50 values of different TAM TKIs (Table 1). The computational analysis supports this notion, as a large inhibitor compound designed for MERTK/AXL presented a higher affinity towards TYRO3 than the kinases it was targeting [116]. These subtle differences can be exploited to improve selectivity between TAM family members, as an overall conserved structure of the kinase domain results in difficulties in creating the potent and selective inhibitors.
Typically, the kinase domain consists of a β-strand N-lobe, and an α-helical C-lobe connected with a hinge. Kinase domain (KD) activity is regulated by the conformational state of the aspartate–phenylalanine–glycine (DFG) motif in the hinge region. In its inactive state, also called DFG-out, phenylalanine disrupts the orientation of the aspartate, effectively inhibiting Mg2+ binding and sterically blocking the ATP binding site. Upon activation, a trans-autophosphorylation of tyrosine residues occurs with conformational changes leading to the reorientation of phenylalanine and positioning of aspartate for two magnesium ions coordination, opening the hinge into a DFG-in state [152]. This mechanism is exploited in small-molecule inhibitor design, as well as an amphiphilic environment of the kinase domain active site, with polar and nonpolar residue clusters within the pocket. The highly conserved kinase structure of the kinase domain makes it difficult to design and develop new small-molecule inhibitors that are both highly selective and potent [115]. Nevertheless, properly implemented drug design can use this specific topography to create an intricate network of interactions to design specific inhibitors, which is crucial in the development of inhibitors for so closely related proteins as TAM family RTKs.
As growing information is gathered on kinase domain structure and its conformational states, several strategies for KD inhibition have been proposed. Currently, there are six different classes of inhibitors, representing different approaches to kinase domain inhibition. Type I inhibitors interact with the ATP-binding site in the active DFG motif (DFG-in) state competitively with ATP. Type II inhibitors occupy the same area, but in a DFG-out state, keeping the kinase domain inactive [116]. Type I/II inhibitors are the most advanced TKIs in development, but are limited by highly conserved KD structure, which often results in the low selectivity and high toxicity of these inhibitors. Both classes are also susceptible to acquired drug resistance through mutations affecting the gatekeeper residues. New-generation type II inhibitors are designed to penetrate an allosteric pocket to overcome drug resistance [152]. Type III inhibitors target specific allosteric sites within the catalytic site, while type IV inhibitors target other allosteric sites. Type III/IV inhibitors are highly selective, as they target specific regulatory sites of enzymes, e.g., phosphorylation sites [153]. Type V inhibitors use protein scaffolds for bivalent binding of KDs’ active sites and other important sites, like the structural motifs or regulatory sites. Type V inhibitors are potent tools in research, but their applications in cancer treatment are limited by the large size of protein scaffolds, which restrict their availability to RTKs’ intracellular KD domains. Type VI inhibitors target active kinase sites and bonds in a covalent way. Different and stable binding lead to high potency and reduced toxicity in comparison to type I and II inhibitors [154,155]. Type VI inhibitors are also more resistant to acquired drug resistance via gatekeeping mutations within ATP-binding sites [155].
In the case of the TAM family, the TKIs of type I and type II are the most prominent and well-studied [156]. These compounds can be aggregated in two distinct structural groups with similar cores, and some non-conventional compounds. First, the CORE-A group consists of smaller compounds, with a hydrogen bond acceptor–substituted phenyl group linked to a hinge-binding heterocycle with a solubilising group (Figure 2). These compounds (SGI7079, bosutinib, gilteritinib, dubermatinib, vandetanib) are all type I inhibitors and were often designed primarily for AXL and present high inhibition of AXL in the low-nM range, with similar inhibition levels of MERTK (Table 1). CORE-A inhibitors are very successful, with three of these compounds (gilteritinib, bosutinib, and vandetanib) developed beyond the preclinical stage with successful clinical trials, and are now approved for treatment in different types of cancer [157].
The second group of compounds, i.e., CORE-B, presents a more complex structure: heterocycle (hinge-binding) optionally ortho-fluoro phenyl group (DFG-binding), 2–4 hydrogen donors/acceptors, and a phenyl group (or para-fluoro phenyl) that binds to allosteric hydrophobic pockets (Figure 2). This scheme is prominent among type II inhibitors, as these compounds are generally bigger and cannot properly dock ATP-binding sites in a DFG-in state [158,159]. This class is developing rapidly, with several compounds in various clinical trial phases (BMS777607, DS-1205c, foretinib, MGCD265, merestinib, ONO-7475, PF-07265807, sitravatinib) and one, cabozantinib, approved for treatment. Several more compounds are yet to be tested for efficacy in clinical trials (Table 1).
As more TAM TKIs are developed, new compounds are introduced, presenting partial structural similarity to previously described cores, -A and -B (rebastinib, amuvatinib, SNS314), or present completely different lead structures (crizotinib, sunitinib, S49076, UN1062, UNC2025). Those compounds are mostly type I inhibitors, the exception being type II rebastinib. These unique cores open new ways for the rational design of TKIs [158,159], as most successful compounds here were designed for different kinases (e.g., crizotinib for ALK/c-Met/ROS1, foretinib for MET/VEGFRs and suntinib for VEGFR2/PDGFRs), but their robust cores are potent against other receptor kinases, TAMs included (Figure 2).

5. Battles for the Future

5.1. Selectivity of TAM Family Inhibitors

As discussed above, a rational approach to developing selective inhibitors within the TAM family is challenging, due to the high structural homology around the active catalytic site. In particular, AXL and MERTK are similar—their ATP-binding site is smaller compared to the more open TYRO3 pocket [115]. Consequently, it is easier to obtain dual AXL/MERTK inhibitors that show selectivity over TYRO3 [115]. When acting on more than one target, there should always be broad consideration of what this might entail—what the concerns might be versus the potential benefits.
Importantly, targeting closely related kinases may involve resistance caused by so-called bypass signalling [77,78]. For example, AXL can cause resistance to MERTK and MERTK can cause resistance to AXL [160]. Given this consideration, in some cases, the simultaneous targeting of both of these kinases may be more effective than the selective inhibition of either kinase. McDaniel et al. showed that the expression of MERTK kinase is increased after treatment with AXL inhibitors in cancer cell lines and patient-derived xenografts [160]. MERTK inhibition resulted in increased sensitivity of head and neck squamous cell carcinoma (HNSCC), triple-negative breast cancer (TNBC), and non-small-cell lung carcinoma (NSCLC) cell lines to AXL inhibition. When both kinases, AXL and MER, were targeted, it caused more robust inhibition of downstream signalling and impaired tumour cell expansion in vitro, as well as reduced tumour growth in vivo [160].
In NSCLC, AXL and MER may have overlapping and complementary roles; furthermore, their role in resistance to therapies and co-occurrence in the tumour microenvironment may outweigh the benefits that the development of such a concept may bring [17]. The inhibition of more than one kinase from the TAM family may be beneficial in some aspects, as described above, but on the other hand, it may also result in increased adverse effects, i.e., inflammation. AXL is often overexpressed in TNBC with a mesenchymal phenotype and in some colon cancer subtypes [161,162,163]. In contrast, MERTK kinase seems to be more abundantly expressed in acute lymphoblastic leukaemias, therefore a selective MERTK inhibition could be an interesting therapeutic strategy in this context [164,165,166,167].

5.2. Combination Therapy

Combination therapies are implemented in cancer treatment for several reasons. First, they increase the effectiveness of the therapy and improve treatment outcomes, particularly when synergistic anticancer effects are achieved. Second, reduced emerging resistance to therapy is observed, which is a key and limiting problem. Studies are being conducted on combining therapies targeting TAM receptors and classical therapies such as radio- or chemotherapy, as well as targeted therapies and immune-checkpoint inhibitors. There is a growing body of knowledge linking TAM kinases to resistance to chemotherapy and radiotherapy in both solid and hematologic malignancies [168,169]. Moreover, in the case of therapies targeting, e.g., EGFR in NSCLC, TAM receptors are known to mediate bypass signalling, resulting in resistance to EGFR inhibitors such as erlotinib, gefitinib, and osimertinib [36,170,171,172]. Moreover, kinases of this family, through their physiological functions in the immune system, can also modulate resistance [50,173,174]. Several TAM inhibitors that are still in development, besides their direct antitumour activity, are known for their immunomodulatory effects on the tumour microenvironment and enhancement of the antitumour immunity [133,141]. Therefore, there are ongoing clinical trials combining TAM TKIs and immune checkpoint inhibitors such as pembrolizumab and nivolumab [175,176,177,178]. Combining TAM-targeted inhibitors with classical, targeted, or immunotherapies seems reasonable to obtain more effective anticancer treatment strategies.

5.3. Biomarkers

Despite the frequent overexpression of TAM proteins in cancer, genetic mutations or gene amplification are rare [77,179]. Due to the lack of confirmed activating mutations or amplifications that could be a suitable biomarker of therapeutic response, extensive work is being done to find robust biomarkers. Finding the appropriate biomarkers is one of the challenges in developing therapies targeting TAM kinases that could lead to clinically successful treatment strategies. Other options include the expression of the TAM family proteins, their phosphorylation and activation status. Moreover, the level of ligands (GAS6 in particular) and soluble forms of the receptors are taken into account as potential biomarkers [179,180]. What is more, it may be a challenge to find a universal biomarker for different cancer types, which is related to tissue-dependent signalling and ligand levels in specific cancer types [95,181]. Currently, in the ongoing clinical trials, i.a., AXL protein levels and AXL phosphorylation status are being analysed [182].

5.4. Potential Concerns

The development of anticancer therapy targeting TAM family receptors requires the consideration of their expression and function in the normal cells of the body. It is worth mentioning that MERTK and AXL kinases are expressed in immune cells such as macrophages, NK cells, and dendritic cells [47,64,183]. Studies of single, double, and triple knockouts of TAM receptors in mice have demonstrated, e.g., inflammation and autoimmunity, with more severe dysfunctions when targeting more than one kinase [39,43,44]. Moreover, as mentioned above, in the case of less selective TAM inhibitors, structurally-related receptors may also be inhibited, leading to increased off-targets and toxicities. TAM receptors promote tissue-specific macrophage polarisation into a pro-tumour M2-like phenotype as AXL and TYRO3 regulate phagocytosis in dendritic cells, whereas MERTK do so in the thymus and retina [64,156,184]. Retinal toxicity is observed under MERTK inhibition and is one of the main concerns in the development of TAM inhibitors [185,186]
As other reports suggest, the activity and downstream signalling of TAM receptors is also regulated by homodimerisation, and this intricate web of interactions and relations makes it difficult to create an inhibitor that is selective, effective, and does not deregulate other signalling pathways [64]. On the other hand, in the case of single-agent anticancer therapy, the emergence of resistance is a well-known and inevitable phenomenon. Upon AXL inhibition, MERTK is upregulated in several cancer models and constitutes one of many mechanisms of drug resistance build-up [160]. So far, mechanisms of resistance to TAM-targeted therapies have not been extensively studied, besides the abovementioned preclinical evaluation of MERTK and AXL’s mutual impact on resistance.

