Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 4.
Published in final edited form as: Crit Rev Oncog. 2011;16(1-2):47–63. doi: 10.1615/critrevoncog.v16.i1-2.60

TAM Receptors in Leukemia: Expression, Signaling, and Therapeutic Implications

Luis Brandão 1, Justine Migdall-Wilson 1, Kristen Eisenman 1, Douglas K Graham 1,*
PMCID: PMC4855299  NIHMSID: NIHMS492406  PMID: 22150307

Abstract

In the past 30 years there has been remarkable progress in the treatment of leukemia and lymphoma. However, current treatments are largely ineffective against relapsed leukemia and, in the case of pediatric patients, are often associated with severe long-term toxicities. Thus, there continues to be a critical need for the development of effective biologically targeted therapies. The TAM family of receptor tyrosine kinases—Tyro3, Axl, and Mer—plays an important role in normal hematopoiesis, including natural killer cell maturation, macrophage function, and platelet activation and signaling. Furthermore, TAM receptor activation leads to upregulation of pro-survival and proliferation signaling pathways, and aberrant TAM receptor expression contributes to cancer development, including myeloid and lymphoid leukemia. This review summarizes the role of TAM receptors in leukemia. We outline TAM receptor expression patterns in different forms of leukemia, describe potential mechanisms leading to their overexpression, and delineate the signaling pathways downstream of receptor activation that have been implicated in leukemogenesis. Finally, we discuss the current research focused on inhibitors against these receptors in an effort to develop new therapeutic strategies for leukemia.

Keywords: leukemia, Axl, Mer, Tyro-3, signal transduction, gene expression, targeted, therapeutics

I. INTRODUCTION

Cancer is the second leading cause of death in the United States, resulting in an estimated 23.2% of all deaths in 2007. While leukemia and lymphoma only account for approximately 7.6% of these deaths (43,370 in 2010), leukemia is the primary cause of cancer-related death in males under 40 and females under 20 years old. Furthermore, cancer is the leading cause of disease-related deaths in children (1–14 years old), and leukemia is the most common pediatric malignancy.13

Between 1975 and 2003, the 5-year relative survival for all leukemia patients has increased significantly from 33.4% to 55.6%. This trend is even more dramatic for children with leukemia, where relative survival rates have increased from 43% to 94.6% in cases of acute lymphoblastic leukemia (ALL) and from 18.7% to 56.1% in cases of acute myeloid leukemia (AML).3 Although much of this improvement stems from changes in treatment regimens and dosing during induction and subsequent courses of therapy, the current success is associated with a 2- to 4-fold increased rate of severe therapy-associated health conditions, including organ damage, infertility, growth decline, reduced mental function, and secondary malignancy.4,5 Furthermore, current treatment protocols are largely ineffective against relapsed leukemia,6 highlighting the need for new antileukemic therapeutic agents.

The TAM family of receptor tyrosine kinases (RTKs) includes Tyro-3, Axl, and Mer. In normal hematopoiesis, TAM receptors inhibit inflammation in dendritic cells and macrophages, promote phagocytosis of apoptotic cells and membranous organelles, and are essential for natural killer (NK) cell maturation.7 The TAM family is also vital for platelet activation, platelet signaling during thrombus stabilization,8,9 and may have an important role in erythropoiesis.10 In addition to their role in normal hematopoiesis, TAM receptors can activate proliferation- and survival-promoting signaling pathways such as those driven by AKT and ERK1/2, which contribute to oncogenesis in multiple cancers, including myeloid and lymphoid leukemia.11

The goal of this review is to summarize the role of the TAM family in leukemia. Because little published data exist on Tyro-3 in hematopoietic malignancy, we have largely focused on Mer and Axl receptor tyrosine kinases. Throughout the discussion, we address leukemia-associated expression patterns observed for TAM receptors, explore potential mechanisms underlying their aberrant expression, and outline the downstream signaling pathways implicated in leukemogenesis. In the final part of our review, we detail the ongoing efforts to develop therapeutic agents against this family of receptors and discuss how specifically targeting TAM receptors can potentially enhance leukemia therapy.

II. TAM RECEPTOR EXPRESSION PATTERNS IN LEUKEMIA

While macrophages, dendritic cells, NK cells, NKT cells, megakaryocytes, and platelets normally express TAM receptors,7,8,10,12,13 they are not expressed in thymocytes, mature T- or B-lymphocytes, or granulocytes.14 However, TAM receptors display altered expression patterns in leukemia. Below and in Table 1, we compare TAM receptor expression in different subsets of leukemia.

TABLE 1.

Expression of TAM Receptors in Leukemia and in the Hematopoietic System

Leukemia Tyro-3 Axl Mer Gas6 Protein S References
Myeloid
    CML Pos Pos Pos 17, 18, 21, 102
    AML Pos Pos Pos Pos 15, 20, 21, 23, 28, 102
    Myeloblastic leukemia Pos Pos Pos 20, 28, 102
    Monoblastic leukemia Pos Pos Pos 20, 28, 102
    Erythroid leukemia Pos Pos Pos 20, 28, 102
    Megakaryoblastic leukemia Pos Pos Pos 20, 28, 102
Lymphoblastic
    ALL Neg Pos Pos Pos 14, 20, 21, 2527
    CLL Pos Pos 21, 95, 102
    Plasma cell leukemia Pos 16
Normal hematopoiesis
    Lymphocytes Neg Neg 14, 20, 21
    Granulocytes Neg 21
    Basophil/Mast cells Pos 7
    Macrophages Pos Pos Pos Pos Pos 7, 13, 14, 21, 103
    Megakaryocytes/platelets Pos Pos Pos Pos 7, 14, 104106
    NK cells, NKT cells Pos Pos Pos Neg Pos 7, 13
    Dendritic cells Pos Pos Pos Pos Pos 7, 13
    Bone marrow Neg Pos Pos Pos 14, 22, 106, 107
    Erythroid cells Neg Pos Pos Pos 10, 108

A. Tyro-3

Tyro-3 (Dtk/Sky/Rse/Brt/Tif) RNA was identified in blasts in 6 of 11 AML patients by RNase protection analysis.15 More recently, a gene expression microarray found Tyro-3 overexpression in multiple myeloma samples relative to autologous B-lymphoblastoid cell lines, and Tyro-3 mRNA transcript was also detected in primary malignant plasma cells from patients with plasma cell leukemia or multiple myeloma.16

B. Axl

Axl (Ufo/Jtk11) was first detected in 1988 as an unidentified gene promoting the transition from chronic phase to blast crisis in two patients with chronic myelogenous leukemia (CML).17 Three years later, two independent groups cloned the human gene from patients with CML and chronic myeloproliferative disorder.18,19 Axl mRNA expression was identified in a large number of cell lines derived from myeloid and erythromegakaryocytic leukemias and was notably absent in lymphocytic cell lines.20 Patient sample analysis confirmed these cell line results, revealing Axl transcript in 56/99 (56.6%) patients with myeloproliferative disorders (AML, CML in chronic phase and in blast crisis, and myelodysplasia), but in only 2.2% of lymphoid leukemias: out of 45 samples, only one patient with chronic lymphocytic leukemia (CLL) had detectable Axl mRNA.21,22 Similarly, Rochlitz et al. identified Axl transcript in 19 of 54 (35%) AML patient samples, observing that patients with increased CD34 expression also displayed higher levels of Axl, and they determined that Axl expression was associated with worse progression-free and overall survival when adjusted for age, Auer rods, and leukocyte counts.23 Lastly, a recent study found increased Axl transcript in myeloid leukemia samples from chemotherapy-resistant patients; furthermore, chemotherapy induced Axl expression in an AML cell line, and addition of Gas6—an Axl ligand—enhanced drug resistance in cells during chemotherapeutic treatment.24

C. Mer

The human Mer gene (MerTK/RP38/Nyk/Tyro12) was initially cloned from a B-lymphoblastoid expression library.14 While normal T- and B-lymphocytes do not express Mer mRNA transcript or protein at any developmental stage,25,26 our lab has detected ectopic Mer mRNA expression in 19/34 (55.8%) patient samples, and Mer protein expression in 8/16 (50%) pediatric T-ALL patient samples.25 Furthermore, a large-scale microarray analysis demonstrated that significantly high levels of Mer transcript exist in the cytogenetic subset of B-ALL patients expressing the E2A-PBX1+ fusion protein.27 Consistently, our lab has identified aberrant Mer protein expression in 16/16 E2A-PBX1+ B-ALL patient samples, whereas 11/12 B-ALL samples without E2A-PBX1 did not express Mer.26 To date, there are no published reports on the role of Mer in myeloid leukemia, but we have detected increased Mer expression in 11/16 AML cell lines and in 17/26 primary patient samples by western blot and flow cytometry.28

The role of Mer in leukemogenesis is further supported by two animal models. Abnormal activation of Eyk, the chicken homologue of Mer, via the naturally occurring RPL30 avian retrovirus, leads to the development of a spectrum of cancers, including lymphomas, in chickens.29 Additionally, ectopic Mer expression in lymphocytes in the Mer transgenic mouse increases the incidence of leukemia/lymphoma.30

III. UPSTREAM REGULATION OF TAM RECEPTOR EXPRESSION

TAM receptor overexpression occurs in many cancers of myeloid lineage, and ectopic expression of Mer, which normal lymphocytes do not express, is found in mantle cell lymphoma, the majority of T cell leukemias, and particular subsets of B cell leukemia.25,27 Although aberrant TAM receptor levels clearly enhance oncogenic potential, much remains unknown about the mechanisms underlying their overexpression. Several studies have begun to explore epigenetic and post-transcriptional regulation of TAM receptor expression, providing us with further insight into the tangled circuitry of cancer progression. Although the studies presented here have been conducted in a variety of systems, their findings may also apply to processes within hematopoietic development and leukemogenesis, which are depicted in Figure 1.

