AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew
Abstract
:Simple Summary
Abstract
1. Introduction
2. Overview of the Structure and Regulation of AXL Receptor
2.1. Structural Features of TAM Receptors and Their Interactions with the Ligands
2.2. AXL Receptor Regulation by Proteolytic Cleavage
3. Overview of AXL-Regulated Signaling
3.1. Docking Sites for Signaling Proteins
3.2. AXL Dimerization Partners and Diversification of Downstream Signaling
4. AXL Functions in Normal Tissues. Lessons from Mice
4.1. Defective Efferocytosis in Immune System of TAM−/− Mice Results in Autoimmunity
4.2. Blindness and Male Infertility in TAM Knockout Mice
4.3. AXL Stimulates Efferocytosis by Regulating Cytoskeletal Dynamics
4.4. AXL Limits the Innate Immune Response
5. AXL and Cancer Cell Motility. Is Efferocytic Machinery Hijacked by Cancer Cells?
5.1. Motile Cancer Cells May Utilize DOCK180-ELMO Signaling Implicated in Efferocytosis
5.2. Are Exosomes in Tumour Microenvironment Involved in AXL-Induced Cell Migration?
6. AXL and Drug Resistance in Cancer
6.1. Survival in Toxic Conditions (Analogy with Professional Phagocytes)
6.2. Protection of Healthy Tissues from Autoimmune Damage and Immune Evasion in Cancer: Common Mechanisms
7. AXL, Epithelial-Mesenchymal Plasticity, Drug Tolerant Persister Cells, and Cancer Metastasis
7.1. Cancer Cells Exist in Distinct Differentiation States
7.2. AXL Belongs to the Mesenchymal Gene Expression Signatures Characterizing Aggressive Cancers
7.3. AXL Is Implicated in Metastasis in Experimental In Vivo Models
7.4. Hypothetical Role of AXL in the Formation of Drug Tolerant Persister Cancer Cells
8. Concluding Remarks
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Antony, J.; Huang, R.Y. AXL-Driven EMT State as a Targetable Conduit in Cancer. Cancer Res. 2017, 77, 3725–3732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antony, J.; Thiery, J.P.; Huang, R.Y. Epithelial-to-mesenchymal transition: Lessons from development, insights into cancer and the potential of EMT-subtype based therapeutic intervention. Phys. Biol. 2019, 16, 041004. [Google Scholar] [CrossRef] [PubMed]
- Colavito, S.A. AXL as a Target in Breast Cancer Therapy. J. Oncol. 2020, 2020, 5291952. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Caberoy, N.B.; Alvarado, G.; Bigcas, J.L.; Li, W. Galectin-3 is a new MerTK-specific eat-me signal. J. Cell Physiol. 2012, 227, 401–407. [Google Scholar] [CrossRef] [Green Version]
- Caberoy, N.B.; Zhou, Y.; Li, W. Tubby and tubby-like protein 1 are new MerTK ligands for phagocytosis. EMBO J. 2010, 29, 3898–3910. [Google Scholar] [CrossRef] [Green Version]
- Nomura, K.; Vilalta, A.; Allendorf, D.H.; Hornik, T.C.; Brown, G.C. Activated Microglia Desialylate and Phagocytose Cells via Neuraminidase, Galectin-3, and Mer Tyrosine Kinase. J. Immunol. 2017, 198, 4792–4801. [Google Scholar] [CrossRef] [Green Version]
- 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]
- 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] [Green Version]
- Lu, Y.; Wan, J.; Yang, Z.; Lei, X.; Niu, Q.; Jiang, L.; Passtoors, W.M.; Zang, A.; Fraering, P.C.; Wu, F. Regulated intramembrane proteolysis of the AXL receptor kinase generates an intracellular domain that localizes in the nucleus of cancer cells. FASEB J. 2017, 31, 1382–1397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. Mol. Biol. Cell 2017, 28, 3123–3131. [Google Scholar] [CrossRef]
- Brand, T.M.; Iida, M.; Corrigan, K.L.; Braverman, C.M.; Coan, J.P.; Flanigan, B.G.; Stein, A.P.; Salgia, R.; Rolff, J.; Kimple, R.J.; et al. The receptor tyrosine kinase AXL mediates nuclear translocation of the epidermal growth factor receptor. Sci. Signal. 2017, 10, eaag1064. [Google Scholar] [CrossRef]
- Ammoun, S.; Provenzano, L.; Zhou, L.; Barczyk, M.; Evans, K.; Hilton, D.A.; Hafizi, S.; Hanemann, C.O. Axl/Gas6/NFκB signalling in schwannoma pathological proliferation, adhesion and survival. Oncogene 2014, 33, 336–346. [Google Scholar] [CrossRef] [Green Version]