6. Future Directions

In summarising the emerging data and collected information, it is becoming clear that the further development of therapies targeting TAM kinases is necessary. It is worth emphasising that small molecular inhibitors are effective, but often highly toxic. On the other hand, antibodies are much more selective and less toxic, but so far do not show as strong a therapeutic effect as small molecular inhibitors. For the further development of TAM-targeted therapies, it would be crucial to find biomarkers stratifying patients to predict which of them may present the greatest therapeutic response. As the effect of targeting TAM kinases, especially AXL and MERTK, is known to increase the effectiveness of other therapies such as chemotherapy, immunotherapy, radiotherapy, and targeted therapies, it is crucial to gather the most in-depth data on the possible and most promising clinical combinations. In addition, further studies should be also implemented to reveal potential toxicities, relevant clinical regimens, therapeutic strategies, and possible bypass mechanisms and drug resistance.

7. Conclusions

In this review, we provide a comprehensive picture of the current status of the TAM receptor inhibition strategies in solid tumour therapy. We present how structural differences between TAM receptors can be exploited to inhibit their activity, with antibodies in various formats, antibody–drug conjugates, protein decoys, and small compounds targeting their extracellular domains and on the other side, the wide array of tyrosine kinase inhibitors directly blocking signal transduction via the intracellular kinase domains.
The great complexity of TAM receptors’ activation mechanisms and their involvement in carcinogenesis make it difficult to develop a drug that is effective, specific, and safe for patients. Hence, we need to further understand how the different structures of low-molecular-weight compounds and antibody formats define their properties and characteristics to overcome acquired drug resistance and make these drugs even more potent in the future. Here, we do not present one approach as better than the other, as we firmly believe that every way to battle cancer is the right way in this never-ending war on cancer.

Author Contributions

Review design: A.M., F.M., D.P., B.K., M.W. and J.P.; manuscript preparation: A.M., F.M. and D.P. All authors have read and agreed to the published version of the manuscript.

Funding

The research was co-financed by the National Centre of Research and Development and Celon Pharma S.A., project “Development of an innovative, first-in-class, small-molecule, double-targeted therapeutic with immunomodulating and cytotoxic activity as a candidate for treatment of leukaemia and solid tumours”, grant number POIR.01.01.01-00-0472/19-00. The first author, Agata Mikołajczyk, is a PhD student in a project financed by the Ministry of Science and Higher Education (50//DW/2017/01/1).