FIGURE 1.

FIGURE 1

Experimentally determined regulators of TAM receptor and ligand gene expression. Nuclear modulators include transcription factors, histone acetylation, promoter methylation, and gene amplification. Outside of the nucleus, several post-transcriptional processes influence protein formation: miRNAs repress translation of Axl and HIF-1a, potentially altering both MER and AXL transcription; YB-1, an RNA-binding protein, inhibits Mer translation unless it is phosphorylated by AKT, a downstream target of Mer activation.

A. Genetic Variation

To date, no activating mutations in the TAM receptor genes have been implicated in malignant transformation, but recent studies have highlighted the potential role of copy number variation in TAM receptor expression. AXL gene amplification and corresponding overexpression of its transcript were found in a CGH-based microarray profiling study of glioblastoma samples,31 and gastric cancer samples displayed increased AXL and MER copy numbers relative to normal controls.32 Additionally, DNA copy number analysis identified AXL gene amplification in 4/4 lapatinib-resistant breast cancer cell lines,33 and GAS6 amplification has also been detected in aggressive mouse mammary tumors.34

Analysis of the Axl transcript has also shown that two alternatively spliced isoforms are expressed in tumor and normal samples at different ratios. However, both isoforms have the same transforming capability, suggesting that receptor overexpression— rather than a structural difference in the transcript or protein—drives the oncogenicity of this receptor.18

B. Transcriptional Regulation

While several putative transcription factors for the TAM receptor genes have been identified based on promoter binding site specificity, gene expression modulation has been most extensively studied in Axl, as it is the only human TAM receptor for which the gene promoter has been fully characterized. Multiple studies have found that AP-2α, Sp1/Sp3 and MZF-1 directly regulate Axl transcription, with MZF-1 levels directly correlating with Axl expression and metastasis in colorectal and cervical cancers.18,3537 More recently, CXCR4/SDF-1 (CXCL12) has been shown to increase transcription of both AXL and TYRO3 in thyroid carcinoma cell lines; although the transcriptional interaction was not further characterized, treatment with a CXCR4 inhibitor did not reduce constitutive Axl expression, suggesting that its overexpression requires additional regulatory mechanisms.38 Another study found that Gas6, the common ligand for both Axl and Mer, was transcriptionally upregulated following progesterone receptor activation in breast cancer cells.39 A complete list of transcription factors and their interactions with the TAM receptor genes has been compiled in Table 2.

TABLE 2.

Transcriptional Regulators of TAM Receptor Expression

Gene Interaction Transcription Factor Effect Cell Type Reference
Tyro-3 Indirect NGF Rat neuronal cells 109
Tyro-3 Indirect CXCR4/SDF-1 (CXCL12) Thyroid carcinoma 38
Axl Direct AP-2a HeLa, 293T, NIH3T3, NSC-34 37
Axl Direct Sp1/Sp3 Rko, HCT116, HeLa 36
Axl Direct MZF-1 Colorectal, cervical cancer 110
Axl Indirect CXCR4/SDF-1 (CXCL12) Thyroid carcinoma 38
Axl Indirect HIF-1a Endothelial cells 111
Axl Indirect Net/Elk3 Endothelial cells 112
Axl Indirect NGF Rat neuronal cells 109
Axl Indirect Prox-1 Blood endothelial cells 113
Axl Indirect E1A Breast cancer 35
Axl Predicted AP-1 24
Axl Predicted C/EBPb 24
Axl Predicted p300 24
Axl Predicted CREB 24
Mer Direct Sp1 ↑, ↓ Mouse Sertoli cells 40
Mer Direct LXR Macrophages 41
Mer Direct Sp3 Mouse Sertoli cells 40
Mer Direct E2F Mouse Sertoli cells 40
Mer Indirect BRLF1 (R) Several 114
Mer Indirect PAX-FKHR Mouse MSCs 115
Mer Indirect GR Mouse MSCs 44
Mer Indirect Net/Elk3 Endothelial cells 112
Mer Indirect HIF-1a Endothelial cells 112
Mer Predicted GATA 40
Mer Predicted MZF-1 40
Gas6 Direct ER-a Breast cancer, normal
mammary
116
Gas6 Indirect PR-B Breast cancer 39

Although the human Mer promoter remains completely uncharacterized, Wong et al. identified Sp1, Sp3, and E2F as transcriptional regulators of the mouse Mertk gene,40 which shares considerable homology with its human counterpart. Liver X receptor (LXR) was recently shown to directly bind the Mertk promoter and induce its transcription in mouse macrophages (without noticeable effects on Axl or Tyro-3 expression), which promoted phagocytosis of apoptotic cells and maintained immunity.41 Another study found that CLL cells display increased LXR expression relative to normal lymphocytes, but the potential relationship with Mer expression was not investigated.42 However, disruption of normal LXR expression in lymphocytes causes age-dependent lymphoid hyperplasia in mouse models,43 findings similar to those observed upon ectopic Mer expression in transgenic mouse lymphocytes.30

Mer has also been identified as a glucocorticoid-responsive gene in mouse mesenchymal stem-like cells: dexamethasone treatment led to a 4.6-fold increase in Mer expression relative to vehicle-treated cells, and ChIP-chip analyses revealed several glucocorticoid binding sequences upstream of the Mertk transcription start site.44 Another study found a similar trend in dexamethasone-induced Mer protein expression on the surface of cultured human macrophages,45 indicating that Mer upregulation may decrease responsiveness to ALL induction therapy, which involves glucocorticoid treatment. Consistently, our laboratory has found increased Mer expression in patients with relapsed leukemia (Eisenman K and Graham DK, unpublished data), suggesting that Mer may serve as a survival or resistance mechanism in leukemic cells following treatment.

Although Mer is upregulated in the E2A-PBX1+ cytogenetic subset of B-ALL patients,26,27,46 expression of the fusion protein does not appear to induce Mer transcription (Sawczyn K and Graham DK, unpublished data) despite the presence of a PBX1-binding site near the Mer promoter.47 However, the PBX domain of the fusion protein is directly responsible for inducing apoptosis in hematopoietic precursor B cell lines,48 consistent with the idea that Mer expression arises as a survival response rather than directly through E2A-PBX1-driven changes in gene transcription.

C. Epigenetic Regulation

Upon identifying Axl as the only consistently upregulated protein tyrosine kinase in drug-resistant AML patient samples, Hong et al. found that treatment of an AML cell line with a chemotherapeutic agent—doxorubicin, cisplatin, or VP16—increased Axl expression, but only when its promoter remained unmethylated.24 Furthermore, a separate study correlated promoter hypomethylation with the degree of Axl overexpression in Kaposi sarcoma: cell lines expressing high levels of Axl had fewer methylated sites, whereas other lines expressing less Axl—including some derived from other cancer types—displayed more promoter methylation.49 Lastly, it was shown that SAHA, a histone deacetylase (HDAC) inhibitor, suppresses Gas6 expression in multiple myeloma cells,50 highlighting another potential mechanism regulating transcription of TAM receptor-related genes.

D. Post-Transcriptional Regulation

Currently, miR-335 is the only microRNA reported to target the Mer 3′UTR.51 However, the study, which compared expression differences between metastatic and non-metastatic breast cancer lines, only used indirect target-validation methods and never assessed Mer protein expression following manipulation of miR-335 levels. In functional studies of miRNAs in leukemic cells, restoration of miR-335 expression levels with a synthetic mimic did not decrease Mer protein expression (Migdall-Wilson J and Graham DK, unpublished data).

Both miR-155 and miR-34a decrease Axl expression in human monocytes.52 miR-155, which promotes tumor growth in numerous leukemias and lymphomas when overexpressed, 53 represses translation of several transcription factors that potentially regulate Axl and Mer expression, including PU.1, CEBPb, CSF1R, and HIF-1α. In addition to showing that miR-34a and miR-199a/b target the Axl 3′UTR, a recent study also determined that both miRNA genes and AXL are regulated by promoter ethylation.54

Evdokimova et al. found that YB-1, a regulatory RNA-binding protein, normally inhibits translation of Mer mRNA; however, in conditions requiring increased expression of stress- and growth-related proteins, Mer translation is de-repressed upon AKT-mediated phosphorylation.55 While this mechanism has not been explored in hematopoietic cells, the fact that AKT is a well-known downstream target of Mer raises the possibility of a positive-feedback loop occurring through this post-transcriptional mechanism.