- Flem-Karlsen, K.; Nyakas, M.; Farstad, I.N.; McFadden, E.; Wernhoff, P.; Jacobsen, K.D.; Flørenes, V.A.; Mælandsmo, G.M. Soluble AXL as a marker of disease progression and survival in melanoma. PLoS ONE 2020, 15, e0227187. [Google Scholar] [CrossRef]
- Song, W.; Wang, H.; Lu, M.; Ni, X.; Bahri, N.; Zhu, S.; Chen, L.; Wu, Y.; Qiu, J.; Fletcher, J.A.; et al. AXL Inactivation Inhibits Mesothelioma Growth and Migration via Regulation of p53 Expression. Cancers 2020, 12, 2757. [Google Scholar] [CrossRef]
- Dantas-Barbosa, C.; Lesluyes, T.; Loarer, F.L.; Chibon, F.; Treilleux, I.; Coindre, J.M.; Meeus, P.; Brahmi, M.; Bally, O.; Ray-Coquard, I.; et al. Expression and role of TYRO3 and AXL as potential therapeutical targets in leiomyosarcoma. Br. J. Cancer 2017, 117, 1787–1797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Migdall-Wilson, J.; Bates, C.; Schlegel, J.; Brandão, L.; Linger, R.M.; DeRyckere, D.; Graham, D.K. Prolonged exposure to a Mer ligand in leukemia: Gas6 favors expression of a partial Mer glycoform and reveals a novel role for Mer in the nucleus. PLoS ONE 2012, 7, e31635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [PubMed] [Green Version]
- 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]
- 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] [PubMed] [Green Version]
- 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]
- Vouri, M.; An, Q.; Pilkington, G.; Hafizi, S. 107—Hetero-interaction amongst Tyro3 and Axl receptor tyrosine kinases diversifies cancer signalling. Eur. J. Cancer 2016, 69, S42. [Google Scholar] [CrossRef]
- Gusenbauer, S.; Vlaicu, P.; Ullrich, A. HGF induces novel EGFR functions involved in resistance formation to tyrosine kinase inhibitors. Oncogene 2013, 32, 3846–3856. [Google Scholar] [CrossRef] [Green Version]
- Meyer, A.S.; Miller, M.A.; Gertler, F.B.; Lauffenburger, D.A. The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells. Sci. Signal. 2013, 6, ra66. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Antony, J.; Tan, T.Z.; Kelly, Z.; Low, J.; Choolani, M.; Recchi, C.; Gabra, H.; Thiery, J.P.; Huang, R.Y. The GAS6-AXL signaling network is a mesenchymal (Mes) molecular subtype-specific therapeutic target for ovarian cancer. Sci. Signal. 2016, 9, ra97. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Ruan, G.X.; Kazlauskas, A. Axl is essential for VEGF-A-dependent activation of PI3K/Akt. EMBO J. 2012, 31, 1692–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- 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] [PubMed]
- Linger, R.M.; Keating, A.K.; Earp, H.S.; Graham, D.K. TAM receptor tyrosine kinases: Biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv. Cancer Res. 2008, 100, 35–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lemke, G.; Burstyn-Cohen, T. TAM receptors and the clearance of apoptotic cells. Ann. N. Y. Acad. Sci. 2010, 1209, 23–29. [Google Scholar] [CrossRef] [Green Version]
- Gallucci, S.; Matzinger, P. Danger signals: SOS to the immune system. Curr. Opin. Immunol. 2001, 13, 114–119. [Google Scholar] [CrossRef]
- Lemke, G. Biology of the TAM receptors. Cold Spring Harb. Perspect. Biol. 2013, 5, a009076. [Google Scholar] [CrossRef]
- Burstyn-Cohen, T.; Lew, E.D.; Través, P.G.; Burrola, P.G.; Hash, J.C.; Lemke, G. Genetic dissection of TAM receptor-ligand interaction in retinal pigment epithelial cell phagocytosis. Neuron 2012, 76, 1123–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Balasubramanian, K.; Schroit, A.J. Aminophospholipid asymmetry: A matter of life and death. Annu. Rev. Physiol. 2003, 65, 701–734. [Google Scholar] [CrossRef]
- Park, S.Y.; Kim, I.S. Engulfment signals and the phagocytic machinery for apoptotic cell clearance. Exp. Mol. Med. 2017, 49, e331. [Google Scholar] [CrossRef] [Green Version]
- Nakaya, M.; Tanaka, M.; Okabe, Y.; Hanayama, R.; Nagata, S. Opposite effects of rho family GTPases on engulfment of apoptotic cells by macrophages. J. Biol. Chem. 2006, 281, 8836–8842. [Google Scholar] [CrossRef] [Green Version]
- Côté, J.F.; Vuori, K. GEF what? Dock180 and related proteins help Rac to polarize cells in new ways. Trends Cell Biol. 2007, 17, 383–393. [Google Scholar] [CrossRef] [Green Version]