Acknowledgments

The authors would like to thank Wojciech Pietrus from Celon Pharma S.A. for his assistance in the graphic preparation of the presented chemical structures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Scott, R.S.; McMahon, E.J.; Pop, S.M.; Reap, E.A.; Caricchio, R.; Cohen, P.L.; Earp, H.S.; Matsushima, G.K. Phagocytosis and Clearance of Apoptotic Cells Is Mediated by MER. Nature 2001, 411, 207–211. [Google Scholar] [CrossRef] [PubMed]
  2. Schulz, N.T.; Paulhiac, C.I.; Lee, L.; Zhou, R. Isolation and Expression Analysis of Tyro3, a Murine Growth Factor Receptor Tyrosine Kinase Preferentially Expressed in Adult Brain. Mol. Brain Res. 1995, 28, 273–280. [Google Scholar] [CrossRef]
  3. Weinger, J.G.; Brosnan, C.F.; Loudig, O.; Goldberg, M.F.; Macian, F.; Arnett, H.A.; Prieto, A.L.; Tsiperson, V.; Shafit-Zagardo, B. Loss of the Receptor Tyrosine Kinase Axl Leads to Enhanced Inflammation in the CNS and Delayed Removal of Myelin Debris during Experimental Autoimmune Encephalomyelitis. J. Neuroinflamm. 2011, 8, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Cavet, M.E.; Smolock, E.M.; Ozturk, O.H.; World, C.; Pang, J.; Konishi, A.; Berk, B.C. Gas6–Axl Receptor Signaling Is Regulated by Glucose in Vascular Smooth Muscle Cells. ATVB 2008, 28, 886–891. [Google Scholar] [CrossRef] [Green Version]
  5. O’Bryan, J.P.; Frye, R.A.; Cogswell, P.C.; Neubauer, A.; Kitch, B.; Prokop, C.; Iii, R.E.; Beau, M.M.L.; Earp, H.S.; LIUl, 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] [PubMed] [Green Version]
  6. Graham, D.; Bowman, G.; Dawson, T.; Stanford, W.; Earp, H.; Snodgrass, H. Cloning and Developmental Expression Analysis of the Murine C-Mer Tyrosine Kinase. Oncogene 1995, 10, 2349–2359. [Google Scholar]
  7. Ohashi, K.; Nagata, K.; Toshima, J.; Nakano, T.; Arita, H.; Tsuda, H.; Suzuki, K.; Mizuno, K. Stimulation of Sky Receptor Tyrosine Kinase by the Product of Growth Arrest-Specific Gene 6. J. Biol. Chem. 1995, 270, 22681–22684. [Google Scholar] [CrossRef] [Green Version]
  8. Sasaki, T.; Knyazev, P.G.; Clout, N.J.; Cheburkin, Y.; Göhring, W.; Ullrich, A.; Timpl, R.; Hohenester, E. Structural Basis for Gas6–Axl Signalling. EMBO J. 2006, 25, 80–87. [Google Scholar] [CrossRef] [Green Version]
  9. Tsou, W.-I.; Nguyen, K.-Q.N.; 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] [Green Version]
  10. Stitt, T.N.; Conn, G.; Gore, M.; Lai, C.; Bruno, J.; Radziejewski, C.; Mattsson, K.; Fisher, J.; Gies, D.R.; Jones, P.F.; et al. The Anticoagulation Factor Protein S and Its Relative, Gas6, Are Ligands for the Tyro 3/Axl Family of Receptor Tyrosine Kinases. Cell 1995, 80, 661–670. [Google Scholar] [CrossRef] [Green Version]
  11. Varnum, B.; Young, C.; Elliott, G.; Garcia, A.; Bartley, T.; Fridell, Y.; Hunt, R.; Trail, G.; Clogston, C.; Toso, R. Axl Receptor Tyrosine Kinase Stimulated by the Vitamin K-Dependent Protein Encoded by Growth-Arrest-Specific Gene 6. Nature 1995, 373, 623–626. [Google Scholar] [CrossRef] [PubMed]
  12. Mercer, J.; Helenius, A. Vaccinia Virus Uses Macropinocytosis and Apoptotic Mimicry to Enter Host Cells. Science 2008, 320, 531–535. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, C.; Li, Q.; Darrow, A.L.; Wang, Y.; Derian, C.K.; Yang, J.; de Garavilla, L.; Andrade-Gordon, P.; Damiano, B.P. Mer Receptor Tyrosine Kinase Signaling Participates in Platelet Function. ATVB 2004, 24, 1118–1123. [Google Scholar] [CrossRef] [PubMed]
  14. Geng, K.; Kumar, S.; Kimani, S.G.; Kholodovych, V.; Kasikara, C.; Mizuno, K.; Sandiford, O.; Rameshwar, P.; Kotenko, S.V.; Birge, R.B. Requirement of Gamma-Carboxyglutamic Acid Modification and Phosphatidylserine Binding for the Activation of Tyro3, Axl, and Mertk Receptors by Growth Arrest-Specific 6. Front. Immunol. 2017, 8, 1521. [Google Scholar] [CrossRef] [PubMed]
  15. Aehnlich, P.; Powell, R.M.; Peeters, M.J.W.; Rahbech, A.; thor Straten, P. TAM Receptor Inhibition–Implications for Cancer and the Immune System. Cancers 2021, 13, 1195. [Google Scholar] [CrossRef] [PubMed]
  16. Li, R.; Chen, J.; Hammonds, G.; Phillips, H.; Armanini, M.; Wood, P.; Bunge, R.; Godowski, P.; Sliwkowski, M.; Mather, J. Identification of Gas6 as a Growth Factor for Human Schwann Cells. J. Neurosci. 1996, 16, 2012–2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Linger, R.M.A.; Cohen, R.A.; Cummings, C.T.; Sather, S.; Migdall-Wilson, J.; Middleton, D.H.G.; Lu, X.; Barón, A.E.; Franklin, W.A.; Merrick, D.T.; et al. Mer or Axl Receptor Tyrosine Kinase Inhibition Promotes Apoptosis, Blocks Growth and Enhances Chemosensitivity of Human Non-Small Cell Lung Cancer. Oncogene 2013, 32, 3420–3431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Demarchi, F.; Verardo, R.; Varnum, B.; Brancolini, C.; Schneider, C. Gas6 Anti-Apoptotic Signaling Requires NF-ΚB Activation. J. Biol. Chem. 2001, 276, 31738–31744. [Google Scholar] [CrossRef] [Green Version]
  19. Paccez, J.D.; Vasques, G.J.; Correa, R.G.; Vasconcellos, J.F.; Duncan, K.; Gu, X.; Bhasin, M.; Libermann, T.A.; Zerbini, L.F. The Receptor Tyrosine Kinase Axl Is an Essential Regulator of Prostate Cancer Proliferation and Tumor Growth and Represents a New Therapeutic Target. Oncogene 2013, 32, 689–698. [Google Scholar] [CrossRef] [Green Version]
  20. Goruppi, S.; Ruaro, E.; Varnum, B.; Schneider, C. Requirement of Phosphatidylinositol 3-Kinase-Dependent Pathway and Src for Gas6-Axl Mitogenic and Survival Activities in NIH 3T3 Fibroblasts. Mol. Cell. Biol. 1997, 17, 4442–4453. [Google Scholar] [CrossRef] [Green Version]
  21. 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]
  22. Knubel, K.H.; Pernu, B.M.; Sufit, A.; Nelson, S.; Pierce, A.M.; Keating, A.K. MerTK Inhibition Is a Novel Therapeutic Approach for Glioblastoma Multiforme. Oncotarget 2014, 5, 1338–1351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Schlegel, J.; Sambade, M.J.; Sather, S.; Moschos, S.J.; Tan, A.-C.; Winges, A.; DeRyckere, D.; Carson, C.C.; Trembath, D.G.; Tentler, J.J.; et al. MERTK Receptor Tyrosine Kinase Is a Therapeutic Target in Melanoma. J. Clin. Investig. 2013, 123, 2257–2267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Davra, V.; Kumar, S.; Geng, K.; Calianese, D.; Mehta, D.; Gadiyar, V.; Kasikara, C.; Lahey, K.C.; Chang, Y.; Wichroski, M.; et al. Axl and Mertk Receptors Cooperate to Promote Breast Cancer Progression by Combined Oncogenic Signaling and Evasion of Host Antitumor Immunity. Cancer Res. 2021, 81, 698–712. [Google Scholar] [CrossRef]
  25. Jansen, F.H.; van Rijswijk, A.; Teubel, W.; van Weerden, W.M.; Reneman, S.; van den Bemd, G.-J.; Roobol, M.J.; Bangma, C.H.; Staal, F.J.T.; Jenster, G. Profiling of Antibody Production against Xenograft-Released Proteins by Protein Microarrays Discovers Prostate Cancer Markers. J. Proteome Res. 2012, 11, 728–735. [Google Scholar] [CrossRef]
  26. Ben-Batalla, I.; Schultze, A.; Wroblewski, M.; Erdmann, R.; Heuser, M.; Waizenegger, J.S.; Riecken, K.; Binder, M.; Schewe, D.; Sawall, S.; et al. Axl, a Prognostic and Therapeutic Target in Acute Myeloid Leukemia Mediates Paracrine Crosstalk of Leukemia Cells with Bone Marrow Stroma. Blood 2013, 122, 2443–2452. [Google Scholar] [CrossRef]
  27. Schmitz, R.; Valls, A.F.; Yerbes, R.; von Richter, S.; Kahlert, C.; Loges, S.; Weitz, J.; Schneider, M.; de Almodovar, C.R.; Ulrich, A.; et al. TAM Receptors Tyro3 and Mer as Novel Targets in Colorectal Cancer. Oncotarget 2016, 7, 56355–56370. [Google Scholar] [CrossRef] [Green Version]
  28. 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]
  29. 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]
  30. Uribe, D.J.; Mandell, E.K.; Watson, A.; Martinez, J.D.; Leighton, J.A.; Ghosh, S.; Rothlin, C.V. The Receptor Tyrosine Kinase AXL Promotes Migration and Invasion in Colorectal Cancer. PLoS ONE 2017, 12, e0179979. [Google Scholar] [CrossRef] [Green Version]
  31. Tian, Y.; Zhang, Z.; Miao, L.; Yang, Z.; Yang, J.; Wang, Y.; Qian, D.; Cai, H.; Wang, Y. Anexelekto (AXL) Increases Resistance to EGFR-TKI and Activation of AKT and ERK1/2 in Non-Small Cell Lung Cancer Cells. Oncol. Res. 2016, 24, 295–303. [Google Scholar] [CrossRef] [PubMed]
  32. Giles, K.M.; Kalinowski, F.C.; Candy, P.A.; Epis, M.R.; Zhang, P.M.; Redfern, A.D.; Stuart, L.M.; Goodall, G.J.; Leedman, P.J. Axl Mediates Acquired Resistance of Head and Neck Cancer Cells to the Epidermal Growth Factor Receptor Inhibitor Erlotinib. Mol. Cancer Ther. 2013, 12, 2541–2558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. 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] [PubMed] [Green Version]