IV. MER AND AXL RECEPTOR SIGNALING IN LEUKEMIA

Mer and Axl activate many different signaling pathways depending on cell type and function, a topic extensively discussed in a review previously published by our lab.11 In this section, we specifically focus on the Mer- and Axl-activated pathways known to play a role in leukemogenesis (Figure 2).

FIGURE 2.

FIGURE 2

Mer/Axl signaling in leukemia. Receptor activation by Gas6 or Protein S leads to activation of MAPK, PI3K/AKT and Jak/STAT pathways, resulting in increased proliferation and survival. Jak/STAT activation has been observed both via direct interaction between Jak and Mer, as well as through interaction between Axl and the type 1 interferon receptor (IFNAR1).

A. Ligands

Gas6 and Protein S are two structurally similar, vitamin K-dependent proteins that activate the TAM receptors.56,57 Both ligands are produced in a wide range of tissues, including the bone marrow, thymus, spleen, and plasma,56,58,59 suggesting that leukemia cells expressing Mer or Axl are constitutively activated through a paracrine mechanism. Consistent with this idea, Gas6 expression has been correlated with the ability of bone marrow stromal cell lines to support hematopoiesis,60 and primary human osteoblasts have been shown to secrete Gas6 in response to Mer-expressing leukemic cells.61 Furthermore, overexpression of Gas6 and Protein S has been reported in several cancers and often correlates with poor prognostic markers.62

Tubby and Tubby-like protein 1 (Tulp1), which also bind Axl and Tyro-3, were recently described as two new Mer ligands important in phagocytosis.63 It is unclear whether these two ligands play a role in leukemogenesis.

B. MAPK (ERK) Pathway

Before the TAM receptor ligands had been identified, initial studies on Mer and Axl downstream signaling implemented chimeric receptors6466 composed of the Axl or Mer intracellular kinase domain fused to an extracellular domain of a receptor with a known ligand. In a study using a colony stimulating factor 1 (CSF1) receptor/Mer chimeric receptor, stimulation with CSF1 led to phosphorylation of Shc, recruitment of Grb2 and downstream phosphorylation of MEK and ERK1/2 kinases, indicating MAPK pathway activation.66 These results were later confirmed with a CD8/Mer chimeric receptor in BaF3 cells,65 and eventually in experiments using the full-length Mer receptor and recombinant Gas6 for activation of the receptor in 293 cells.67 More recently, our lab demonstrated that Mer expression mediates chemotherapy-induced activation of ERK1/2 in the 697 (human B-ALL) cell line,26 and related studies showed that Axl stimulation activates the ERK1/2 kinases through Shc, Grb2, and Ras.64,68 Interestingly, results of other investigations have suggested that Axl-mediated ERK1/2 activation is specifically required to induce proliferation and depends on PI3K/AKT and Src activity.69,70

C. PI3K/AKT/mTOR

Many studies exploring downstream activation of the MAPK pathway have also identified the PI3K/AKT kinase cascade as an important component of Axl/Mer signaling.64,66,67,69 Several groups showed that the p85 subunit of PI3K binds to both Mer and Axl, and determined that this interaction requires both ligand binding and kinase activity.64,68,71

In response to Axl stimulation, PI3K/AKT pathway activation is important for both cell proliferation—via ERK1/2 activation—as well as survival, which occurs through phosphorylation of Bad, a Bcl-2 family member;69,70 furthermore, this antiapoptotic effect appears to be independent of MAPK activity.69 Interestingly, studies investigating how Mer stimulation affects the PI3K/AKT pathway were less conclusive, partly because they were performed in different cell types: in the Ba/F3 lymphoid cell line, Mer-mediated PI3K activation increased cell survival upon IL-3 withdrawal and minimally impacted cell proliferation.65 However, in the 32D monocytic cell line, Mer-mediated PI3K activation alone did not sufficiently prevent apoptosis; instead, investigators found that the MAPK and PI3K pathways play parallel roles: inhibition of both cascades was necessary to elicit growth factor withdrawal–induced cell death.71

mTOR kinase, a key regulator of protein synthesis and cell growth, is one of the downstream effectors of the PI3K pathway. Goruppi et al. showed that rapamycin, an mTOR inhibitor, blocked Axl-mediated proliferation and survival in NIH 3T3 cells.69 Furthermore, S6 kinase (S6K)—a downstream target of mTOR—is activated upon stimulation of a Mer chimeric receptor in NIH/3T3 cells.66

D. NF-kB

The NF-kB family of transcription factors is a key regulator of cell growth, development, and survival. Aberrant NF-kB activation has been observed in various forms of leukemia/lymphoma72 and has also been implicated downstream of Mer and Axl/Gas6 signaling:65,73,74 luciferase reporter assays revealed a 10-fold increase in NF-kB transcriptional activity in Ba/F3 cells expressing a constitutively active chimeric Mer receptor, along with more robust proliferation than vector-only control cells.65 In NIH/3T3 cells, Gas6 induced a transient decrease in the level of IkB, which binds to and inhibits NF-kB, resulting in enhanced NF-kB DNA-binding activity and an NF-kB–dependent increase in expression of Bcl-xL, an anti-apoptotic protein.73 Likewise, expression of a dominant negative IkB—which keeps NF-kB in a permanently bound, inactive state—inhibited Gas6-mediated survival, further implying that the anti-apoptotic effects downstream of TAM receptor activation require NF-kB activity.73 Lastly, NF-kB activation depends upon PI3K/AKT activation, suggesting that extensive cross-talk exists between the various pathways downstream of Mer or Axl.65,73

E. Jak/STAT Pathway

Constitutive activation of STAT proteins has been observed both in AML and ALL, as well as in a variety of other tumors (reviewed in Benekli et al.75). Jak/STAT activation arises through various mechanisms, including cytokine overexpression and autocrine/paracrine stimulation,76 as well as expression of a TEL-JAK2 fusion protein.77

Both constitutive Jak activation and STAT phosphorylation have been associated with Mer and Axl activity. COS cells expressing a constitutively active chicken Mer receptor (CD8-Eyk chimera) displayed constitutive activation of STAT1, STAT3, and Jak1; the CD8-Eyk chimera also co-immunoprecipitated with Jak1.78 More recently, Gas6-induced Axl stimulation led to STAT1 activation in mouse bone marrow dendritic cells, an effect dependent upon type I interferon (IFN) receptor expression.79 Interestingly, Axl co-immunoprecipitated with the IFN receptor R1 chain, suggesting a possible mechanism for TAM receptor activation of the Jak/STAT pathway.

V. THERAPEUTIC TARGETING

Several novel biologically targeted therapies have shown great promise in improving therapeutic outcomes in leukemia. The most striking examples are imatinib, dasatinib, and nilotinib, tyrosine kinase inhibitors used to treat CML and Philadelphia chromosome-positive (Ph+) ALL.80 Imatinib therapy resulted in a 73.8% rate of complete cytogenetic response after 19 months, compared with 8.5% for patients on earlier standards of care.81 Other examples include lestaurtinib, a FLT3 tyrosine kinase inhibitor currently in clinical trials for treatment of MLL-rearranged ALL, and rituximab, a monoclonal antibody against CD20 currently used in therapy for CD20+ leukemia/lymphoma.82,83

Mer and Axl represent two novel targets for the development of new anti-leukemia therapies, and multiple studies of receptor inhibition highlight their potential within this realm. Mer inhibition significantly delays leukemia progression in a human ALL cell line xenograft (Brandão and Graham, unpublished data), and Axl inhibition reduces growth of lung and breast cancer xenograft tumors in immuno compromised mice.84 Furthermore, inhibition of Mer dampens pro-survival pathway activation in a human B-ALL cell line and renders it more sensitive to a spectrum of chemotherapeutic agents currently used in the clinic.26 Similarly, Mer and Axl inhibition also increases apoptosis and chemosensitivity in astrocytoma cell lines.85

Three different strategies are available to hinder TAM receptor signaling: ligand sinks, antibody-mediated downregulation/blocking, and small-molecule kinase inhibitors. Our lab has shown that treatment with a Mer-Fc fusion construct—one example of a ligand sink—inhibits Gas6 signaling, apoptotic cell engulfment by macrophages, and platelet aggregation.86 Such a construct could be used as adjuvant therapy to transiently inhibit TAM signaling in leukemia cells at the time of chemotherapy administration.

Two separate groups have reported that Axl-specific antibodies inhibit signaling and enhance the effect of standard chemotherapies on tumor cells.84,87,88 Zhang et al. showed that treatment with a polyclonal anti-Axl antibody significantly diminishes the motility and invasive properties of human breast cancer cells.88 Furthermore, Ye et al. demonstrated that an anti-Axl monoclonal antibody blocks Gas6 signaling, reduces Axl surface expression, and synergizes with standard chemotherapy agents to inhibit tumor growth in xenograft models of lung and breast cancer.87 While there are no published studies regarding the use of Mer-specific antibodies as a therapeutic agent, our lab has characterized monoclonal anti-Mer antibodies that decrease surface expression of Mer in human ALL cell lines.