- Liau, N.P.D.; Laktyushin, A.; Lucet, I.S.; Murphy, J.M.; Yao, S.; Whitlock, E.; Callaghan, K.; Nicola, N.A.; Kershaw, N.J.; Babon, J.J. The molecular basis of JAK/STAT inhibition by SOCS1. Nat. Commun. 2018, 9, 1558. [Google Scholar] [CrossRef]
- Rothlin, C.V.; Lemke, G. TAM receptor signaling and autoimmune disease. Curr. Opin. Immunol. 2010, 22, 740–746. [Google Scholar] [CrossRef] [Green Version]
- Yamazaki, M.; Maruyama, S.; Abé, T.; Tsuneki, M.; Kato, H.; Izumi, K.; Tanuma, J.I.; Cheng, J.; Saku, T. Rac1-dependent phagocytosis of apoptotic cells by oral squamous cell carcinoma cells: A possible driving force for tumor progression. Exp. Cell Res. 2020, 392, 112013. [Google Scholar] [CrossRef]
- Nguyen, K.Q.; Tsou, W.I.; Calarese, D.A.; Kimani, S.G.; Singh, S.; Hsieh, S.; Liu, Y.; Lu, B.; Wu, Y.; Garforth, S.J.; et al. Overexpression of MERTK receptor tyrosine kinase in epithelial cancer cells drives efferocytosis in a gain-of-function capacity. J. Biol. Chem. 2014, 289, 25737–25749. [Google Scholar] [CrossRef] [Green Version]
- Koren, E.; Fuchs, Y. Modes of Regulated Cell Death in Cancer. Cancer Discov. 2021, 11, 245–265. [Google Scholar] [CrossRef] [PubMed]
- Abu-Thuraia, A.; Gauthier, R.; Chidiac, R.; Fukui, Y.; Screaton, R.A.; Gratton, J.P.; Côté, J.F. Axl phosphorylates Elmo scaffold proteins to promote Rac activation and cell invasion. Mol. Cell Biol. 2015, 35, 76–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, W.; Xiong, X.; Abdalla, A.; Alejo, S.; Zhu, L.; Lu, F.; Sun, H. HGF-induced formation of the MET-AXL-ELMO2-DOCK180 complex promotes RAC1 activation, receptor clustering, and cancer cell migration and invasion. J. Biol. Chem. 2018, 293, 15397–15418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Xu, J.; Jia, L.; Ma, H.; Li, Y.; Ma, Z.; Zhao, Y. Axl gene knockdown inhibits the metastasis properties of hepatocellular carcinoma via PI3K/Akt-PAK1 signal pathway. Tumour Biol. 2014, 35, 3809–3817. [Google Scholar] [CrossRef]
- Abu-Thuraia, A.; Goyette, M.A.; Boulais, J.; Delliaux, C.; Apcher, C.; Schott, C.; Chidiac, R.; Bagci, H.; Thibault, M.P.; Davidson, D.; et al. AXL confers cell migration and invasion by hijacking a PEAK1-regulated focal adhesion protein network. Nat. Commun. 2020, 11, 3586. [Google Scholar] [CrossRef]
- Gao, Y.; Qin, Y.; Wan, C.; Sun, Y.; Meng, J.; Huang, J.; Hu, Y.; Jin, H.; Yang, K. Small Extracellular Vesicles: A Novel Avenue for Cancer Management. Front. Oncol. 2021, 11, 638357. [Google Scholar] [CrossRef]
- Matsumura, S.; Minamisawa, T.; Suga, K.; Kishita, H.; Akagi, T.; Ichiki, T.; Ichikawa, Y.; Shiba, K. Subtypes of tumour cell-derived small extracellular vesicles having differently externalized phosphatidylserine. J. Extracell. Vesicles 2019, 8, 1579541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Graham, D.K.; DeRyckere, D.; Davies, K.D.; Earp, H.S. The TAM family: Phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer. Nat. Rev. Cancer 2014, 14, 769–785. [Google Scholar] [CrossRef] [PubMed]
- Park, M.; Kim, J.W.; Kim, K.M.; Kang, S.; Kim, W.; Kim, J.K.; Cho, Y.; Lee, H.; Baek, M.C.; Bae, J.H.; et al. Circulating Small Extracellular Vesicles Activate TYRO3 to Drive Cancer Metastasis and Chemoresistance. Cancer Res. 2021, 81, 3539–3553. [Google Scholar] [CrossRef] [PubMed]
- Ekman, C.; Stenhoff, J.; Dahlbäck, B. Gas6 is complexed to the soluble tyrosine kinase receptor Axl in human blood. J. Thromb. Haemost. 2010, 8, 838–844. [Google Scholar] [CrossRef]
- Kanzaki, R.; Naito, H.; Kise, K.; Takara, K.; Eino, D.; Minami, M.; Shintani, Y.; Funaki, S.; Kawamura, T.; Kimura, T.; et al. Gas6 derived from cancer-associated fibroblasts promotes migration of Axl-expressing lung cancer cells during chemotherapy. Sci. Rep. 2017, 7, 10613. [Google Scholar] [CrossRef] [Green Version]
- Wu, G.; Ma, Z.; Cheng, Y.; Hu, W.; Deng, C.; Jiang, S.; Li, T.; Chen, F.; Yang, Y. Targeting Gas6/TAM in cancer cells and tumor microenvironment. Mol. Cancer 2018, 17, 20. [Google Scholar] [CrossRef] [Green Version]