  34. Yan, D.; Parker, R.E.; Wang, X.; Frye, S.V.; Earp, H.S.; DeRyckere, D.; Graham, D.K. MERTK Promotes Resistance to Irreversible EGFR Tyrosine Kinase Inhibitors in Non–Small Cell Lung Cancers Expressing Wild-Type EGFR Family Members. Clin. Cancer Res. 2018, 24, 6523–6535. [Google Scholar] [CrossRef] [Green Version]
  35. 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]
  36. 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]
  37. Thomson, S.; Petti, F.; Sujka-Kwok, I.; Mercado, P.; Bean, J.; Monaghan, M.; Seymour, S.L.; Argast, G.M.; Epstein, D.M.; Haley, J.D. A Systems View of Epithelial–Mesenchymal Transition Signaling States. Clin. Exp. Metastasis 2011, 28, 137–155. [Google Scholar] [CrossRef] [Green Version]
  38. Morimoto, M.; Horikoshi, Y.; Nakaso, K.; Kurashiki, T.; Kitagawa, Y.; Hanaki, T.; Sakamoto, T.; Honjo, S.; Umekita, Y.; Fujiwara, Y.; et al. Oncogenic Role of TYRO3 Receptor Tyrosine Kinase in the Progression of Pancreatic Cancer. Cancer Lett. 2020, 470, 149–160. [Google Scholar] [CrossRef]
  39. Lu, Q.; Lemke, G. Homeostatic Regulation of the Immune System by Receptor Tyrosine Kinases of the Tyro 3 Family. Science 2001, 293, 306–311. [Google Scholar] [CrossRef]
  40. Carrera Silva, E.A.; Chan, P.Y.; Joannas, L.; Errasti, A.E.; Gagliani, N.; Bosurgi, L.; Jabbour, M.; Perry, A.; Smith-Chakmakova, F.; Mucida, D.; et al. T Cell-Derived Protein S Engages TAM Receptor Signaling in Dendritic Cells to Control the Magnitude of the Immune Response. Immunity 2013, 39, 160–170. [Google Scholar] [CrossRef] [Green Version]
  41. Lemke, G.; Rothlin, C.V. Immunobiology of the TAM Receptors. Nat. Rev. Immunol. 2008, 8, 327–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Rothlin, C.V.; Ghosh, S.; Zuniga, E.I.; Oldstone, M.B.A.; Lemke, G. TAM Receptors Are Pleiotropic Inhibitors of the Innate Immune Response. Cell 2007, 131, 1124–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. 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] [PubMed]
  44. Cohen, P.L.; Caricchio, R.; Abraham, V.; Camenisch, T.D.; Jennette, J.C.; Roubey, R.A.S.; Earp, H.S.; Matsushima, G.; Reap, E.A. Delayed Apoptotic Cell Clearance and Lupus-like Autoimmunity in Mice Lacking the c-Mer Membrane Tyrosine Kinase. J. Exp. Med. 2002, 196, 135–140. [Google Scholar] [CrossRef] [PubMed]
  45. Tirado-Gonzalez, I.; Descot, A.; Soetopo, D.; Nevmerzhitskaya, A.; Schäffer, A.; Kur, I.-M.; Czlonka, E.; Wachtel, C.; Tsoukala, I.; Müller, L.; et al. AXL Inhibition in Macrophages Stimulates Host-versus-Leukemia Immunity and Eradicates Naïve and Treatment-Resistant Leukemia. Cancer Discov. 2021, 11, 2924–2943. [Google Scholar] [CrossRef]
  46. Zhou, Y.; Fei, M.; Zhang, G.; Liang, W.-C.; Lin, W.; Wu, Y.; Piskol, R.; Ridgway, J.; McNamara, E.; Huang, H.; et al. Blockade of the Phagocytic Receptor MerTK on Tumor-Associated Macrophages Enhances P2X7R-Dependent STING Activation by Tumor-Derived CGAMP. Immunity 2020, 52, 357–373.e9. [Google Scholar] [CrossRef]
  47. 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]
  48. Giroud, P.; Renaudineau, S.; Gudefin, L.; Calcei, A.; Menguy, T.; Rozan, C.; Mizrahi, J.; Caux, C.; Duong, V.; Valladeau-Guilemond, J. Expression of TAM-R in Human Immune Cells and Unique Regulatory Function of MerTK in IL-10 Production by Tolerogenic DC. Front. Immunol. 2020, 11, 564133. [Google Scholar] [CrossRef]
  49. Cook, R.S.; Jacobsen, K.M.; Wofford, A.M.; DeRyckere, D.; Stanford, J.; Prieto, A.L.; Redente, E.; Sandahl, M.; Hunter, D.M.; Strunk, K.E.; et al. MerTK Inhibition in Tumor Leukocytes Decreases Tumor Growth and Metastasis. J. Clin. Investig. 2013, 123, 3231–3242. [Google Scholar] [CrossRef] [Green Version]
  50. Aguilera, T.A.; Rafat, M.; Castellini, L.; Shehade, H.; Kariolis, M.S.; Hui, A.B.-Y.; 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] [Green Version]
  51. Peeters, M.J.W.; Dulkeviciute, D.; Draghi, A.; Ritter, C.; Rahbech, A.; Skadborg, S.K.; Seremet, T.; Carnaz Simões, A.M.; Martinenaite, E.; Halldórsdóttir, H.R.; et al. MERTK Acts as a Costimulatory Receptor on Human CD8+ T Cells. Cancer Immunol. Res. 2019, 7, 1472–1484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Fantl, W.J.; Johnson, D.E.; Williams, L.T. Signalling by Receptor Tyrosine Kinases. Annu. Rev. Biochem. 1993, 62, 453–481. [Google Scholar] [CrossRef] [PubMed]
  53. Ullrich, A.; Schlessinger, J. Signal Transduction by Receptors with Tyrosine Kinase Activity. Cell 1990, 61, 203–212. [Google Scholar] [CrossRef]
  54. Lew, E.D.; Oh, J.; Burrola, P.G.; Lax, I.; Zagórska, A.; Través, P.G.; Schlessinger, J.; Lemke, G. Differential TAM Receptor–Ligand–Phospholipid Interactions Delimit Differential TAM Bioactivities. eLife 2014, 3, e03385. [Google Scholar] [CrossRef] [PubMed]
  55. Lemmon, M.A.; Schlessinger, J. Cell Signaling by Receptor Tyrosine Kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [Green Version]
  56. Ling, L.; Templeton, D.; Kung, H.-J. Identification of the Major Autophosphorylation Sites of Nyk/Mer, an NCAM-Related Receptor Tyrosine Kinase. J. Biol. Chem. 1996, 271, 18355–18362. [Google Scholar] [CrossRef] [Green Version]
  57. Zhu, C.; Wei, Y.; Wei, X. AXL Receptor Tyrosine Kinase as a Promising Anti-Cancer Approach: Functions, Molecular Mechanisms and Clinical Applications. Mol. Cancer 2019, 18, 153. [Google Scholar] [CrossRef] [Green Version]
  58. Braunger, J.; Schleithoff, L.; Schulz, A.S.; Kessler, H.; Lammers, R.; Ullrich, A.; Bartram, C.R.; Janssen, J.W. Intracellular Signaling of the Ufo/Axl Receptor Tyrosine Kinase Is Mediated Mainly by a Multi-Substrate Docking-Site. Oncogene 1997, 14, 2619–2631. [Google Scholar] [CrossRef] [Green Version]
  59. Auyez, A.; Sayan, A.E.; Kriajevska, M.; Tulchinsky, E. AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew. Cancers 2021, 13, 4864. [Google Scholar] [CrossRef]
  60. Bellosta, P.; Costa, M.; Lin, D.A.; Basilico, C. The Receptor Tyrosine Kinase ARK Mediates Cell Aggregation by Homophilic Binding. Mol. Cell. Biol. 1995, 15, 614–625. [Google Scholar] [CrossRef] [Green Version]
  61. Burchert, A.; Attar, E.C.; McCloskey, P.; Fridell, Y.-W.C.; Liu, E.T. Determinants for Transformation Induced by the Axl Receptor Tyrosine Kinase. Oncogene 1998, 16, 3177–3187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Korshunov, V.A. Axl-Dependent Signalling: A Clinical Update. Clin. Sci. 2012, 122, 361–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Pierce, A.; Bliesner, B.; Xu, M.; Nielsen-Preiss, S.; Lemke, G.; Tobet, S.; Wierman, M.E. Axl and Tyro3 Modulate Female Reproduction by Influencing Gonadotropin-Releasing Hormone Neuron Survival and Migration. Mol. Endocrinol. 2008, 22, 2481–2495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. 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] [PubMed]
  65. Brown, J.E.; Krodel, M.; Pazos, M.; Lai, C.; Prieto, A.L. Cross-Phosphorylation, Signaling and Proliferative Functions of the Tyro3 and Axl Receptors in Rat2 Cells. PLoS ONE 2012, 7, e36800. [Google Scholar] [CrossRef] [Green Version]
  66. Vouri, M.; Croucher, D.R.; Kennedy, S.P.; An, Q.; Pilkington, G.J.; Hafizi, S. Axl-EGFR Receptor Tyrosine Kinase Hetero-Interaction Provides EGFR with Access to pro-Invasive Signalling in Cancer Cells. Oncogenesis 2016, 5, e266. [Google Scholar] [CrossRef] [Green Version]
  67. McShane, L.; Tabas, I.; Lemke, G.; Kurowska-Stolarska, M.; Maffia, P. TAM Receptors in Cardiovascular Disease. Cardiovasc. Res. 2019, 115, 1286–1295. [Google Scholar] [CrossRef] [Green Version]
  68. Mudduluru, G.; Ceppi, P.; Kumarswamy, R.; Scagliotti, G.V.; Papotti, M.; Allgayer, H. Regulation of Axl Receptor Tyrosine Kinase Expression by MiR-34a and MiR-199a/b in Solid Cancer. Oncogene 2011, 30, 2888–2899. [Google Scholar] [CrossRef]
  69. Kurowska-Stolarska, M.; Alivernini, S.; Melchor, E.G.; Elmesmari, A.; Tolusso, B.; Tange, C.; Petricca, L.; Gilchrist, D.S.; Di Sante, G.; Keijzer, C.; et al. MicroRNA-34a Dependent Regulation of AXL Controls the Activation of Dendritic Cells in Inflammatory Arthritis. Nat. Commun. 2017, 8, 15877. [Google Scholar] [CrossRef]
  70. Thorp, E.; Vaisar, T.; Subramanian, M.; Mautner, L.; Blobel, C.; Tabas, I. Shedding of the Mer Tyrosine Kinase Receptor Is Mediated by ADAM17 Protein through a Pathway Involving Reactive Oxygen Species, Protein Kinase Cδ, and P38 Mitogen-Activated Protein Kinase (MAPK). J. Biol. Chem. 2011, 286, 33335–33344. [Google Scholar] [CrossRef] [Green Version]
  71. Merilahti, J.A.M.; Ojala, V.K.; Knittle, A.M.; Pulliainen, A.T.; Elenius, K. Genome-Wide Screen of Gamma-Secretase–Mediated Intramembrane Cleavage of Receptor Tyrosine Kinases. MBoC 2017, 28, 3123–3131. [Google Scholar] [CrossRef] [PubMed]