Although there are few reports of TAM receptor-specific small-molecule inhibitors, several inhibitors developed against closely related receptor tyrosine kinases,89 such as c-Kit and Met, are also active against Axl and/or Mer. Foretinib (GSK1363089, GlaxoS-mithKline), a Met inhibitor currently in several clinical trials for use against solid tumors, inhibits in vitro Axl phosphorylation in the low nanomolar range (IC50=11 nM).33 MP470 (Amuvatinib, SuperGen), originally designed as a c-Kit inhibitor, inhibits Axl activation90,91 and is currently in a Phase II clinical trial to test it as combination therapy with Platinum-Etoposide against small cell lung carcinoma. Crizotinib (PF-2341066, Pfizer), a Met inhibitor also active against ALK kinase, inhibits Axl with an IC50=322 nM,92 and is currently being evaluated in a variety of clinical trials. Lastly, BMS777607 (Bristol-Meyers Squibb)—also developed against Met—has low nanomolar activity in vitro against Axl, Mer and Tyro-3 (IC50= 1.1, 14, and 4.3 nM, respectively) and has been evaluated in a Phase I dose-escalation clinical trial.93

Unlike the inhibitors mentioned above, R428 (Rigel) was developed specifically against Axl.94 Treatment with R428, which inhibits Axl with an IC50 = 14 nM but shows low activity for Mer and Tyro3, blocked both in vitro invasion and in vivo metastatic potential of a human breast cancer cell line, and also prolonged survival in a mouse model of post-mastectomy metastasis.94 In a separate study, R428 was also shown to induce apoptosis in CLL B cells.95

In contrast with Axl, there is no information on specific small molecule inhibitors targeting Mer or Tyro-3. In a crystal structural study of Mer, a library of kinase inhibitors was screened in vitro against the kinase domain of the receptor96: several compounds were identified, but no further reports of biological activity or toxicity are available.

VI. CONCLUSION

In this review, we have described the role of the TAM receptor family in leukemia to spotlight the aberrant expression patterns within different subsets and illustrate how receptor activation upregulates particular proliferative and antiapoptotic signaling pathways known to contribute to leukemogenesis. Although the mechanisms underlying abnormal TAM receptor expression are still unclear, the existing data—insight gained from studies conducted on various cell types—underscore how multiple, and perhaps context-dependent, processes facilitate overexpression of TAM receptors in leukemic cells.

The preclinical data presented here strongly support the development of specific inhibitors of TAM receptors: Mer and/or Axl inhibition can enhance leukemia cell sensitivity to chemotherapeutic agents currently used in the clinic, thus requiring lower doses to achieve equal or better efficiency and decreasing the severity of side effects observed with current therapies. Given that patients with inactivating Mer mutations and TAM receptor-knockout mice experience late-onset health defects,97101 transient TAM inhibition during chemotherapy will not likely bear the same long-term consequences. Furthermore, current immunophenotyping assays can easily be adapted to characterize TAM expression in leukemia patient samples,25,28 highlighting the diagnostic value of TAM family receptors. For all of these reasons, we believe that incorporating TAM receptor-targeted therapies into treatment regimens for leukemia, as well as for other cancers, is an exciting strategy in the ongoing fight against this devastating disease.

Acknowledgments

This article was supported in part by grants from the National Institutes of Health (RO1CA137078, DKG); the National Heart, Lung, and Blood Institute (F32HL096416, LB); and from the American Cancer Society (RSG-08-291-01-LIB, DKG).