- Myers, S.H.; Brunton, V.G.; Unciti-Broceta, A. AXL Inhibitors in Cancer: A Medicinal Chemistry Perspective. J. Med. Chem. 2016, 59, 3593–3608. [Google Scholar] [CrossRef] [Green Version]
- Wium, M.; Ajayi-Smith, A.F.; Paccez, J.D.; Zerbini, L.F. The Role of the Receptor Tyrosine Kinase Axl in Carcinogenesis and Development of Therapeutic Resistance: An Overview of Molecular Mechanisms and Future Applications. Cancers 2021, 13, 1521. [Google Scholar] [CrossRef]
- 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]
- Wang, C.; Jin, H.; Wang, N.; Fan, S.; Wang, Y.; Zhang, Y.; Wei, L.; Tao, X.; Gu, D.; Zhao, F.; et al. Gas6/Axl Axis Contributes to Chemoresistance and Metastasis in Breast Cancer through Akt/GSK-3β/β-catenin Signaling. Theranostics 2016, 6, 1205–1219. [Google Scholar] [CrossRef] [PubMed]
- Guo, Z.; Li, Y.; Zhang, D.; Ma, J. Axl inhibition induces the antitumor immune response which can be further potentiated by PD-1 blockade in the mouse cancer models. Oncotarget 2017, 8, 89761–89774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Sen, T.; Tong, P.; Diao, L.; Li, L.; Fan, Y.; Hoff, J.; Heymach, J.V.; Wang, J.; Byers, L.A. Targeting AXL and mTOR Pathway Overcomes Primary and Acquired Resistance to WEE1 Inhibition in Small-Cell Lung Cancer. Clin. Cancer Res. 2017, 23, 6239–6253. [Google Scholar] [CrossRef] [Green Version]
- Du, W.; Huang, H.; Sorrelle, N.; Brekken, R.A. Sitravatinib potentiates immune checkpoint blockade in refractory cancer models. JCI Insight 2018, 3, e124184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Okura, N.; Nishioka, N.; Yamada, T.; Taniguchi, H.; Tanimura, K.; Katayama, Y.; Yoshimura, A.; Watanabe, S.; Kikuchi, T.; Shiotsu, S.; et al. ONO-7475, a Novel AXL Inhibitor, Suppresses the Adaptive Resistance to Initial EGFR-TKI Treatment in. Clin. Cancer Res. 2020, 26, 2244–2256. [Google Scholar] [CrossRef]
- Hong, J.; Maacha, S.; Belkhiri, A. Transcriptional upregulation of c-MYC by AXL confers epirubicin resistance in esophageal adenocarcinoma. Mol. Oncol. 2018, 12, 2191–2208. [Google Scholar] [CrossRef] [Green Version]
- Ludwig, K.F.; Du, W.; Sorrelle, N.B.; Wnuk-Lipinska, K.; Topalovski, M.; Toombs, J.E.; Cruz, V.H.; Yabuuchi, S.; Rajeshkumar, N.V.; Maitra, A.; et al. Small-Molecule Inhibition of Axl Targets Tumor Immune Suppression and Enhances Chemotherapy in Pancreatic Cancer. Cancer Res. 2018, 78, 246–255. [Google Scholar] [CrossRef] [Green Version]
- McDaniel, N.K.; Iida, M.; Nickel, K.P.; Longhurst, C.A.; Fischbach, S.R.; Rodems, T.S.; Kranjac, C.A.; Bo, A.Y.; Luo, Q.; Gallagher, M.M.; et al. AXL Mediates Cetuximab and Radiation Resistance Through Tyrosine 821 and the c-ABL Kinase Pathway in Head and Neck Cancer. Clin. Cancer Res. 2020, 26, 4349–4359. [Google Scholar] [CrossRef]
- Boshuizen, J.; Koopman, L.A.; Krijgsman, O.; Shahrabi, A.; van den Heuvel, E.G.; Ligtenberg, M.A.; Vredevoogd, D.W.; Kemper, K.; Kuilman, T.; Song, J.Y.; et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat. Med. 2018, 24, 203–212. [Google Scholar] [CrossRef] [PubMed]
- Flem-Karlsen, K.; McFadden, E.; Omar, N.; Haugen, M.H.; Øy, G.F.; Ryder, T.; Gullestad, H.P.; Hermann, R.; Mælandsmo, G.M.; Flørenes, V.A. Targeting AXL and the DNA Damage Response Pathway as a Novel Therapeutic Strategy in Melanoma. Mol. Cancer Ther. 2020, 19, 895–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boshuizen, J.; Pencheva, N.; Krijgsman, O.; Altimari, D.D.; Castro, P.G.; de Bruijn, B.; Ligtenberg, M.A.; Gresnigt-Van den Heuvel, E.; Vredevoogd, D.W.; Song, J.Y.; et al. Cooperative Targeting of Immunotherapy-Resistant Melanoma and Lung Cancer by an AXL-Targeting Antibody-Drug Conjugate and Immune Checkpoint Blockade. Cancer Res. 2021, 81, 1775–1787. [Google Scholar] [CrossRef] [PubMed]