  72. Orme, J.J.; Du, Y.; Vanarsa, K.; Mayeux, J.; Li, L.; Mutwally, A.; Arriens, C.; Min, S.; Hutcheson, J.; Davis, L.S.; et al. Heightened Cleavage of Axl Receptor Tyrosine Kinase by ADAM Metalloproteases May Contribute to Disease Pathogenesis in SLE. Clin. Immunol. 2016, 169, 58–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Sather, S.; Kenyon, K.D.; Lefkowitz, J.B.; Liang, X.; Varnum, B.C.; Henson, P.M.; Graham, D.K. A Soluble Form of the Mer Receptor Tyrosine Kinase Inhibits Macrophage Clearance of Apoptotic Cells and Platelet Aggregation. Blood 2007, 109, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
  74. Weinger, J.G.; Omari, K.M.; Marsden, K.; Raine, C.S.; Shafit-Zagardo, B. Up-Regulation of Soluble Axl and Mer Receptor Tyrosine Kinases Negatively Correlates with Gas6 in Established Multiple Sclerosis Lesions. Am. J. Pathol. 2009, 175, 283–293. [Google Scholar] [CrossRef] [Green Version]
  75. Gal, A.; Li, Y.; Thompson, D.A.; Weir, J.; Orth, U.; Jacobson, S.G.; Apfelstedt-Sylla, E.; Vollrath, D. Mutations in MERTK, the Human Orthologue of the RCS Rat Retinal Dystrophy Gene, Cause Retinitis Pigmentosa. Nat. Genet. 2000, 26, 270–271. [Google Scholar] [CrossRef]
  76. D’Cruz, P.M. Mutation of the Receptor Tyrosine Kinase Gene Mertk in the Retinal Dystrophic RCS Rat. Hum. Mol. Genet. 2000, 9, 645–651. [Google Scholar] [CrossRef] [Green Version]
  77. 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]
  78. Niederst, M.J.; Engelman, J.A. Bypass Mechanisms of Resistance to Receptor Tyrosine Kinase Inhibition in Lung Cancer. Sci. Signal. 2013, 6, re6. [Google Scholar] [CrossRef] [Green Version]
  79. 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, 10. [Google Scholar]
  80. Nagata, K.; Ohashi, K.; Nakano, T.; Arita, H.; Zong, C.; Hanafusa, H.; Mizuno, K. Identification of the Product of Growth Arrest-Specific Gene 6 as a Common Ligand for Axl, Sky, and Mer Receptor Tyrosine Kinases. J. Biol. Chem. 1996, 271, 30022–30027. [Google Scholar] [CrossRef] [Green Version]
  81. 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] [PubMed] [Green Version]
  82. Zagórska, A.; Través, P.G.; Lew, E.D.; Dransfield, I.; Lemke, G. Diversification of TAM Receptor Tyrosine Kinase Function. Nat. Immunol. 2014, 15, 920–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Sadahiro, H.; Kang, K.-D.; Gibson, J.T.; Minata, M.; Yu, H.; Shi, J.; Chhipa, R.; Chen, Z.; Lu, S.; Simoni, Y.; et al. Activation of the Receptor Tyrosine Kinase AXL Regulates the Immune Microenvironment in Glioblastoma. Cancer Res. 2018, 78, 3002–3013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Uehara, S.; Fukuzawa, Y.; Matuyama, T.; Gotoh, K. Role of Tyro3, Axl, and Mer Receptors and Their Ligands (Gas6, and Protein S) in Patients with Hepatocellular Carcinoma. JCT 2017, 8, 112–130. [Google Scholar] [CrossRef] [Green Version]
  85. Davra, V.; Kimani, S.; Calianese, D.; Birge, R. Ligand Activation of TAM Family Receptors-Implications for Tumor Biology and Therapeutic Response. Cancers 2016, 8, 107. [Google Scholar] [CrossRef] [PubMed]
  86. Uehara, H.; Shacter, E. Auto-Oxidation and Oligomerization of Protein S on the Apoptotic Cell Surface Is Required for Mer Tyrosine Kinase-Mediated Phagocytosis of Apoptotic Cells. J. Immunol. 2008, 180, 2522–2530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lemke, G. Biology of the TAM Receptors. Cold Spring Harb. Perspect. Biol. 2013, 5, a009076. [Google Scholar] [CrossRef]
  88. 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] [Green Version]
  89. Di Stasi, R.; De Rosa, L.; D’Andrea, L.D. Therapeutic Aspects of the Axl/Gas6 Molecular System. Drug Discov. Today 2020, 25, 2130–2148. [Google Scholar] [CrossRef]
  90. Wu, X.; Ma, W.; Zhou, Q.; Yan, H.; Lim, Z.-F.; Huang, M.; Deng, C.; Yu, X.; Su, H.; Komo, S.; et al. AXL–GAS6 Expression Can Predict for Adverse Prognosis in Non-Small Cell Lung Cancer with Brain Metastases. J. Cancer Res. Clin. Oncol. 2017, 143, 1947–1957. [Google Scholar] [CrossRef] [Green Version]
  91. Mao, S.; Wu, Y.; Wang, R.; Guo, Y.; Bi, D.; Ma, W.; Zhang, W.; Zhang, J.; Yan, Y.; Yao, X. Overexpression of GAS6 Promotes Cell Proliferation and Invasion in Bladder Cancer by Activation of the PI3K/AKT Pathway. OTT 2020, 13, 4813–4824. [Google Scholar] [CrossRef] [PubMed]
  92. Buehler, M.; Tse, B.; Leboucq, A.; Jacob, F.; Caduff, R.; Fink, D.; Goldstein, D.R.; Heinzelmann-Schwarz, V. Meta-Analysis of Microarray Data Identifies GAS6 Expression as an Independent Predictor of Poor Survival in Ovarian Cancer. BioMed Res. Int. 2013, 2013, 238284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Whitman, S.P.; Kohlschmidt, J.; Maharry, K.; Volinia, S.; Mrózek, K.; Nicolet, D.; Schwind, S.; Becker, H.; Metzeler, K.H.; Mendler, J.H.; et al. GAS6 Expression Identifies High-Risk Adult AML Patients: Potential Implications for Therapy. Leukemia 2014, 28, 1252–1258. [Google Scholar] [CrossRef] [Green Version]
  94. Cardone, C.; Liguori, G.; Troiani, T.; Nappi, A.; Amoroso, N.; Iaffaioli, V.R.; Romano, C.; Botti, G.; Vitagliano, D.; Martini, G.; et al. Expression of Axl Receptor and Its Ligand Gas6 in Colorectal Cancer (Crc). Ann. Oncol. 2014, 25, iv73. [Google Scholar] [CrossRef]
  95. 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] [Green Version]
  96. Al Kafri, N.; Hafizi, S. Tumour-Secreted Protein S (ProS1) Activates a Tyro3-Erk Signalling Axis and Protects Cancer Cells from Apoptosis. Cancers 2019, 11, 1843. [Google Scholar] [CrossRef] [Green Version]
  97. Ubil, E.; Caskey, L.; Holtzhausen, A.; Hunter, D.; Story, C.; Earp, H.S. Tumor-Secreted Pros1 Inhibits Macrophage M1 Polarization to Reduce Antitumor Immune Response. J. Clin. Investig. 2018, 128, 2356–2369. [Google Scholar] [CrossRef] [Green Version]
  98. Chen, T.J.; Mydel, P.; Benedyk-Machaczka, M.; Kamińska, M.; Kalucka, U.; Blø, M.; Furriol, J.; Gausdal, G.; Lorens, J.; Osman, T.; et al. AXL Targeting by a Specific Small Molecule or Monoclonal Antibody Inhibits Renal Cell Carcinoma Progression in an Orthotopic Mice Model. Physiol. Rep. 2021, 9, e15140. [Google Scholar] [CrossRef]
  99. Leconet, W.; Larbouret, C.; Chardès, T.; Thomas, G.; Neiveyans, M.; Busson, M.; Jarlier, M.; Radosevic-Robin, N.; Pugnière, 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]
  100. Yu, H.; Liu, R.; Ma, B.; Li, X.; Yen, H.; Zhou, Y.; Krasnoperov, V.; Xia, Z.; Zhang, X.; Bove, A.M.; et al. Axl Receptor Tyrosine Kinase Is a Potential Therapeutic Target in Renal Cell Carcinoma. Br. J. Cancer 2015, 113, 616–625. [Google Scholar] [CrossRef] [Green Version]
  101. Leconet, W.; Chentouf, M.; du Manoir, S.; Chevalier, C.; Sirvent, A.; Aït-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]
  102. Duan, Y.; Luo, L.; Qiao, C.; Li, X.; Wang, J.; Liu, H.; Zhou, T.; Shen, B.; Lv, M.; Feng, J. A Novel Human Anti-AXL Monoclonal Antibody Attenuates Tumour Cell Migration. Scand. J. Immunol. 2019, 90, e12777. [Google Scholar] [CrossRef] [PubMed]
  103. 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]
  104. Alvarado, D.; Vitale, L.; Murphy, M.; O’Neill, T.; Natoli, E.; Lillquist, J.; Crew, L.; Wasiuk, A.; Weidlick, J.; Sisson, C.; et al. 550 An Axl-Targeting Monoclonal Antibody That Inhibits Axl Activity and Potently Stimulates the Innate Immune Response. J. ImmunoTherapy of Cancer 2020, 8, A334.1. [Google Scholar]
  105. Chien, C.-W.; Hou, P.-C.; Wu, H.-C.; Chang, Y.-L.; Lin, S.-C.; Lin, S.-C.; Lin, B.-W.; Lee, J.-C.; Chang, Y.-J.; Sun, H.S.; et al. Targeting TYRO3 Inhibits Epithelial–Mesenchymal Transition and Increases Drug Sensitivity in Colon Cancer. Oncogene 2016, 35, 5872–5881. [Google Scholar] [CrossRef]
  106. Takeda, S.; Andreu-Agullo, C.; Sridhar, S.; Halberg, N.; Lorenz, I.C.; Tavazoie, S.; Kurth, I.; Tavazoie, M. Abstract LB-277: Characterization of the Anti-Cancer and Immunologic Activity of RGX-019, a Novel Pre-Clinical Stage Humanized Monoclonal Antibody Targeting the MERTK Receptor. Cancer Res. 2019, 79, LB-277. [Google Scholar] [CrossRef]
  107. Ballantine, L.; Midgley, A.; Harris, D.; Richards, E.; Burgess, S.; Beresford, M.W. Increased Soluble Phagocytic Receptors SMer, STyro3 and SAxl and Reduced Phagocytosis in Juvenile-Onset Systemic Lupus Erythematosus. Pediatr. Rheumatol. 2015, 13, 10. [Google Scholar] [CrossRef] [Green Version]
  108. Kariolis, M.S.; Miao, Y.R.; Diep, A.; Nash, S.E.; Olcina, M.M.; Jiang, D.; Jones, D.S.; Kapur, S.; Mathews, I.I.; Koong, A.C.; et al. Inhibition of the GAS6/AXL Pathway Augments the Efficacy of Chemotherapies. J. Clin. Investig. 2016, 127, 183–198. [Google Scholar] [CrossRef] [Green Version]
  109. Duan, Y.; Hu, B.; Qiao, C.; Luo, L.; Li, X.; Wang, J.; Liu, H.; Zhou, T.; Shen, B.; Lv, M.; et al. Engineered AXL-ECD-Fc Variants That Abolish the AXL/Gas6 Interaction Suppress Tumor Cell Migration. Oncol. Lett. 2019, 17, 5784–5792. [Google Scholar] [CrossRef] [Green Version]