ABBREVIATIONS

ALL

acute lymphoblastic leukemia

AML

acute myeloid leukemia

CLL

chronic lymphocytic leukemia

CML

chronic myelogenous leukemia

CSF1

colony stimulating factor 1

HDAC

histone deacetylase

IFN

type I interferon

LXR

liver X receptor

NK

natural killer

NKT

natural killer T cell

RTK

receptor tyrosine kinase

TAM

Tyro3, Axl, and Mer

REFERENCES

  • 1.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009 Jul-Aug;59(4):225–249. doi: 10.3322/caac.20006. [DOI] [PubMed] [Google Scholar]
  • 2.Kaatsch P. Epidemiology of childhood cancer. Cancer Treat Rev. 2010 Jun;36(4):277–285. doi: 10.1016/j.ctrv.2010.02.003. [DOI] [PubMed] [Google Scholar]
  • 3.Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Waldron W, Altekruse SF, Kosary CL, Ruhl J, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Chen HS, Feuer EJ, Cronin KA, Edwards BK. SEER Cancer Statistics Review. 2011:1975–2008. [Google Scholar]
  • 4.Landier W, Bhatia S. Cancer survivorship: a pediatric perspective. Oncologist. 2008 Nov;13(11):1181–1192. doi: 10.1634/theoncologist.2008-0104. [DOI] [PubMed] [Google Scholar]
  • 5.Oeffinger KC, Mertens AC, Sklar CA, Kawashima T, Hudson MM, Meadows AT, Friedman DL, Marina N, Hobbie W, Kadan-Lottick NS, Schwartz CL, Leisenring W, Robison LL. Chronic health conditions in adult survivors of childhood cancer. N Engl J Med. 2006 Oct 12;355(15):1572–1582. doi: 10.1056/NEJMsa060185. [DOI] [PubMed] [Google Scholar]
  • 6.Nguyen K, Devidas M, Cheng SC, La M, Raetz EA, Carroll WL, Winick NJ, Hunger SP, Gaynon PS, Loh ML. Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children’s Oncology Group study. Leukemia. 2008 Dec;22(12):2142–2150. doi: 10.1038/leu.2008.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lemke G, Rothlin CV. Immunobiology of the TAM receptors. Nat Rev Immunol. 2008 May;8(5):327–336. doi: 10.1038/nri2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Angelillo-Scherrer A, Burnier L, Flores N, Savi P, DeMol M, Schaeffer P, Herbert JM, Lemke G, Goff SP, Matsushima GK, Earp HS, Vesin C, Hoylaerts MF, Plaisance S, Collen D, Conway EM, Wehrle-Haller B, Carmeliet P. Role of Gas6 receptors in platelet signaling during thrombus stabilization and implications for antithrombotic therapy. J Clin Invest. 2005 Feb;115(2):237–246. doi: 10.1172/JCI22079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gould WR, Baxi SM, Schroeder R, Peng YW, Leadley RJ, Peterson JT, Perrin LA. Gas6 receptors Axl, Sky and Mer enhance platelet activation and regulate thrombotic responses. J Thromb Haemost. 2005 Apr;3(4):733–741. doi: 10.1111/j.1538-7836.2005.01186.x. [DOI] [PubMed] [Google Scholar]
  • 10.Angelillo-Scherrer A, Burnier L, Lambrechts D, Fish RJ, Tjwa M, Plaisance S, Sugamele R, DeMol M, Martinez-Soria E, Maxwell PH, Lemke G, Goff SP, Matsushima GK, Earp HS, Chanson M, Collen D, Izui S, Schapira M, Conway EM, Carmeliet P. Role of Gas6 in erythropoiesis and anemia in mice. J Clin Invest. 2008 Feb;118(2):583–596. doi: 10.1172/JCI30375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Linger RM, Keating AK, Earp HS, Graham DK. TAM receptor tyrosine kinases: biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv Cancer Res. 2008;100:35–83. doi: 10.1016/S0065-230X(08)00002-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Caraux A, Lu Q, Fernandez N, Riou S, Di Santo JP, Raulet DH, Lemke G, Roth C. Natural killer cell differentiation driven by Tyro3 receptor tyrosine kinases. Nat Immunol. 2006 Jul;7(7):747–754. doi: 10.1038/ni1353. [DOI] [PubMed] [Google Scholar]
  • 13.Behrens EM, Gadue P, Gong SY, Garrett S, Stein PL, Cohen PL. The mer receptor tyrosine kinase: expression and function suggest a role in innate immunity. Eur J Immunol. 2003 Aug;33(8):2160–2167. doi: 10.1002/eji.200324076. [DOI] [PubMed] [Google Scholar]
  • 14.Graham DK, Dawson TL, Mullaney DL, Snodgrass HR, Earp HS. Cloning and mRNA expression analysis of a novel human protooncogene, c-mer. Cell Growth Differ. 1994 Jun;5(6):647–657. [PubMed] [Google Scholar]
  • 15.Crosier PS, Hall LR, Vitas MR, Lewis PM, Crosier KE. Identification of a novel receptor tyrosine kinase expressed in acute myeloid leukemic blasts. Leuk Lymphoma. 1995 Aug;18(5–6):443–449. doi: 10.3109/10428199509059643. [DOI] [PubMed] [Google Scholar]
  • 16.De Vos J, Couderc G, Tarte K, Jourdan M, Requirand G, Delteil MC, Rossi JF, Mechti N, Klein B. Identifying intercellular signaling genes expressed in malignant plasma cells by using complementary DNA arrays. Blood. 2001 Aug 1;98(3):771–780. doi: 10.1182/blood.v98.3.771. [DOI] [PubMed] [Google Scholar]
  • 17.Liu E, Hjelle B, Bishop JM. Transforming genes in chronic myelogenous leukemia. Proc Natl Acad Sci U S A. 1988 Mar;85(6):1952–1956. doi: 10.1073/pnas.85.6.1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.O’Bryan JP, Frye RA, Cogswell PC, Neubauer A, Kitch B, Prokop C, Espinosa R, 3rd, Le Beau MM, Earp HS, Liu ET. axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase. Mol Cell Biol. 1991 Oct;11(10):5016–5031. doi: 10.1128/mcb.11.10.5016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Janssen JW, Schulz AS, Steenvoorden AC, Schmidberger M, Strehl S, Ambros PF, Bartram CR. A novel putative tyrosine kinase receptor with oncogenic potential. Oncogene. 1991 Nov;6(11):2113–2120. [PubMed] [Google Scholar]
  • 20.Challier C, Uphoff CC, Janssen JW, Drexler HG. Differential expression of the ufo/axl oncogene in human leukemia-lymphoma cell lines. Leukemia. 1996 May;10(5):781–787. [PubMed] [Google Scholar]
  • 21.Neubauer A, Fiebeler A, Graham DK, O’Bryan JP, Schmidt CA, Barckow P, Serke S, Siegert W, Snodgrass HR, Huhn D. Expression of axl, a transforming receptor tyrosine kinase, in normal and malignant hematopoiesis. Blood. 1994 Sep 15;84(6):1931–1941. [PubMed] [Google Scholar]
  • 22.Neubauer A, Burchert A, Maiwald C, Gruss HJ, Serke S, Huhn D, Wittig B, Liu E. Recent progress on the role of Axl, a receptor tyrosine kinase, in malignant transformation of myeloid leukemias. Leuk Lymphoma. 1997 Mar;25(1–2):91–96. doi: 10.3109/10428199709042499. [DOI] [PubMed] [Google Scholar]
  • 23.Rochlitz C, Lohri A, Bacchi M, Schmidt M, Nagel S, Fopp M, Fey MF, Herrmann R, Neubauer A. Axl expression is associated with adverse prognosis and with expression of Bcl-2 and CD34 in de novo acute myeloid leukemia (AML): results from a multicenter trial of the Swiss Group for Clinical Cancer Research (SAKK) Leukemia. 1999 Sep;13(9):1352–1358. doi: 10.1038/sj.leu.2401484. [DOI] [PubMed] [Google Scholar]
  • 24.Hong CC, Lay JD, Huang JS, Cheng AL, Tang JL, Lin MT, Lai GM, Chuang SE. Receptor tyrosine kinase AXL is induced by chemotherapy drugs and overexpression of AXL confers drug resistance in acute myeloid leukemia. Cancer Lett. 2008 Sep 18;268(2):314–324. doi: 10.1016/j.canlet.2008.04.017. [DOI] [PubMed] [Google Scholar]
  • 25.Graham DK, Salzberg DB, Kurtzberg J, Sather S, Matsushima GK, Keating AK, Liang X, Lovell MA, Williams SA, Dawson TL, Schell MJ, Anwar AA, Snodgrass HR, Earp HS. Ectopic expression of the proto-oncogene Mer in pediatric T-cell acute lymphoblastic leukemia. Clin Cancer Res. 2006 May 1;12(9):2662–2669. doi: 10.1158/1078-0432.CCR-05-2208. [DOI] [PubMed] [Google Scholar]
  • 26.Linger RM, DeRyckere D, Brandao L, Sawczyn KK, Jacobsen KM, Liang X, Keating AK, Graham DK. Mer receptor tyrosine kinase is a novel therapeutic target in pediatric B-cell acute lymphoblastic leukemia. Blood. 2009 Sep 24;114(13):2678–2687. doi: 10.1182/blood-2009-03-209247. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 27.Yeoh EJ, Ross ME, Shurtleff SA, Williams WK, Patel D, Mahfouz R, Behm FG, Raimondi SC, Relling MV, Patel A, Cheng C, Campana D, Wilkins D, Zhou X, Li J, Liu H, Pui CH, Evans WE, Naeve C, Wong L, Downing JR. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell. 2002 Mar;1(2):133–143. doi: 10.1016/s1535-6108(02)00032-6. [DOI] [PubMed] [Google Scholar]
  • 28.Eisenman K, Sather S, McGranahan A, Liang X, Desch A, Cannon C, Martinson H, Keating AK, Graham DK. Mer as a Potential Novel Target in Acute Myeloid Leukemia. Pediatr Blood Cancer. 2010 Jun;54(6):790. [Google Scholar]
  • 29.Jia R, Mayer BJ, Hanafusa T, Hanafusa H. A novel oncogene, v-ryk, encoding a truncated receptor tyrosine kinase is transduced into the RPL30 virus without loss of viral sequences. J Virol. 1992 Oct;66(10):5975–5987. doi: 10.1128/jvi.66.10.5975-5987.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Keating AK, Salzberg DB, Sather S, Liang X, Nickoloff S, Anwar A, Deryckere D, Hill K, Joung D, Sawczyn KK, Park J, Curran-Everett D, McGavran L, Meltesen L, Gore L, Johnson GL, Graham DK. Lymphoblastic leukemia/lymphoma in mice overex-pressing the Mer (MerTK) receptor tyrosine kinase. Oncogene. 2006 Oct 5;25(45):6092–6100. doi: 10.1038/sj.onc.1209633. [DOI] [PubMed] [Google Scholar]