- Meel, M.H.; de Gooijer, M.C.; Metselaar, D.S.; Sewing, A.C.P.; Zwaan, K.; Waranecki, P.; Breur, M.; Buil, L.C.M.; Lagerweij, T.; Wedekind, L.E.; et al. Combined Therapy of AXL and HDAC Inhibition Reverses Mesenchymal Transition in Diffuse Intrinsic Pontine Glioma. Clin. Cancer Res. 2020, 26, 3319–3332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aguilera, T.A.; Giaccia, A.J. Molecular Pathways: Oncologic Pathways and Their Role in T-cell Exclusion and Immune Evasion-A New Role for the AXL Receptor Tyrosine Kinase. Clin. Cancer Res. 2017, 23, 2928–2933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aguilera, T.A.; Rafat, M.; Castellini, L.; Shehade, H.; Kariolis, M.S.; Hui, A.B.; Stehr, H.; von Eyben, R.; Jiang, D.; Ellies, L.G.; et al. Reprogramming the immunological microenvironment through radiation and targeting Axl. Nat. Commun. 2016, 7, 13898. [Google Scholar] [CrossRef] [Green Version]
- Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef] [Green Version]
- Tsukita, Y.; Fujino, N.; Miyauchi, E.; Saito, R.; Fujishima, F.; Itakura, K.; Kyogoku, Y.; Okutomo, K.; Yamada, M.; Okazaki, T.; et al. Axl kinase drives immune checkpoint and chemokine signalling pathways in lung adenocarcinomas. Mol. Cancer 2019, 18, 24. [Google Scholar] [CrossRef] [Green Version]
- Akalu, Y.T.; Rothlin, C.V.; Ghosh, S. TAM receptor tyrosine kinases as emerging targets of innate immune checkpoint blockade for cancer therapy. Immunol. Rev. 2017, 276, 165–177. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Nieto, M.A. Epithelial plasticity: A common theme in embryonic and cancer cells. Science 2013, 342, 1234850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhillon, A.S.; Tulchinsky, E. FRA-1 as a driver of tumour heterogeneity: A nexus between oncogenes and embryonic signalling pathways in cancer. Oncogene 2015, 34, 4421–4428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stemmler, M.P.; Eccles, R.L.; Brabletz, S.; Brabletz, T. Non-redundant functions of EMT transcription factors. Nat. Cell Biol. 2019, 21, 102–112. [Google Scholar] [CrossRef] [PubMed]
- Derynck, R.; Weinberg, R.A. EMT and Cancer: More Than Meets the Eye. Dev. Cell 2019, 49, 313–316. [Google Scholar] [CrossRef]
- Pastushenko, I.; Blanpain, C. EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol. 2019, 29, 212–226. [Google Scholar] [CrossRef] [Green Version]
- Bakir, B.; Chiarella, A.M.; Pitarresi, J.R.; Rustgi, A.K. EMT, MET, Plasticity, and Tumor Metastasis. Trends Cell Biol. 2020, 30, 764–776. [Google Scholar] [CrossRef]
- Pastushenko, I.; Mauri, F.; Song, Y.; de Cock, F.; Meeusen, B.; Swedlund, B.; Impens, F.; Van Haver, D.; Opitz, M.; Thery, M.; et al. Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis. Nature 2021, 589, 448–455. [Google Scholar] [CrossRef]
- Xu, M.Z.; Chan, S.W.; Liu, A.M.; Wong, K.F.; Fan, S.T.; Chen, J.; Poon, R.T.; Zender, L.; Lowe, S.W.; Hong, W.; et al. AXL receptor kinase is a mediator of YAP-dependent oncogenic functions in hepatocellular carcinoma. Oncogene 2011, 30, 1229–1240. [Google Scholar] [CrossRef] [Green Version]
- Sayan, A.E.; Stanford, R.; Vickery, R.; Grigorenko, E.; Diesch, J.; Kulbicki, K.; Edwards, R.; Pal, R.; Greaves, P.; Jariel-Encontre, I.; et al. Fra-1 controls motility of bladder cancer cells via transcriptional upregulation of the receptor tyrosine kinase AXL. Oncogene 2012, 31, 1493–1503. [Google Scholar] [CrossRef] [Green Version]
- Dang, T.T.; Esparza, M.A.; Maine, E.A.; Westcott, J.M.; Pearson, G.W. ΔNp63α Promotes Breast Cancer Cell Motility through the Selective Activation of Components of the Epithelial-to-Mesenchymal Transition Program. Cancer Res. 2015, 75, 3925–3935. [Google Scholar] [CrossRef] [Green Version]
- Rankin, E.B.; Fuh, K.C.; Castellini, L.; Viswanathan, K.; Finger, E.C.; Diep, A.N.; LaGory, E.L.; Kariolis, M.S.; Chan, A.; Lindgren, D.; et al. Direct regulation of GAS6/AXL signaling by HIF promotes renal metastasis through SRC and MET. Proc. Natl. Acad. Sci. USA 2014, 111, 13373–13378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lehmann, W.; Mossmann, D.; Kleemann, J.; Mock, K.; Meisinger, C.; Brummer, T.; Herr, R.; Brabletz, S.; Stemmler, M.P.; Brabletz, T. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat. Commun. 2016, 7, 10498. [Google Scholar] [CrossRef] [PubMed]