  110. Kariolis, M.S.; Miao, Y.R.; Jones, D.S.; 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]
  111. Kimani, S.G.; Kumar, S.; Bansal, N.; Singh, K.; Kholodovych, V.; Comollo, T.; Peng, Y.; Kotenko, S.V.; Sarafianos, S.G.; Bertino, J.R.; et al. Small Molecule Inhibitors Block Gas6-Inducible TAM Activation and Tumorigenicity. Sci. Rep. 2017, 7, 43908. [Google Scholar] [CrossRef] [PubMed]
  112. 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]
  113. Tie, J.-K.; Stafford, D.W. Functional Study of the Vitamin K Cycle Enzymes in Live Cells. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2017; Volume 584, pp. 349–394. ISBN 978-0-12-812213-6. [Google Scholar]
  114. Linger, R.M.A.; Keating, A.K.; Earp, H.S.; Graham, D.K. TAM Receptor Tyrosine Kinases: Biologic Functions, Signaling, and Potential Therapeutic Targeting in Human Cancer. In Advances in Cancer Research; Elsevier: Amsterdam, The Netherlands, 2008; Volume 100, pp. 35–83. ISBN 978-0-12-374358-9. [Google Scholar]
  115. Liu, M.-H.; Chen, S.-B.; Yu, J.; Liu, C.-J.; Zhang, X.-J. Promiscuity and Selectivity of Small-Molecule Inhibitors across TAM Receptor Tyrosine Kinases in Pediatric Leukemia. J. Mol. Graph. Model. 2017, 75, 125–131. [Google Scholar] [CrossRef] [PubMed]
  116. Baladi, T.; Abet, V.; Piguel, S. State-of-the-Art of Small Molecule Inhibitors of the TAM Family: The Point of View of the Chemist. Eur. J. Med. Chem. 2015, 105, 220–237. [Google Scholar] [CrossRef]
  117. Zhang, Y.-X.; Knyazev, P.G.; Cheburkin, Y.V.; Sharma, K.; Knyazev, Y.P.; Őrfi, L.; Szabadkai, I.; Daub, H.; Kéri, G.; Ullrich, A. AXL Is a Potential Target for Therapeutic Intervention in Breast Cancer Progression. Cancer Res. 2008, 68, 1905–1915. [Google Scholar] [CrossRef] [Green Version]
  118. Remsing Rix, L.L.; Rix, U.; Colinge, J.; Hantschel, O.; Bennett, K.L.; Stranzl, T.; Müller, A.; Baumgartner, C.; Valent, P.; Augustin, M.; et al. Global Target Profile of the Kinase Inhibitor Bosutinib in Primary Chronic Myeloid Leukemia Cells. Leukemia 2009, 23, 477–485. [Google Scholar] [CrossRef] [Green Version]
  119. Davis, M.I.; Hunt, J.P.; Herrgard, S.; Ciceri, P.; Wodicka, L.M.; Pallares, G.; Hocker, M.; Treiber, D.K.; Zarrinkar, P.P. Comprehensive Analysis of Kinase Inhibitor Selectivity. Nat. Biotechnol. 2011, 29, 1046–1051. [Google Scholar] [CrossRef]
  120. Mori, M.; Kaneko, N.; Ueno, Y.; Yamada, M.; Tanaka, R.; Saito, R.; Shimada, I.; Mori, K.; Kuromitsu, S. Gilteritinib, a FLT3/AXL Inhibitor, Shows Antileukemic Activity in Mouse Models of FLT3 Mutated Acute Myeloid Leukemia. Investig. New Drugs 2017, 35, 556–565. [Google Scholar] [CrossRef] [Green Version]
  121. Yakes, F.M.; Chen, J.; Tan, J.; Yamaguchi, K.; Shi, Y.; Yu, P.; Qian, F.; Chu, F.; Bentzien, F.; Cancilla, B.; et al. Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth. Mol. Cancer Ther. 2011, 10, 2298–2308. [Google Scholar] [CrossRef] [Green Version]
  122. 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] [Green Version]
  123. Cui, J.J.; Tran-Dubé, M.; Shen, H.; Nambu, M.; Kung, P.-P.; Pairish, M.; Jia, L.; Meng, J.; Funk, L.; Botrous, I.; et al. Structure Based Drug Design of Crizotinib (PF-02341066), a Potent and Selective Dual Inhibitor of Mesenchymal–Epithelial Transition Factor (c-MET) Kinase and Anaplastic Lymphoma Kinase (ALK). J. Med. Chem. 2011, 54, 6342–6363. [Google Scholar] [CrossRef] [PubMed]
  124. Karaman, M.W.; Herrgard, S.; Treiber, D.K.; Gallant, P.; Atteridge, C.E.; Campbell, B.T.; Chan, K.W.; Ciceri, P.; Davis, M.I.; Edeen, P.T.; et al. A Quantitative Analysis of Kinase Inhibitor Selectivity. Nat. Biotechnol. 2008, 26, 127–132. [Google Scholar] [CrossRef] [PubMed]
  125. Mollard, A.; Warner, S.L.; Call, L.T.; Wade, M.L.; Bearss, J.J.; Verma, A.; Sharma, S.; Vankayalapati, H.; Bearss, D.J. Design, Synthesis, and Biological Evaluation of a Series of Novel AXL Kinase Inhibitors. ACS Med. Chem. Lett. 2011, 2, 907–912. [Google Scholar] [CrossRef] [PubMed]
  126. Schroeder, G.M.; An, Y.; Cai, Z.-W.; Chen, X.-T.; Clark, C.; Cornelius, L.A.M.; 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]
  127. Cui, X.; Zheng, X.; Jiang, J.; Tan, F.; Ding, L.; Hu, P. Simultaneous Determination of a Novel C-Met/AXL Dual-Target Small-Molecule Inhibitor BPI-9016M and Its Metabolites in Human Plasma by Liquid Chromatography-Tandem Mass Spectrometry: Application in a Pharmacokinetic Study in Chinese Advanced Solid Tumor Patients. J. Chromatogr. B 2017, 1068–1069, 33–40. [Google Scholar] [CrossRef]
  128. Jimbo, T.; Hatanaka, M.; Komatsu, T.; Taira, T.; Kumazawa, K.; Maeda, N.; Suzuki, T.; Ota, M.; Haginoya, N.; Isoyama, T.; et al. DS-1205b, a Novel Selective Inhibitor of AXL Kinase, Blocks Resistance to EGFR-Tyrosine Kinase Inhibitors in a Non-Small Cell Lung Cancer Xenograft Model. Oncotarget 2019, 10, 5152–5167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. 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]
  130. 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] [Green Version]
  131. Xi, N.; Zhang, Y.; Wang, Z.; Wu, Y.; Wang, T. Abstract 1755: CT053PTSA, a Novel c-MET and VEGFR2 Inhibitor, Potently Suppresses Angiogenesis and Tumor Growth. Cancer Res. 2014, 74, 1755. [Google Scholar] [CrossRef]
  132. Ruvolo, P.P.; Ma, H.; Ruvolo, V.R.; Zhang, X.; Mu, H.; Schober, W.; Hernandez, I.; Gallardo, M.; Khoury, J.D.; Cortes, J.; et al. Anexelekto/MER Tyrosine Kinase Inhibitor ONO-7475 Arrests Growth and Kills FMS-like Tyrosine Kinase 3-Internal Tandem Duplication Mutant Acute Myeloid Leukemia Cells by Diverse Mechanisms. Haematologica 2017, 102, 2048–2057. [Google Scholar] [CrossRef] [Green Version]
  133. 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] [PubMed] [Green Version]
  134. 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] [PubMed] [Green Version]
  135. Hsu, J.; Chong, C.; Goon, L.; Balayan, J.; Wu, S.; Johnson, E.; Lorenzana, G.; Bannen, L.; Nguyen, L.; Scheffold, C.; et al. XL092, a Multi-Targeted Inhibitor of MET, VEGFR2, AXL and MER with an Optimized Pharmacokinetic Profile. Eur. J. Cancer 2020, 138, S16. [Google Scholar] [CrossRef]
  136. Minson, K.A.; Smith, C.C.; DeRyckere, D.; Libbrecht, C.; Lee-Sherick, A.B.; Huey, M.G.; Lasater, E.A.; Kirkpatrick, G.D.; Stashko, M.A.; Zhang, W.; et al. The MERTK/FLT3 Inhibitor MRX-2843 Overcomes Resistance-Conferring FLT3 Mutations in Acute Myeloid Leukemia. JCI Insight 2016, 1, e85630. [Google Scholar] [CrossRef] [PubMed]
  137. Burbridge, M.F.; Bossard, C.J.; Saunier, C.; Fejes, I.; Bruno, A.; Léonce, 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]
  138. Tanaka, M.; Siemann, D.W. Therapeutic Targeting of the Gas6/Axl Signaling Pathway in Cancer. Int. J. Mol. Sci. 2021, 22, 9953. [Google Scholar] [CrossRef]
  139. 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] [Green Version]
  140. Lai, S.; Li, R.; Raha, P.; Hu, Y.; Yan, J.; Zhang, H.; Marotta, A.; Zhang, Z. Abstract B148: Activity of the TAM Kinase-Targeting Compound, SLC-391, Is Mediated by the Engagement of the Immune System in CT-26 Syngeneic Mouse Model. Mol. Cancer Ther. 2018, 17, B148. [Google Scholar] [CrossRef]
  141. Rios-Doria, J.; Favata, M.; Lasky, K.; Feldman, P.; Lo, Y.; Yang, G.; Stevens, C.; Wen, X.; Sehra, S.; Katiyar, K.; et al. A Potent and Selective Dual Inhibitor of AXL and MERTK Possesses Both Immunomodulatory and Tumor-Targeted Activity. Front. Oncol. 2020, 10, 598477. [Google Scholar] [CrossRef]
  142. 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] [Green Version]
  143. Kim, H.D.; Park, E.J.; Choi, E.K.; Song, S.Y.; Hoe, K.-L.; Kim, D.-U. G-749 Promotes Receptor Tyrosine Kinase TYRO3 Degradation and Induces Apoptosis in Both Colon Cancer Cell Lines and Xenograft Mouse Models. Front. Pharmacol. 2021, 12, 730241. [Google Scholar] [CrossRef] [PubMed]
  144. Suárez, R.M.; Chevot, F.; Cavagnino, A.; Saettel, N.; Radvanyi, F.; Piguel, S.; Bernard-Pierrot, I.; Stoven, V.; Legraverend, M. Inhibitors of the TAM Subfamily of Tyrosine Kinases: Synthesis and Biological Evaluation. Eur. J. Med. Chem. 2013, 61, 2–25. [Google Scholar] [CrossRef] [PubMed]
  145. 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] [PubMed] [Green Version]
  146. McIver, A.L.; Zhang, W.; Liu, Q.; Jiang, X.; Stashko, M.A.; Nichols, J.; Miley, M.J.; Norris-Drouin, J.; Machius, M.; DeRyckere, D.; et al. Discovery of Macrocyclic Pyrimidines as MerTK-Specific Inhibitors. Chem. Med. Chem. 2017, 12, 207–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Zhang, W.; McIver, A.L.; Stashko, M.A.; DeRyckere, D.; Branchford, B.R.; Hunter, D.; Kireev, D.; Miley, M.J.; Norris-Drouin, J.; Stewart, W.M.; et al. Discovery of Mer Specific Tyrosine Kinase Inhibitors for the Treatment and Prevention of Thrombosis. J. Med. Chem. 2013, 56, 9693–9700. [Google Scholar] [CrossRef] [Green Version]