  • 31.Margareto J, Leis O, Larrarte E, Pomposo IC, Garibi JM, Lafuente JV. DNA copy number variation and gene expression analyses reveal the implication of specific oncogenes and genes in GBM. Cancer Invest. 2009 Jun;27(5):541–548. doi: 10.1080/07357900802563044. [DOI] [PubMed] [Google Scholar]
  • 32.Kubo T, Kuroda Y, Shimizu H, Kokubu A, Okada N, Hosoda F, Arai Y, Nakamura Y, Taniguchi H, Yanagihara K, Imoto I, Inazawa J, Hirohashi S, Shibata T. Resequencing and copy number analysis of the human tyrosine kinase gene family in poorly differentiated gastric cancer. Carcinogenesis. 2009 Nov;30(11):1857–1864. doi: 10.1093/carcin/bgp206. [DOI] [PubMed] [Google Scholar]
  • 33.Liu L, Greger J, Shi H, Liu Y, Greshock J, Annan R, Halsey W, Sathe GM, Martin AM, Gilmer TM. Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: activation of AXL. Cancer Res. 2009 Sep 1;69(17):6871–6878. doi: 10.1158/0008-5472.CAN-08-4490. [DOI] [PubMed] [Google Scholar]
  • 34.Abba MC, Fabris VT, Hu Y, Kittrell FS, Cai WW, Donehower LA, Sahin A, Medina D, Aldaz CM. Identification of novel amplification gene targets in mouse and human breast cancer at a syntenic cluster mapping to mouse ch8A1 and human ch13q34. Cancer Res. 2007 May 1;67(9):4104–4112. doi: 10.1158/0008-5472.CAN-06-4672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Liao Y, Yu D, Hung MC. Novel approaches for chemosensitization of breast cancer cells: the E1A story. Adv Exp Med Biol. 2007;608:144–169. doi: 10.1007/978-0-387-74039-3_11. [DOI] [PubMed] [Google Scholar]
  • 36.Mudduluru G, Allgayer H. The human receptor tyrosine kinase Axl gene—promoter characterization and regulation of constitutive expression by Sp1, Sp3 and CpG methylation. Biosci Rep. 2008 Jun;28(3):161–176. doi: 10.1042/BSR20080046. [DOI] [PubMed] [Google Scholar]
  • 37.Orso F, Jager R, Calogero RA, Schorle H, Sismondi P, De Bortoli M, Taverna D. AP-2alpha regulates migration of GN-11 neurons via a specific genetic programme involving the Axl receptor tyrosine kinase. BMC Biol. 2009;7:25. doi: 10.1186/1741-7007-7-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Avilla E, Guarino V, Visciano C, Liotti F, Svelto M, Krishnamoorthy G, Franco R, Melillo RM. Activation of TYRO3/AXL tyrosine kinase receptors in thyroid cancer. Cancer Res. 2011 Mar 1;71(5):1792–1804. doi: 10.1158/0008-5472.CAN-10-2186. [DOI] [PubMed] [Google Scholar]
  • 39.Richer JK, Jacobsen BM, Manning NG, Abel MG, Wolf DM, Horwitz KB. Differential gene regulation by the two progesterone receptor isoforms in human breast cancer cells. J Biol Chem. 2002 Feb 15;277(7):5209–5218. doi: 10.1074/jbc.M110090200. [DOI] [PubMed] [Google Scholar]
  • 40.Wong CC, Lee WM. The proximal cis-acting elements Sp1, Sp3 and E2F regulate mouse mer gene transcription in Sertoli cells. Eur J Biochem. 2002 Aug;269(15):3789–3800. doi: 10.1046/j.1432-1033.2002.03092.x. [DOI] [PubMed] [Google Scholar]
  • 41.A-Gonzalez N, Bensinger SJ, Hong C, Beceiro S, Bradley MN, Zelcer N, Deniz J, Ramirez C, Diaz M, Gallardo G, de Galarreta CR, Salazar J, Lopez F, Edwards P, Parks J, Andujar M, Tontonoz P, Castrillo A. Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR. Immunity. 2009 Aug 21;31(2):245–258. doi: 10.1016/j.immuni.2009.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Geyeregger R, Shehata M, Zeyda M, Kiefer FW, Stuhlmeier KM, Porpaczy E, Zlabinger GJ, Jager U, Stulnig TM. Liver X receptors interfere with cytokine-induced proliferation and cell survival in normal and leukemic lymphocytes. J Leukoc Biol. 2009 Nov;86(5):1039–1048. doi: 10.1189/jlb.1008663. [DOI] [PubMed] [Google Scholar]
  • 43.Bensinger SJ, Bradley MN, Joseph SB, Zelcer N, Janssen EM, Hausner MA, Shih R, Parks JS, Edwards PA, Jamieson BD, Tontonoz P. LXR signaling couples sterol metabolism to proliferation in the acquired immune response. Cell. 2008 Jul 11;134(1):97–111. doi: 10.1016/j.cell.2008.04.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.So AY, Cooper SB, Feldman BJ, Manuchehri M, Yamamoto KR. Conservation analysis predicts in vivo occupancy of glucocorticoid receptor-binding sequences at glucocorticoid-induced genes. Proc Natl Acad Sci U S A. 2008 Apr 15;105(15):5745–5749. doi: 10.1073/pnas.0801551105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.McColl A, Bournazos S, Franz S, Perretti M, Morgan BP, Haslett C, Dransfield I. Glucocorticoids induce protein S-dependent phagocytosis of apoptotic neutrophils by human macrophages. J Immunol. 2009 Aug 1;183(3):2167–2175. doi: 10.4049/jimmunol.0803503. [DOI] [PubMed] [Google Scholar]
  • 46.Chiaretti S, Li X, Gentleman R, Vitale A, Wang KS, Mandelli F, Foa R, Ritz J. Gene expression profiles of B-lineage adult acute lymphocytic leukemia reveal genetic patterns that identify lineage derivation and distinct mechanisms of transformation. Clin Cancer Res. 2005 Oct 15;11(20):7209–7219. doi: 10.1158/1078-0432.CCR-04-2165. [DOI] [PubMed] [Google Scholar]
  • 47.Fujita PA, Rhead B, Zweig AS, Hinrichs AS, Karolchik D, Cline MS, Goldman M, Barber GP, Clawson H, Coelho A, Diekhans M, Dreszer TR, Giardine BM, Harte RA, Hillman-Jackson J, Hsu F, Kirkup V, Kuhn RM, Learned K, Li CH, Meyer LR, Pohl A, Raney BJ, Rosenbloom KR, Smith KE, Haussler D, Kent WJ. The UCSC Genome Browser database: update 2011. Nucleic Acids Res. 2011 Jan;39:D876–D882. doi: 10.1093/nar/gkq963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Smith KS, Jacobs Y, Chang CP, Cleary ML. Chimeric oncoprotein E2A-Pbx1 induces apoptosis of hematopoietic cells by a p53-independent mechanism that is suppressed by Bcl-2. Oncogene. 1997 Jun 19;14(24):2917–2926. doi: 10.1038/sj.onc.1201249. [DOI] [PubMed] [Google Scholar]
  • 49.Liu R, Gong M, Li X, Zhou Y, Gao W, Tulpule A, Chaudhary PM, Jung J, Gill PS. Induction, regulation, and biologic function of Axl receptor tyrosine kinase in Kaposi sarcoma. Blood. 2010 Jul 15;116(2):297–305. doi: 10.1182/blood-2009-12-257154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Hideshima T, Akiyama M, Chauhan D, Munshi N, Gu X, Bailey C, Joseph M, Libermann TA, Richon VM, Marks PA, Anderson KC. Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications. Proc Natl Acad Sci U S A. 2004 Jan 13;101(2):540–545. doi: 10.1073/pnas.2536759100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massague J. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008 Jan 10;451(7175):147–152. doi: 10.1038/nature06487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kurowska-Stolarska M, Ballantine L, Stolarski B, Hunter J, Hueber A, Gracie JA, Liew FY, McInnes IB. miR-155 and miR-34a regulate proinflammatory cytokine production by human monocytes. Annals of the Rheumatic Diseases. 2010 Mar 1;69(Suppl 2):A30. [Google Scholar]
  • 53.Bryant A, Lutherborrow M, Ma D. The clini-copathological relevance of microRNA in normal and malignant haematopoiesis. Pathology. 2009;41(3):204–213. doi: 10.1080/00313020902756287. [DOI] [PubMed] [Google Scholar]
  • 54.Mudduluru G, Ceppi P, Kumarswamy R, Scagliotti GV, Papotti M, Allgayer H. Regulation of Axl receptor tyrosine kinase expression by miR-34a and miR-199a/b in solid cancer. Oncogene. 2011 Jun 23;30(25):2888–2899. doi: 10.1038/onc.2011.13. [DOI] [PubMed] [Google Scholar]
  • 55.Evdokimova V, Ruzanov P, Anglesio MS, Sorokin AV, Ovchinnikov LP, Buckley J, Triche TJ, Sonenberg N, Sorensen PH. Akt-mediated YB-1 phosphorylation activates translation of silent mRNA species. Mol Cell Biol. 2006 Jan;26(1):277–292. doi: 10.1128/MCB.26.1.277-292.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Stitt TN, Conn G, Gore M, Lai C, Bruno J, Radziejewski C, Mattsson K, Fisher J, Gies DR, Jones PF, Masiakowski P, Ryan TE, Tobkes NJ, Chen DH, DiStefano PS, Long GL, Basilico C, Goldfarb MP, Lemke G, Glass DJ, Yancopoulos GD. The anticoagulation factor protein S and its relative, Gas6, are ligands for the Tyro 3/Axl family of receptor tyrosine kinases. Cell. 1995 Feb 24;80(4):661–670. doi: 10.1016/0092-8674(95)90520-0. [DOI] [PubMed] [Google Scholar]