- Siemens, H.; Jackstadt, R.; Hünten, S.; Kaller, M.; Menssen, A.; Götz, U.; Hermeking, H. miR-34 and SNAIL form a double-negative feedback loop to regulate epithelial-mesenchymal transitions. Cell Cycle 2011, 10, 4256–4271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mackiewicz, M.; Huppi, K.; Pitt, J.J.; Dorsey, T.H.; Ambs, S.; Caplen, N.J. Identification of the receptor tyrosine kinase AXL in breast cancer as a target for the human miR-34a microRNA. Breast Cancer Res. Treat. 2011, 130, 663–679. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Choi, W.; Porten, S.; Kim, S.; Willis, D.; Plimack, E.R.; Hoffman-Censits, J.; Roth, B.; Cheng, T.; Tran, M.; Lee, I.L.; et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 2014, 25, 152–165. [Google Scholar] [CrossRef] [Green Version]
- Tan, T.Z.; Miow, Q.H.; Huang, R.Y.; Wong, M.K.; Ye, J.; Lau, J.A.; Wu, M.C.; Bin Abdul Hadi, L.H.; Soong, R.; Choolani, M.; et al. Functional genomics identifies five distinct molecular subtypes with clinical relevance and pathways for growth control in epithelial ovarian cancer. EMBO Mol. Med. 2013, 5, 1051–1066. [Google Scholar] [CrossRef]
- Tan, T.Z.; Rouanne, M.; Tan, K.T.; Huang, R.Y.; Thiery, J.P. Molecular Subtypes of Urothelial Bladder Cancer: Results from a Meta-cohort Analysis of 2411 Tumors. Eur. Urol. 2019, 75, 423–432. [Google Scholar] [CrossRef]
- 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]
- 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] [Green Version]
- Colucci-Guyon, E.; Portier, M.M.; Dunia, I.; Paulin, D.; Pournin, S.; Babinet, C. Mice lacking vimentin develop and reproduce without an obvious phenotype. Cell 1994, 79, 679–694. [Google Scholar] [CrossRef]
- Li, Y.; Ye, X.; Tan, C.; Hongo, J.A.; Zha, J.; Liu, J.; Kallop, D.; Ludlam, M.J.; Pei, L. Axl as a potential therapeutic target in cancer: Role of Axl in tumor growth, metastasis and angiogenesis. Oncogene 2009, 28, 3442–3455. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Del Pozo Martin, Y.; Park, D.; Ramachandran, A.; Ombrato, L.; Calvo, F.; Chakravarty, P.; Spencer-Dene, B.; Derzsi, S.; Hill, C.S.; Sahai, E.; et al. Mesenchymal Cancer Cell-Stroma Crosstalk Promotes Niche Activation, Epithelial Reversion, and Metastatic Colonization. Cell Rep. 2015, 13, 2456–2469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, L.; Zhang, Z.; Gao, D.; Luo, J.; Tu, Z.C.; Li, Z.; Peng, L.; Ren, X.; Ding, K. 4-Oxo-1,4-dihydroquinoline-3-carboxamide Derivatives as New Axl Kinase Inhibitors. J. Med. Chem. 2016, 59, 6807–6825. [Google Scholar] [CrossRef] [PubMed]
- Szabadkai, I.; Torka, R.; Garamvölgyi, R.; Baska, F.; Gyulavári, P.; Boros, S.; Illyés, E.; Choidas, A.; Ullrich, A.; Őrfi, L. Discovery of N-[4-(Quinolin-4-yloxy)phenyl]benzenesulfonamides as Novel AXL Kinase Inhibitors. J. Med. Chem. 2018, 61, 6277–6292. [Google Scholar] [CrossRef] [PubMed]
- Davra, V.; Kumar, S.; Geng, K.; Calianese, D.; Mehta, D.; Gadiyar, V.; Kasikara, C.; Lahey, K.C.; Chang, Y.J.; 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] [PubMed]
- 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] [Green Version]
- Kasikara, C.; Davra, V.; Calianese, D.; Geng, K.; Spires, T.E.; Quigley, M.; Wichroski, M.; Sriram, G.; Suarez-Lopez, L.; Yaffe, M.B.; et al. Pan-TAM Tyrosine Kinase Inhibitor BMS-777607 Enhances Anti-PD-1 mAb Efficacy in a Murine Model of Triple-Negative Breast Cancer. Cancer Res. 2019, 79, 2669–2683. [Google Scholar] [CrossRef] [Green Version]
- 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. 2017, 127, 183–198. [Google Scholar] [CrossRef] [Green Version]
- Rankin, E.B.; Fuh, K.C.; Taylor, T.E.; Krieg, A.J.; Musser, M.; Yuan, J.; Wei, K.; Kuo, C.J.; Longacre, T.A.; Giaccia, A.J. AXL is an essential factor and therapeutic target for metastatic ovarian cancer. Cancer Res. 2010, 70, 7570–7579. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Kanlikilicer, P.; Ozpolat, B.; Aslan, B.; Bayraktar, R.; Gurbuz, N.; Rodriguez-Aguayo, C.; Bayraktar, E.; Denizli, M.; Gonzalez-Villasana, V.; Ivan, C.; et al. Therapeutic Targeting of AXL Receptor Tyrosine Kinase Inhibits Tumor Growth and Intraperitoneal Metastasis in Ovarian Cancer Models. Mol. Ther. Nucleic Acids 2017, 9, 251–262. [Google Scholar] [CrossRef] [Green Version]