  148. Fujino, N.; Kubo, H.; Maciewicz, R.A. Phenotypic Screening Identifies Axl Kinase as a Negative Regulator of an Alveolar Epithelial Cell Phenotype. Lab. Investig. 2017, 97, 1047–1062. [Google Scholar] [CrossRef]
  149. Liu, J.; Zhang, W.; Stashko, M.A.; DeRyckere, D.; Cummings, C.T.; Hunter, D.; Yang, C.; Jayakody, C.N.; Cheng, N.; Simpson, C.; et al. UNC1062, a New and Potent Mer Inhibitor. Eur. J. Med. Chem. 2013, 65, 83–93. [Google Scholar] [CrossRef] [Green Version]
  150. 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]
  151. Powell, N.A.; Hoffman, J.K.; Ciske, F.L.; Kohrt, J.T.; Baxi, S.M.; Peng, Y.-W.; Zhong, M.; Catana, C.; Ohren, J.; Perrin, L.A.; et al. Optimization of Highly Selective 2,4-Diaminopyrimidine-5-Carboxamide Inhibitors of Sky Kinase. Bioorganic Med. Chem. Lett. 2013, 23, 1051–1055. [Google Scholar] [CrossRef]
  152. Vijayan, R.S.K.; He, P.; Modi, V.; Duong-Ly, K.C.; Ma, H.; Peterson, J.R.; Dunbrack, R.L.; Levy, R.M. Conformational Analysis of the DFG-Out Kinase Motif and Biochemical Profiling of Structurally Validated Type II Inhibitors. J. Med. Chem. 2015, 58, 466–479. [Google Scholar] [CrossRef] [Green Version]
  153. Bhullar, K.S.; Lagarón, N.O.; McGowan, E.M.; Parmar, I.; Jha, A.; Hubbard, B.P.; Rupasinghe, H.P.V. Kinase-Targeted Cancer Therapies: Progress, Challenges and Future Directions. Mol. Cancer 2018, 17, 48. [Google Scholar] [CrossRef] [PubMed]
  154. Kannaiyan, R.; Mahadevan, D. A Comprehensive Review of Protein Kinase Inhibitors for Cancer Therapy. Expert Rev. Anticancer Ther. 2018, 18, 1249–1270. [Google Scholar] [CrossRef] [PubMed]
  155. Roskoski, R. 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]
  156. Myers, K.V.; Amend, S.R.; Pienta, K.J. Targeting Tyro3, Axl and MerTK (TAM Receptors): Implications for Macrophages in the Tumor Microenvironment. Mol. Cancer 2019, 18, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Imran, M.; Asdaq, S.M.B.; Khan, S.A.; Unnikrishnan Meenakshi, D.; Alamri, A.S.; Alsanie, W.F.; Alhomrani, M.; Mohzari, Y.; Alrashed, A.; AlMotairi, M.; et al. Innovations and Patent Trends in the Development of USFDA Approved Protein Kinase Inhibitors in the Last Two Decades. Pharmaceuticals 2021, 14, 710. [Google Scholar] [CrossRef] [PubMed]
  158. Feneyrolles, C.; Spenlinhauer, A.; Guiet, L.; Fauvel, B.; Daydé-Cazals, B.; Warnault, P.; Chevé, G.; Yasri, A. Axl Kinase as a Key Target for Oncology: Focus on Small Molecule Inhibitors. Mol. Cancer Ther. 2014, 13, 2141–2148. [Google Scholar] [CrossRef] [Green Version]
  159. Myers, S.H.; Brunton, V.G.; Unciti-Broceta, A. AXL Inhibitors in Cancer: A Medicinal Chemistry Perspective: Miniperspective. J. Med. Chem. 2016, 59, 3593–3608. [Google Scholar] [CrossRef] [Green Version]
  160. McDaniel, N.K.; Cummings, C.T.; Iida, M.; Hülse, J.; Pearson, H.E.; Vasileiadi, E.; Parker, R.E.; Orbuch, R.A.; Ondracek, O.J.; Welke, N.B.; et al. MERTK Mediates Intrinsic and Adaptive Resistance to AXL-Targeting Agents. Mol. Cancer Ther. 2018, 17, 2297–2308. [Google Scholar] [CrossRef] [Green Version]
  161. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [Green Version]
  162. Martinelli, E.; Martini, G.; Cardone, C.; Troiani, T.; Liguori, G.; Vitagliano, D.; Napolitano, S.; Morgillo, F.; Rinaldi, B.; Melillo, R.M.; et al. AXL Is an Oncotarget in Human Colorectal Cancer. Oncotarget 2015, 6, 23281–23296. [Google Scholar] [CrossRef] [Green Version]
  163. Guinney, J.; Dienstmann, R.; Wang, X.; de Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The Consensus Molecular Subtypes of Colorectal Cancer. Nat. Med. 2015, 21, 1350–1356. [Google Scholar] [CrossRef] [PubMed]
  164. 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]
  165. Brandao, L.N.; Winges, A.; Christoph, S.; Sather, S.; Migdall-Wilson, J.; Schlegel, J.; McGranahan, A.; Gao, D.; Liang, X.; DeRyckere, D.; et al. Inhibition of MerTK Increases Chemosensitivity and Decreases Oncogenic Potential in T-Cell Acute Lymphoblastic Leukemia. Blood Cancer J. 2013, 3, e101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Graham, D.K.; Salzberg, D.B.; Kurtzberg, J.; Sather, S.; Matsushima, G.K.; Keating, A.K.; Liang, X.; Lovell, M.A.; Williams, S.A.; Dawson, T.L.; et al. Ectopic Expression of the Proto-Oncogene Mer in Pediatric T-Cell Acute Lymphoblastic Leukemia. Clin. Cancer Res. 2006, 12, 2662–2669. [Google Scholar] [CrossRef] [Green Version]
  167. Summers, R.J.; Ryan, J.; Minson, K.A.; Frye, S.V.; Wang, X.; Earp, H.S., III.; DeRyckere, D.; Graham, D.K. Mertk Is a Therapeutic Target in Early T-Precursor Acute Lymphoblastic Leukemia. Blood 2017, 130, 2536. [Google Scholar]
  168. Tormoen, G.W.; Blair, T.C.; Bambina, S.; Kramer, G.; Baird, J.; Rahmani, R.; Holland, J.M.; McCarty, O.J.T.; Baine, M.J.; Verma, V.; et al. Targeting MerTK Enhances Adaptive Immune Responses After Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 93–103. [Google Scholar] [CrossRef]
  169. 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]
  170. Taniguchi, H.; Yamada, T.; Wang, R.; Tanimura, K.; Adachi, Y.; Nishiyama, A.; Tanimoto, A.; Takeuchi, S.; Araujo, L.H.; Boroni, M.; et al. AXL Confers Intrinsic Resistance to Osimertinib and Advances the Emergence of Tolerant Cells. Nat. Commun. 2019, 10, 259. [Google Scholar] [CrossRef]
  171. Xie, S.; Li, Y.; Li, X.; Wang, L.; Yang, N.; Wang, Y.; Wei, H. Mer receptor tyrosine kinase is frequently overexpressed in human non-small cell lung cancer, confirming resistance to erlotinib. Oncotarget 2015, 6, 9206–9219. [Google Scholar] [CrossRef] [Green Version]
  172. Kim, D.; Bach, D.-H.; Fan, Y.-H.; Luu, T.-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] [Green Version]
  173. Elkabets, M.; Pazarentzos, E.; Juric, D.; Sheng, Q.; Pelossof, R.A.; Brook, S.; Benzaken, A.O.; Rodon, J.; Morse, N.; Yan, J.J.; et al. AXL Mediates Resistance to PI3Kα Inhibition by Activating the EGFR/PKC/MTOR Axis in Head and Neck and Esophageal Squamous Cell Carcinomas. Cancer Cell 2015, 27, 533–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Terry, S.; Abdou, A.; Engelsen, A.S.T.; Buart, S.; Dessen, P.; Corgnac, S.; Collares, D.; Meurice, G.; Gausdal, G.; Baud, V.; et al. AXL Targeting Overcomes Human Lung Cancer Cell Resistance to NK- and CTL-Mediated Cytotoxicity. Cancer Immunol. Res. 2019, 7, 1789–1802. [Google Scholar] [CrossRef] [PubMed]
  175. Terry, S.; Dalban, C.; Rioux-Leclercq, N.; Adam, J.; Meylan, M.; Buart, S.; Bougoüin, A.; Lespagnol, A.; Dugay, F.; Moreno, I.C.; et al. Association of AXL and PD-L1 Expression with Clinical Outcomes in Patients with Advanced Renal Cell Carcinoma Treated with PD-1 Blockade. Clin. Cancer Res. 2021, 27, 6749–6760. [Google Scholar] [CrossRef] [PubMed]
  176. Krebs, M.; Brunsvig, P.; Helland, Å.; Viñolas, N.; Aix, S.; Carcereny, E.; Gomez, M.D.; Perez, J.M.T.; Arriola, E.; Campelo, R.G.; et al. P1.01-72 A Phase II Study of Selective AXL Inhibitor Bemcentinib and Pembrolizumab in Patients with NSCLC Refractory to Anti-PD(L)1. J. Thorac. Oncol. 2019, 14, S388. [Google Scholar] [CrossRef]
  177. Oliva, M.; Chepeha, D.; Araujo, D.V.; Diaz-Mejia, J.J.; Olson, P.; Prawira, A.; Spreafico, A.; Bratman, S.V.; Shek, T.; de Almeida, J.; et al. Antitumor Immune Effects of Preoperative Sitravatinib and Nivolumab in Oral Cavity Cancer: SNOW Window-of-Opportunity Study. J. Immunother. Cancer 2021, 9, e003476. [Google Scholar] [CrossRef] [PubMed]
  178. Spicer, J.; Helland, Å.; Carcereny, E.; Arriola, E.; Gomez, M.D.; Trigo Perez, J.M.; Thompson, J.; Strauss, J.; Ortega Granados, A.L.; Felip, E.; et al. 362 A PhII Study of Bemcentinib, a First-in-Class Selective AXL Kinase Inhibitor, in Combination with Pembrolizumab in Pts with Previously-Treated Advanced NSCLC: Updated Clinical & Translational Analysis. Christie Sch. Oncol. 2020, A221. [Google Scholar]
  179. Colavito, S.A. AXL as a Target in Breast Cancer Therapy. J. Oncol. 2020, 2020, 5291952. [Google Scholar] [CrossRef] [Green Version]
  180. Zhang, S.; Xu, X.S.; Yang, J.X.; Guo, J.H.; Chao, T.F.; Tong, Y. The Prognostic Role of Gas6/Axl Axis in Solid Malignancies: A Meta-Analysis and Literature Review. OTT 2018, 11, 509–519. [Google Scholar] [CrossRef] [Green Version]
  181. 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]
  182. Schoumacher, M.; Burbridge, M. Key Roles of AXL and MER Receptor Tyrosine Kinases in Resistance to Multiple Anticancer Therapies. Curr. Oncol. Rep. 2017, 19, 19. [Google Scholar] [CrossRef] [Green Version]