  • 57.Varnum BC, Young C, Elliott G, Garcia A, Bartley TD, Fridell YW, Hunt RW, Trail G, Clogston C, Toso RJ, Yanagihara D, Bennett L, Sylber M, Merewether LA, Tseng A, Escobar E, Edison TL, Yamane HK. Axl receptor tyrosine kinase stimulated by the vitamin K-dependent protein encoded by growth-arrest-specific gene 6. Nature. 1995 Feb 16;373(6515):623–626. doi: 10.1038/373623a0. [DOI] [PubMed] [Google Scholar]
  • 58.Balogh I, Hafizi S, Stenhoff J, Hansson K, Dahlback B. Analysis of Gas6 in human platelets and plasma. Arterioscler Thromb Vasc Biol. 2005 Jun;25(6):1280–1286. doi: 10.1161/01.ATV.0000163845.07146.48. [DOI] [PubMed] [Google Scholar]
  • 59.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 Aug;13(8):4976–4985. doi: 10.1128/mcb.13.8.4976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dormady SP, Zhang XM, Basch RS. Hematopoietic progenitor cells grow on 3T3 fibroblast monolayers that overexpress growth arrest-specific gene-6 (GAS6) Proc Natl Acad Sci U S A. 2000 Oct 24;97(22):12260–12265. doi: 10.1073/pnas.97.22.12260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shiozawa Y, Pedersen EA, Taichman RS. 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 Feb;38(2):132–140. doi: 10.1016/j.exphem.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Linger RM, Keating AK, Earp HS, Graham DK. Taking aim at Mer and Axl receptor tyrosine kinases as novel therapeutic targets in solid tumors. Expert Opin Ther Targets. 2010 Oct;14(10):1073–1090. doi: 10.1517/14728222.2010.515980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Caberoy NB, Zhou Y, Li W. Tubby and tubby-like protein 1 are new MerTK ligands for phagocytosis. EMBO J. 2010 Dec 1;29(23):3898–3910. doi: 10.1038/emboj.2010.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Fridell YW, Jin Y, Quilliam LA, Burchert A, McCloskey P, Spizz G, Varnum B, Der C, Liu ET. Differential activation of the Ras/extracellular-signal-regulated protein kinase pathway is responsible for the biological consequences induced by the Axl receptor tyrosine kinase. Mol Cell Biol. 1996 Jan;16(1):135–145. doi: 10.1128/mcb.16.1.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Georgescu MM, Kirsch KH, Shishido T, Zong C, Hanafusa H. Biological effects of c-Mer receptor tyrosine kinase in hematopoietic cells depend on the Grb2 binding site in the receptor and activation of NF-kappaB. Mol Cell Biol. 1999 Feb;19(2):1171–1181. doi: 10.1128/mcb.19.2.1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ling L, Kung HJ. Mitogenic signals and transforming potential of Nyk, a newly identified neural cell adhesion molecule-related receptor tyrosine kinase. Mol Cell Biol. 1995 Dec;15(12):6582–6592. doi: 10.1128/mcb.15.12.6582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chen J, Carey K, Godowski PJ. Identification of Gas6 as a ligand for Mer, a neural cell adhesion molecule related receptor tyrosine kinase implicated in cellular transformation. Oncogene. 1997 May 1;14(17):2033–2039. doi: 10.1038/sj.onc.1201039. [DOI] [PubMed] [Google Scholar]
  • 68.Braunger J, Schleithoff L, Schulz AS, Kessler H, Lammers R, Ullrich A, Bartram CR, Janssen JW. Intracellular signaling of the Ufo/Axl receptor tyrosine kinase is mediated mainly by a multi-substrate docking-site. Oncogene. 1997 Jun 5;14(22):2619–2631. doi: 10.1038/sj.onc.1201123. [DOI] [PubMed] [Google Scholar]
  • 69.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 Aug;17(8):4442–4453. doi: 10.1128/mcb.17.8.4442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sainaghi PP, Castello L, Bergamasco L, Galletti M, Bellosta P, Avanzi GC. Gas6 induces proliferation in prostate carcinoma cell lines expressing the Axl receptor. J Cell Physiol. 2005 Jul;204(1):36–44. doi: 10.1002/jcp.20265. [DOI] [PubMed] [Google Scholar]
  • 71.Guttridge KL, Luft JC, Dawson TL, Kozlowska E, Mahajan NP, Varnum B, Earp HS. Mer receptor tyrosine kinase signaling: prevention of apoptosis and alteration of cytoskeletal architecture without stimulation or proliferation. J Biol Chem. 2002 Jul 5;277(27):24057–24066. doi: 10.1074/jbc.M112086200. [DOI] [PubMed] [Google Scholar]
  • 72.Lopez-Guerra M, Colomer D. NF-kappaB as a therapeutic target in chronic lymphocytic leukemia. Expert Opin Ther Targets. 2010 Mar;14(3):275–288. doi: 10.1517/14728221003598930. [DOI] [PubMed] [Google Scholar]
  • 73.Demarchi F, Verardo R, Varnum B, Brancolini C, Schneider C. Gas6 anti-apoptotic signaling requires NF-kappa B activation. J Biol Chem. 2001 Aug 24;276(34):31738–31744. doi: 10.1074/jbc.M104457200. [DOI] [PubMed] [Google Scholar]
  • 74.Tibrewal N, Wu Y, D’Mello V, Akakura R, George TC, Varnum B, Birge RB. Autophosphorylation docking site Tyr-867 in Mer receptor tyrosine kinase allows for dissociation of multiple signaling pathways for phagocytosis of apoptotic cells and down-modulation of lipopolysaccharide-inducible NF-kappaB transcriptional activation. J Biol Chem. 2008 Feb 8;283(6):3618–3627. doi: 10.1074/jbc.M706906200. [DOI] [PubMed] [Google Scholar]
  • 75.Benekli M, Baer MR, Baumann H, Wetzler M. Signal transducer and activator of transcription proteins in leukemias. Blood. 2003 Apr 15;101(8):2940–2954. doi: 10.1182/blood-2002-04-1204. [DOI] [PubMed] [Google Scholar]
  • 76.Qazi S, Uckun FM. Gene expression profiles of infant acute lymphoblastic leukaemia and its prognostically distinct subsets. Br J Haematol. 2010 Jun;149(6):865–873. doi: 10.1111/j.1365-2141.2010.08177.x. [DOI] [PubMed] [Google Scholar]
  • 77.Lacronique V, Boureux A, Valle VD, Poirel H, Quang CT, Mauchauffe M, Berthou C, Lessard M, Berger R, Ghysdael J, Bernard OA. A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia. Science. 1997 Nov 14;278(5341):1309–1312. doi: 10.1126/science.278.5341.1309. [DOI] [PubMed] [Google Scholar]
  • 78.Zong C, Yan R, August A, Darnell JE, Jr, Hanafusa H. Unique signal transduction of Eyk: constitutive stimulation of the JAK-STAT pathway by an oncogenic receptor-type tyrosine kinase. EMBO J. 1996 Sep 2;15(17):4515–4525. [PMC free article] [PubMed] [Google Scholar]
  • 79.Rothlin CV, Ghosh S, Zuniga EI, Oldstone MB, Lemke G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell. 2007 Dec 14;131(6):1124–1136. doi: 10.1016/j.cell.2007.10.034. [DOI] [PubMed] [Google Scholar]
  • 80.O’Hare T, Deininger MW. Toward a cure for chronic myeloid leukemia. Clin Cancer Res. 2008 Dec 15;14(24):7971–7974. doi: 10.1158/1078-0432.CCR-08-1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.O’Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, Cornelissen JJ, Fischer T, Hochhaus A, Hughes T, Lechner K, Nielsen JL, Rousselot P, Reiffers J, Saglio G, Shepherd J, Simonsson B, Gratwohl A, Goldman JM, Kantarjian H, Taylor K, Verhoef G, Bolton AE, Capdeville R, Druker BJ. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2003 Mar 13;348(11):994–1004. doi: 10.1056/NEJMoa022457. [DOI] [PubMed] [Google Scholar]
  • 82.Small D. Targeting FLT3 for the treatment of leukemia. Semin Hematol. 2008 Jul;45(3 Suppl 2):S17–S21. doi: 10.1053/j.seminhematol.2008.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Griffin TC, Weitzman S, Weinstein H, Chang M, Cairo M, Hutchison R, Shiramizu B, Wiley J, Woods D, Barnich M, Gross TG. A study of rituximab and ifosfamide, carboplatin, and etoposide chemotherapy in children with recurrent/refractory B-cell (CD20+) non-Hodgkin lymphoma and mature B-cell acute lymphoblastic leukemia: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2009 Feb;52(2):177–181. doi: 10.1002/pbc.21753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li Y, Ye X, Tan C, Hongo JA, Zha J, Liu J, Kallop D, Ludlam MJ, Pei L. Axl as a potential therapeutic target in cancer: role of Axl in tumor growth, metastasis and angiogenesis. Oncogene. 2009 Oct 1;28(39):3442–3455. doi: 10.1038/onc.2009.212. [DOI] [PubMed] [Google Scholar]
  • 85.Keating AK, Kim GK, Jones AE, Donson AM, Ware K, Mulcahy JM, Salzberg DB, Foreman NK, Liang X, Thorburn A, Graham DK. Inhibition of Mer and Axl receptor tyrosine kinases in astrocytoma cells leads to increased apoptosis and improved chemosensitivity. Mol Cancer Ther. 2010 May;9(5):1298–1307. doi: 10.1158/1535-7163.MCT-09-0707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sather S, Kenyon KD, Lefkowitz JB, Liang X, Varnum BC, Henson PM, Graham DK. A soluble form of the Mer receptor tyrosine kinase inhibits macrophage clearance of apoptotic cells and platelet aggregation. Blood. 2007 Feb 1;109(3):1026–1033. doi: 10.1182/blood-2006-05-021634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.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 anti-cancer therapies. Oncogene. 2010 Sep 23;29(38):5254–5264. doi: 10.1038/onc.2010.268. [DOI] [PubMed] [Google Scholar]