- Divine, L.M.; Nguyen, M.R.; Meller, E.; Desai, R.A.; Arif, B.; Rankin, E.B.; Bligard, K.H.; Meyerson, C.; Hagemann, I.S.; Massad, M.; et al. AXL modulates extracellular matrix protein expression and is essential for invasion and metastasis in endometrial cancer. Oncotarget 2016, 7, 77291–77305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mills, K.A.; Quinn, J.M.; Roach, S.T.; Palisoul, M.; Nguyen, M.; Noia, H.; Guo, L.; Fazal, J.; Mutch, D.G.; Wickline, S.A.; et al. p5RHH nanoparticle-mediated delivery of AXL siRNA inhibits metastasis of ovarian and uterine cancer cells in mouse xenografts. Sci. Rep. 2019, 9, 4762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, Q.; Grimmig, T.; Gonzalez, G.; Giobbie-Hurder, A.; Berg, G.; Carr, N.; Wilson, B.J.; Banerjee, P.; Ma, J.; Gold, J.S.; et al. ATP-binding cassette member B5 (ABCB5) promotes tumor cell invasiveness in human colorectal cancer. J. Biol. Chem. 2018, 293, 11166–11178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, L.; Lei, Y.; Hou, J.; Wu, J.; Lv, G. Implications of the Receptor Tyrosine Kinase Axl in Gastric Cancer Progression. OncoTargets Ther. 2020, 13, 5901–5911. [Google Scholar] [CrossRef]
- Choi, Y.J.; Kim, J.H.; Rho, J.K.; Kim, J.S.; Choi, C.M.; Kim, W.S.; Son, J.; Lee, J.C. AXL and MET receptor tyrosine kinases are essential for lung cancer metastasis. Oncol. Rep. 2017, 37, 2201–2208. [Google Scholar] [CrossRef] [Green Version]
- Hoj, J.P.; Mayro, B.; Pendergast, A.M. A TAZ-AXL-ABL2 Feed-Forward Signaling Axis Promotes Lung Adenocarcinoma Brain Metastasis. Cell Rep. 2019, 29, 3421–3434.e3428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taverna, J.A.; Hung, C.N.; DeArmond, D.T.; Chen, M.; Lin, C.L.; Osmulski, P.A.; Gaczynska, M.E.; Wang, C.M.; Lucio, N.D.; Chou, C.W.; et al. Single-Cell Proteomic Profiling Identifies Combined AXL and JAK1 Inhibition as a Novel Therapeutic Strategy for Lung Cancer. Cancer Res. 2020, 80, 1551–1563. [Google Scholar] [CrossRef] [Green Version]
- Shibue, T.; Weinberg, R.A. EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 611–629. [Google Scholar] [CrossRef] [Green Version]
- Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
- Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.C.; Choi, H.; El Rayes, T.; Ryu, S.; Troeger, J.; et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef]
- Sharma, S.V.; Lee, D.Y.; Li, B.; Quinlan, M.P.; Takahashi, F.; Maheswaran, S.; McDermott, U.; Azizian, N.; Zou, L.; Fischbach, M.A.; et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 2010, 141, 69–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tulchinsky, E.; Demidov, O.; Kriajevska, M.; Barlev, N.A.; Imyanitov, E. EMT: A mechanism for escape from EGFR-targeted therapy in lung cancer. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 29–39. [Google Scholar] [CrossRef]
- De Conti, G.; Dias, M.H.; Bernards, R. Fighting Drug Resistance through the Targeting of Drug-Tolerant Persister Cells. Cancers 2021, 13, 1118. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, M.; Siemann, D.W. Gas6/Axl Signaling Pathway in the Tumor Immune Microenvironment. Cancers 2020, 12, 1850. [Google Scholar] [CrossRef] [PubMed]
- Mohme, M.; Riethdorf, S.; Pantel, K. Circulating and disseminated tumour cells—Mechanisms of immune surveillance and escape. Nat. Rev. Clin. Oncol. 2017, 14, 155–167. [Google Scholar] [CrossRef]
Cancer Type | Type of Study | Method of AXL Inhibition | Therapeutic Agent | Drug Class | Reference |
---|---|---|---|---|---|
Breast cancer | (*) CDX | mAb (YW327.6S2) | Anti-VEGF mAb | VEGFR-Targeted therapy | [60] |
Breast cancer | Cell lines-derived xenografts (CDX) | AXL shRNA, AXLi (R428) | Doxorubicin | Chemotherapy | [61] |
Ovarian cancer | Mouse CDX | AXLi (R428) | Anti-PD-1 mAb | Immunotherapy | [62] |
Lung cancer | CDX | mAb (YW327.6S2) | Erlotinib | EGFR-Targeted therapy | [60] |
Lung cancer | CDX | mAb (YW327.6S2) | Paclitaxel + Carboplatin | Chemotherapy | [60] |
Lung cancer, NSCLC | CDX | AXL shRNA, AXLi | Erlotinib | EGFR-targeted therapy | [63] |
Lung cancer, SCLC | CDX | AXLi (TP0903) | AZD1775 | WEE1-targeted therapy | [64] |
Lung cancer, breast cancer | Mouse CDX, orthotopic model for breast cancer | TAMi (Sitravatinib) | Anti-PD-1 mAb (Nivolumab) | Immunotherapy | [65] |
Lung cancer, NSCLC | CDX, (**) PDX | AXLi (NPS1034) | Osimertinib, | EGFR-targeted therapy | [66] |
Lung cancer, NSCLC | Human CDX | AXLi (ONO-7475) | Osimertinib | EGFR-targeted therapy | [67] |
Esophageal adenocarcinoma | Mouse CDX | AXLi (R428) | Epirubicin | Chemotherapy | [68] |
Pancreatic cancer | Transgenic model (KrasLSL-G12D; Cdkn2alox/lox; Ptf1aCre/+); orthotopic model | AXLi (R428) | Gemcitabine | Chemotherapy | [69] |
Head and Neck Cancer | PDX | AXLi (R428) | Cetuximab or radiation | EGFR-targeted therapy or radiotherapy | [70] |
Cutaneous melanoma | PDX | (***) AXL-107-MMAE (or EnaV) | Vemurafenib + Trametinib | BRAFV600E + MEK − targeted therapy | [71] |
Cutaneous melanoma | PDX | AXLi (R428) | AZD7762 | CHK1/CHK2-targeted therapy | [72] |
Cutaneous melanoma; lung cancer | CDX, PDX | (***) AXL-107-MMAE (or EnaV | Anti-PD-1 mAb (Pembrolizumab) | Immunotherapy | [73] |
Glioblastoma | Mouse spheroid-derived xenografts | AXLi (R428) | Anti-PD-1 mAb (Nivolumab) | Immunotherapy | [9] |
Diffuse intrinsic pontine glioma | PDX; mouse allografts | AXLi (R428) | Panobinostat | Histone deacetylase inhibitors | [74] |
Cancer Type | Type of Study | Method of AXL Inhibition | Reference |
---|---|---|---|
Breast cancer | Orthotopic model | shRNA | [102] |
Breast cancer | Intravenous metastasis assay | Anti-AXL mAb YW327.6S2 | [60] |
Breast cancer | Orthotopic model and lung metastasis assay | AXLi R428 | [103] |
Breast cancer | Lung metastasis assay | AXLi R428; shRNA | [104] |
Breast cancer | (*) CDX | AXLi R428; shRNA | [101] |
Breast cancer | CDX | AXLi | [105] |
Breast cancer | CDX | AXLi | [106] |
Breast cancer | Orthotopic model | CRISPR/Cas9 gene inactivation | [107] |
Triple-Negative Breast Cancer | Orthotopic model | shRNAi | [99] |
Triple-Negative Breast Cancer | CDX or (**) PDX | Anti-AXL mAb 20G7-D9 | [108] |
Triple-Negative Breast Cancer | CDX | pan-TAM kinase inhibitor BMS-777607 | [109] |
HER2 + Breast Cancer | Genetically modified mice | Germline knockout | [27] |
Breast cancer, cutaneous melanoma | CDX | Pan-TAM inhibitor LDC1267 | [80] |
Ovarian, breast, pancreatic cancer | CDX | MYD1-72 Fc decoy receptor | [110] |
Ovarian cancer | Peritoneal xenografts | shRNA, sAXL (acts as a decoy receptor) | [111] |
Ovarian cancer | Intraperitoneal injections | MYD1 Fc decoy receptor | [112] |
Ovarian cancer | CDX | AXL-aptamer | [113] |
Endometrial cancer | Orthotopic | shRNA | [114] |
Uterine cancer | CDX | siRNA | [115] |
Pancreatic cancer | CDX | Anti-AXL mAb 10C9 | [100] |
Colorectal cancer | CDX (effect on dissemination in the bloodstream) | Ectopic expression of AXL | [116] |
Gastric cancer | CDX | shRNA | [117] |
Clear cell renal cell carcinoma | Lung metastasis assay | shRNA, sAXL-IgG1 fusion | [91] |
NSCLC | Lung metastasis assay | shRNA | [118] |
NSCLC | Intracardiac injections | shRNA | [119] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
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. https://doi.org/10.3390/cancers13194864
Auyez A, Sayan AE, Kriajevska M, Tulchinsky E. AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew. Cancers. 2021; 13(19):4864. https://doi.org/10.3390/cancers13194864
Chicago/Turabian StyleAuyez, Almira, A. Emre Sayan, Marina Kriajevska, and Eugene Tulchinsky. 2021. "AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew" Cancers 13, no. 19: 4864. https://doi.org/10.3390/cancers13194864
APA StyleAuyez, A., Sayan, A. E., Kriajevska, M., & Tulchinsky, E. (2021). AXL Receptor in Cancer Metastasis and Drug Resistance: When Normal Functions Go Askew. Cancers, 13(19), 4864. https://doi.org/10.3390/cancers13194864