  183. Caraux, A.; Lu, Q.; Fernandez, N.; Riou, S.; Di Santo, J.P.; Raulet, D.H.; Lemke, G.; Roth, C. Natural Killer Cell Differentiation Driven by Tyro3 Receptor Tyrosine Kinases. Nat. Immunol. 2006, 7, 747–754. [Google Scholar] [CrossRef] [PubMed]
  184. Nandrot, E.F.; Dufour, E.M. Mertk in Daily Retinal Phagocytosis: A History in the Making. In Retinal Degenerative Diseases; Advances in Experimental Medicine and Biology; Anderson, R.E., Hollyfield, J.G., LaVail, M.M., Eds.; Springer: New York, NY, USA, 2010; Volume 664, pp. 133–140. ISBN 978-1-4419-1398-2. [Google Scholar]
  185. Inoue, S.; Yamane, Y.; Tsukamoto, S.; Azuma, H.; Nagao, S.; Murai, N.; Nishibata, K.; Fukushima, S.; Ichikawa, K.; Nakagawa, T.; et al. Discovery of a Potent and Selective Axl Inhibitor in Preclinical Model. Bioorganic Med. Chem. 2021, 39, 116137. [Google Scholar] [CrossRef] [PubMed]
  186. Vollrath, D.; Yasumura, D.; Benchorin, G.; Matthes, M.T.; Feng, W.; Nguyen, N.M.; Sedano, C.D.; Calton, M.A.; LaVail, M.M. Tyro3 Modulates Mertk-Associated Retinal Degeneration. PLoS Genet. 2015, 11, e1005723. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Different strategies of inhibition of TAM receptor activity. Abbreviations: GAS6, Growth arrest-specific 6 ligand; PROS1, Protein S; IgG, immunoglobulin G antibodies; scFv, single-chain variable fragment antibodies; ADC, antibody–drug conjugates; LMW, low-molecular-weight compounds; TKI, tyrosine kinase inhibitors; IgG-like domains, immunoglobulin-like domains; FNIII domains, fibronectin type III domains; AA, amino acid; ECD, extracellular domain; KD, kinase domain.
Figure 1. Different strategies of inhibition of TAM receptor activity. Abbreviations: GAS6, Growth arrest-specific 6 ligand; PROS1, Protein S; IgG, immunoglobulin G antibodies; scFv, single-chain variable fragment antibodies; ADC, antibody–drug conjugates; LMW, low-molecular-weight compounds; TKI, tyrosine kinase inhibitors; IgG-like domains, immunoglobulin-like domains; FNIII domains, fibronectin type III domains; AA, amino acid; ECD, extracellular domain; KD, kinase domain.
Cancers 14 02488 g001
Figure 2. Different structures of TAM family kinase inhibitors. TAM receptors are divided into three groups, based on their base core structure: CORE-A compounds, with hydrogen bond acceptor–substituted phenyl group linked to a hinge-binding heterocycle with a solubilising group; CORE-B compounds, with heterocycle (hinge-binding) optionally ortho-fluoro phenyl group (binding DFG motif), 2–4 hydrogen donors/acceptors, and a phenyl group (or para-fluoro phenyl) (binding to allosteric hydrophobic pockets); and CORE-X compounds, that cannot be clearly assigned to either -A, or -B groups, as they present only partial structural similarity to other cores, or present completely different lead structures. Compounds approved for treatment are presented in bold, compounds currently in clinical trials are presented in italics, and compounds that did not enter the clinical phase are presented in plain text.
Figure 2. Different structures of TAM family kinase inhibitors. TAM receptors are divided into three groups, based on their base core structure: CORE-A compounds, with hydrogen bond acceptor–substituted phenyl group linked to a hinge-binding heterocycle with a solubilising group; CORE-B compounds, with heterocycle (hinge-binding) optionally ortho-fluoro phenyl group (binding DFG motif), 2–4 hydrogen donors/acceptors, and a phenyl group (or para-fluoro phenyl) (binding to allosteric hydrophobic pockets); and CORE-X compounds, that cannot be clearly assigned to either -A, or -B groups, as they present only partial structural similarity to other cores, or present completely different lead structures. Compounds approved for treatment are presented in bold, compounds currently in clinical trials are presented in italics, and compounds that did not enter the clinical phase are presented in plain text.
Cancers 14 02488 g002
Table 1. TAM family kinase domain inhibitors in cancer therapy.
Table 1. TAM family kinase domain inhibitors in cancer therapy.
InhibitorStatus 1CoreTypeInhibitory ParametersReferences
Bosutinib;
SKI-606; PF5208763;
Bosulif
APPROVEDAIAXL IC50 = 174 nM
MERTK IC50 = 110 nM
TYRO3 IC50 = 61 nM
[117,118,119]
Gilteritinib;
ASP2215; Xospata
APPROVEDAIAXL IC50 = 0.73 nM
MERTK IC50 = 5 nM
[120]
Vandetanib;
ZD6474; Caprelsa
APPROVEDAIAXL IC50 = 250 nM
MERTK IC50 = 1400 nM
TYRO3 IC50 = 93 nM
[119]
Cabozantinib;
XL 184; BMS-907351; Cabometyx
APPROVEDBIIAXL IC50 = 7 nM[121]
BGB324;
R428;
Bemcentinib
APPROVEDXIAXL IC50 = 14 nM[122]
Crizotinib
PF-02341066; Xalkori
APPROVEDXIAXL IC50 = 294 nM[123]
Sunitinib;
SU 11248; Sutent
APPROVEDXIAXL IC50 = 9 nM[124]
TP-0903;
Dubermatinib
CLINICAL TRIALS
NCT04518345
AIAXL IC50 = 27 nM[125]
BMS777607;
ASLAN002
CLINICAL TRIALS
NCT01721148
NCT00605618
BIIAXL IC50 = 1.1 nM
MERTK IC50 = 14 nM
TYRO3 IC50 = 4.3 nM
[126]
BPI-9016MCLINICAL TRIALS
NCT02929290
NCT02478866
BIIAXL IC50 = 9 nM[127]
DS-1205b/cCLINICAL TRIALS
NCT03599518
NCT03255083
(TERMINATED)
BIIAXL IC50 = 1.3 nM
MERTK IC50 = 63 nM
[128]
Foretinib;
XL880;
GSK1363089
CLINICAL TRIALS
NCT00920192
NCT01147484
NCT01138384
NCT00742131
NCT00725764
NCT00726323
NCT00725712
NCT00743067
NCT01068587
BIIAXL IC50 = 11 nM[129]
Merestinib; LY2801653CLINICAL TRIALS
NCT03125239
NCT03027284
NCT02779738
NCT02745769
BIIAXL IC50 = 2 nM
MERTK IC50 = 10 nM
[125,130]
MGCD265;
Glesatinib
CLINICAL TRIALS
NCT02954991
BIIn/a
Ningetinib; CT053PTSACLINICAL TRIALS
NCT04577703
NCT03758287
BIIAXL IC50 < 1.0 nM[131]
ONO-7475CLINICAL TRIALS
NCT03176277
BIIAXL IC50 = 0.7 nM
MERTK IC50 = 1 nM
TYRO3 IC50 = 1.9 nM
[132]
PF-07265807;
ARRY-067; PF-5807
CLINICAL TRIALS
NCT04458259
BIIn/a
RXDX-106;
CEP-40783
CLINICAL TRIALS
NCT03454243 (TERMINATED)
BIIAXL IC50 = 0.31 nM
MERTK IC50 = 1.89 nM
TYRO3 IC50 = 3.5 nM
[133]
Sitravatinib;
MGCD516
CLINICAL TRIALS
NCT04123704
NCT03575598
NCT04472650
NCT04772612
NCT04921358
NCT04727996
NCT04800614
NCT04904302
NCT04925986
NCT05176925
NCT05104801
NCT05255276
NCT04887194
NCT04734262
NCT02954991
NCT03606174
NCT04887870
BIIAXL IC50 = 1.5 nM
MERTK IC50 = 2 nM
[134]
XL092CLINICAL TRIALS
NCT03845166
NCT05176483
BIIAXL IC50 = 3.4 nM
MERTK IC50 = 7.2 nM
[135]
Amuvatinib;
MP470
CLINICAL TRIALS
NCT01357395
NCT00894894
NCT00881166
XIAXL IC50 = 10 nM[57]
MRX-2843;
UNC2371
CLINICAL TRIALS
NCT03510104
NCT04762199
NCT04872478
XIAXL IC50 = 15 nM
MERTK IC50 = 1.3 nM
TYRO3 IC50 = 17 nM
[136]
S49076CLINICAL TRIALS
ISRCTN11619481
XIAXL IC50 = 7 nM
MERTK IC50 = 2 nM
[137]
SNS314CLINICAL TRIALS
NCT00519662
XIAXL IC50 = 84 nM[138]
Rebastinib;
DCC-2036
CLINICAL TRIALS
NCT00827138
XIIAXL IC50 = 42 nM[139]
SLC-391CLINICAL TRIALS
NCT05278845
NCT03990454
Xn/aAXL IC50 = 9.6 nM
MERTK IC50 = 42.3 nM
TYRO3 IC50 = 44 nM
[140]
INCB081776CLINICAL TRIALS
NCT03522142
n/an/aAXL IC50 = 0.61 nM
MERTK IC50 = 3.17 nM
TYRO3 IC50 = 101 nM
[141]
Q702CLINICAL TRIALS
NCT04648254
n/an/an/a
SGI7079PRECLINICALAIAXL IC50 = 58 nM[142]
SK-G-801;
G-801
PRECLINICALAIAXL IC50 = 20 nM[143]
6g; purine analogue of BMS777607PRECLINICALBIIAXL IC50 = 39 nM
MERTK IC50 = 42 nM
TYRO3 IC50 = 65 nM
[144]
LDC1267PRECLINICALBIIAXL IC50 = 29 nM
MERTK IC50 = 5 nM
TYRO3 IC50 = 8 nM
[47]
NPS-1034PRECLINICALBIIAXL IC50 = 10.3 nM[145]
TAM-IN-2PRECLINICALBIIn/a
UNC2541PRECLINICALBIIMERTK IC50 = 4.4 nM[146]
UNC2881PRECLINICALBIIAXL IC50 = 360 nM
MERTK IC50 = 4.3 nM
TYRO3 IC50 = 250 nM
[147]
2-D08PRECLINICALXIAXL IC50 = 0.49 nM[148]
UNC1062PRECLINICALXIAXL IC50 = 85 nM
MERTK IC50 = 1.1 nM
TYRO3 IC50 = 60 nM
[23,149]
UNC2025PRECLINICALXIAXL IC50 = 14 nM
MERTK IC50 = 0.7 nM
TYRO3 IC50 = 18 nM
[150]
Compound 19PRECLINICALXn/aTYRO3 IC50 = 10 nM[151]
Abbreviations: n/a, not available; IC50, half-maximal inhibitory concentration. 1 The table lists NCT/ISRCTN clinical trials numbers only for completed or recruiting trials. For RXDX-106, the NCT number is given for terminated study (sponsor’s decision) as it was the only clinical trial for this compound.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mikolajczyk, A.; Mitula, F.; Popiel, D.; Kaminska, B.; Wieczorek, M.; Pieczykolan, J. Two-Front War on Cancer—Targeting TAM Receptors in Solid Tumour Therapy. Cancers 2022, 14, 2488. https://doi.org/10.3390/cancers14102488

AMA Style

Mikolajczyk A, Mitula F, Popiel D, Kaminska B, Wieczorek M, Pieczykolan J. Two-Front War on Cancer—Targeting TAM Receptors in Solid Tumour Therapy. Cancers. 2022; 14(10):2488. https://doi.org/10.3390/cancers14102488

Chicago/Turabian Style

Mikolajczyk, Agata, Filip Mitula, Delfina Popiel, Bozena Kaminska, Maciej Wieczorek, and Jerzy Pieczykolan. 2022. "Two-Front War on Cancer—Targeting TAM Receptors in Solid Tumour Therapy" Cancers 14, no. 10: 2488. https://doi.org/10.3390/cancers14102488

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