  • 88.Zhang YX, Knyazev PG, Cheburkin YV, Sharma K, Knyazev YP, Orfi L, Szabadkai I, Daub H, Keri G, Ullrich A. AXL is a potential target for therapeutic intervention in breast cancer progression. Cancer Res. 2008 Mar 15;68(6):1905–1915. doi: 10.1158/0008-5472.CAN-07-2661. [DOI] [PubMed] [Google Scholar]
  • 89.Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002 Dec 6;298(5600):1912–1934. doi: 10.1126/science.1075762. [DOI] [PubMed] [Google Scholar]
  • 90.Mahadevan D, Cooke L, Riley C, Swart R, Simons B, Della Croce K, Wisner L, Iorio M, Shakalya K, Garewal H, Nagle R, Bearss D. A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors. Oncogene. 2007 Jun 7;26(27):3909–3919. doi: 10.1038/sj.onc.1210173. [DOI] [PubMed] [Google Scholar]
  • 91.Qi W, Cooke LS, Stejskal A, Riley C, Croce KD, Saldanha JW, Bearss D, Mahadevan D. MP470, a novel receptor tyrosine kinase inhibitor, in combination with Erlotinib inhibits the HER family/PI3K/ Akt pathway and tumor growth in prostate cancer. BMC Cancer. 2009;9:142. doi: 10.1186/1471-2407-9-142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung PP, Nambu MD, Los G, Bender SL, Mroczkowski B, Christensen JG. An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007 May 1;67(9):4408–4417. doi: 10.1158/0008-5472.CAN-06-4443. [DOI] [PubMed] [Google Scholar]
  • 93.Schroeder GM, An Y, Cai ZW, Chen XT, Clark C, Cornelius LA, Dai J, Gullo-Brown J, Gupta A, Henley B, Hunt JT, Jeyaseelan R, Kamath A, Kim K, Lippy J, Lombardo LJ, Manne V, Oppenheimer S, Sack JS, Schmidt RJ, Shen G, Stefanski K, Tokarski JS, Trainor GL, Wautlet BS, Wei D, Williams DK, Zhang Y, Fargnoli J, Borzilleri RM. 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 Mar 12;52(5):1251–1254. doi: 10.1021/jm801586s. [DOI] [PubMed] [Google Scholar]
  • 94.Holland SJ, Pan A, Franci C, Hu Y, Chang B, Li W, Duan M, Torneros A, Yu J, Heckrodt TJ, Zhang J, Ding P, Apatira A, Chua J, Brandt R, Pine P, Goff D, Singh R, Payan DG, Hitoshi Y. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res. 2010 Feb 15;70(4):1544–1554. doi: 10.1158/0008-5472.CAN-09-2997. [DOI] [PubMed] [Google Scholar]
  • 95.Ghosh AK, Secreto C, Boysen J, Sassoon T, Shanafelt TD, Mukhopadhyay D, Kay NE. The novel receptor tyrosine kinase Axl is constitutively active in B-cell chronic lymphocytic leukemia and acts as a docking site of nonreceptor kinases: implications for therapy. Blood. 2011 Feb 10;117(6):1928–1937. doi: 10.1182/blood-2010-09-305649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Huang X, Finerty P, Jr, Walker JR, Butler-Cole C, Vedadi M, Schapira M, Parker SA, Turk BE, Thompson DA, Dhe-Paganon S. Structural insights into the inhibited states of the Mer receptor tyrosine kinase. J Struct Biol. 2009 Feb;165(2):88–96. doi: 10.1016/j.jsb.2008.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.McHenry CL, Liu Y, Feng W, Nair AR, Feathers KL, Ding X, Gal A, Vollrath D, Sieving PA, Thompson DA. MERTK arginine-844-cysteine in a patient with severe rod-cone dystrophy: loss of mutant protein function in transfected cells. Invest Ophthalmol Vis Sci. 2004 May;45(5):1456–1463. doi: 10.1167/iovs.03-0909. [DOI] [PubMed] [Google Scholar]
  • 98.Gal A, Li Y, Thompson DA, Weir J, Orth U, Jacobson SG, Apfelstedt-Sylla E, Vollrath D. Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nat Genet. 2000 Nov;26(3):270–271. doi: 10.1038/81555. [DOI] [PubMed] [Google Scholar]
  • 99.Tschernutter M, Jenkins SA, Waseem NH, Saihan Z, Holder GE, Bird AC, Bhattacharya SS, Ali RR, Webster AR. Clinical characterisation of a family with retinal dystrophy caused by mutation in the Mertk gene. Br J Ophthalmol. 2006 Jun;90(6):718–723. doi: 10.1136/bjo.2005.084897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lu Q, Gore M, Zhang Q, Camenisch T, Boast S, Casagranda F, Lai C, Skinner MK, Klein R, Matsushima GK, Earp HS, Goff SP, Lemke G. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature. 1999 Apr 22;398(6729):723–728. doi: 10.1038/19554. [DOI] [PubMed] [Google Scholar]
  • 101.Camenisch TD, Koller BH, Earp HS, Matsushima GK. A novel receptor tyrosine kinase, Mer, inhibits TNF-alpha production and lipopolysaccharide-induced endotoxic shock. J Immunol. 1999 Mar 15;162(6):3498–3503. [PubMed] [Google Scholar]
  • 102.Dirks W, Rome D, Ringel F, Jager K, MacLeod RA, Drexler HG. Expression of the growth arrest-specific gene 6 (GAS6) in leukemia and lymphoma cell lines. Leuk Res. 1999 Jul;23(7):643–651. doi: 10.1016/s0145-2126(99)00075-2. [DOI] [PubMed] [Google Scholar]
  • 103.Graham DK, Bowman GW, Dawson TL, Stanford WL, Earp HS, Snodgrass HR. Cloning and developmental expression analysis of the murine c-mer tyrosine kinase. Oncogene. 1995 Jun 15;10(12):2349–2359. [PubMed] [Google Scholar]
  • 104.Angelillo-Scherrer A, de Frutos P, Aparicio C, Melis E, Savi P, Lupu F, Arnout J, Dewerchin M, Hoylaerts M, Herbert J, Collen D, Dahlback B, Carmeliet P. Deficiency or inhibition of Gas6 causes platelet dysfunction and protects mice against thrombosis. Nat Med. 2001 Feb;7(2):215–221. doi: 10.1038/84667. [DOI] [PubMed] [Google Scholar]
  • 105.Cosemans JM, Van Kruchten R, Olieslagers S, Schurgers LJ, Verheyen FK, Munnix IC, Waltenberger J, Angelillo-Scherrer A, Hoylaerts MF, Carmeliet P, Heemskerk JW. Potentiating role of Gas6 and Tyro3, Axl and Mer (TAM) receptors in human and murine platelet activation and thrombus stabilization. J Thromb Haemost. 2010 Aug;8(8):1797–1808. doi: 10.1111/j.1538-7836.2010.03935.x. [DOI] [PubMed] [Google Scholar]
  • 106.Avanzi GC, Gallicchio M, Cavalloni G, Gammaitoni L, Leone F, Rosina A, Boldorini R, Monga G, Pegoraro L, Varnum B, Aglietta M. GAS6, the ligand of Axl and Rse receptors, is expressed in hematopoietic tissue but lacks mitogenic activity. Exp Hematol. 1997 Nov;25(12):1219–1226. [PubMed] [Google Scholar]
  • 107.Crosier PS, Freeman SA, Orlic D, Bodine DM, Crosier KE. The Dtk receptor tyrosine kinase, which binds protein S, is expressed during hematopoiesis. Exp Hematol. 1996 Feb;24(2):318–323. [PubMed] [Google Scholar]
  • 108.Tang H, Chen S, Wang H, Wu H, Lu Q, Han D. TAM receptors and the regulation of erythropoiesis in mice. Haematologica. 2009 Mar;94(3):326–334. doi: 10.3324/haematol.13635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zheng Y, Zhang L, Lu Q, Wang X, Yu F. NGF-induced Tyro3 and Axl function as survival factors for differentiating PC12 cells. Biochem Biophys Res Commun. 2009 Jan 16;378(3):371–375. doi: 10.1016/j.bbrc.2008.11.049. [DOI] [PubMed] [Google Scholar]
  • 110.Mudduluru G, Vajkoczy P, Allgayer H. Myeloid zinc finger 1 induces migration, invasion, and in vivo metastasis through Axl gene expression in solid cancer. Mol Cancer Res. 2010 Feb;8(2):159–169. doi: 10.1158/1541-7786.MCR-09-0326. [DOI] [PubMed] [Google Scholar]
  • 111.Manalo DJ, Rowan A, Lavoie T, Natarajan L, Kelly BD, Ye SQ, Garcia JG, Semenza GL. Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood. 2005 Jan 15;105(2):659–669. doi: 10.1182/blood-2004-07-2958. [DOI] [PubMed] [Google Scholar]
  • 112.Gross C, Dubois-Pot H, Wasylyk B. The ternary complex factor Net/Elk-3 participates in the transcriptional response to hypoxia and regulates HIF-1 alpha. Oncogene. 2008 Feb 21;27(9):1333–1341. doi: 10.1038/sj.onc.1210736. [DOI] [PubMed] [Google Scholar]
  • 113.Petrova TV, Makinen T, Makela TP, Saarela J, Virtanen I, Ferrell RE, Finegold DN, Kerjaschki D, Yla-Herttuala S, Alitalo K. Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor. EMBO J. 2002 Sep 2;21(17):4593–4599. doi: 10.1093/emboj/cdf470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Li Y, Mahajan NP, Webster-Cyriaque J, Bhende P, Hong GK, Earp HS, Kenney S. The C-mer gene is induced by Epstein-Barr virus immediateearly protein BRLF1. J Virol. 2004 Nov;78(21):11778–11785. doi: 10.1128/JVI.78.21.11778-11785.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Khan J, Bittner ML, Saal LH, Teichmann U, Azorsa DO, Gooden GC, Pavan WJ, Trent JM, Meltzer PS. cDNA microarrays detect activation of a myogenic transcription program by the PAX3-FKHR fusion oncogene. Proc Natl Acad Sci U S A. 1999 Nov 9;96(23):13264–13269. doi: 10.1073/pnas.96.23.13264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mo R, Tony Zhu Y, Zhang Z, Rao SM, Zhu YJ. GAS6 is an estrogen-inducible gene in mammary epithelial cells. Biochem Biophys Res Commun. 2007 Feb 2;353(1):189–194. doi: 10.1016/j.bbrc.2006.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES