Next Article in Journal
Anti-Amnesic Effect of Synbiotic Supplementation Containing Corni fructus and Limosilactobacillus reuteri in DSS-Induced Colitis Mice
Next Article in Special Issue
Establishment of Mucoepidermoid Carcinoma Cell Lines from Surgical and Recurrence Biopsy Specimens
Previous Article in Journal
Vitamin D in Neurological Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

May EPH/Ephrin Targeting Revolutionize Lung Cancer Treatment?

by
Iason Psilopatis
1,2,
Ioannis Karniadakis
3,
Konstantinos Stylianos Danos
1,
Kleio Vrettou
1,
Kleita Michaelidou
4,
Konstantinos Mavridis
5,
Sofia Agelaki
4,6,* and
Stamatios Theocharis
1,*
1
First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10, Goudi, 11527 Athens, Greece
2
Department of Gynecology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt—Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
3
Second Department of Propaedeutic Surgery, “Laiko” General Hospital, 17 Agiou Thoma Street, 11527 Athens, Greece
4
Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, 71003 Herakleion, Greece
5
Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, 70013 Herakleion, Greece
6
Department of Medical Oncology, University General Hospital of Herakleion, Vassilika Vouton, 71110 Herakleion, Greece
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 93; https://doi.org/10.3390/ijms24010093
Submission received: 4 November 2022 / Revised: 10 December 2022 / Accepted: 17 December 2022 / Published: 21 December 2022
(This article belongs to the Special Issue Molecular Advances in Cancer Therapy 2.0)

Abstract

:
Lung cancer (LC) is the leading cause of cancer death in the United States. Erythropoietin-producing hepatocellular receptors (EPHs) comprise the largest receptor tyrosine kinases (RTKs) family in mammals. EPHs along with their ligands, EPH-family receptor-interacting proteins (ephrins), have been found to be either up- or downregulated in LC cells, hence exhibiting a defining role in LC carcinogenesis and tumor progression. In their capacity as membrane-bound molecules, EPHs/ephrins may represent feasible targets in the context of precision cancer treatment. In order to investigate available therapeutics targeting the EPH/ephrin system in LC, a literature review was conducted, using the MEDLINE, LIVIVO, and Google Scholar databases. EPHA2 is the most well-studied EPH/ephrin target in LC treatment. The targeting of EPHA2, EPHA3, EPHA5, EPHA7, EPHB4, EPHB6, ephrin-A1, ephrin-A2, ephrin-B2, and ephrin-B3 in LC cells or xenograft models not only directly correlates with a profound LC suppression but also enriches the effects of well-established therapeutic regimens. However, the sole clinical trial incorporating a NSCLC patient could not describe objective anti-cancer effects after anti-EPHA2 antibody administration. Collectively, EPHs/ephrins seem to represent promising treatment targets in LC. However, large clinical trials still need to be performed, with a view to examining the effects of EPH/ephrin targeting in the clinical setting.

1. Introduction

Lung cancer (LC) represents the second most common malignancy and the leading cancer death cause in the United States, with the mean age of patients upon diagnosis at 70 years of age [1]. According to the American Cancer Society, about 236,740 new cases of LC will be diagnosed and about 130,180 patients will die from LC in the United States in 2022 [1]. LC is divided into two distinct histologic classes: the more aggressive small-cell LC (SCLC) and the more common non-small-cell LC (NSCLC) [2]. The main NSCLC subtypes are adenocarcinoma, squamous cell carcinoma, and large cell carcinoma [2]. Pulmonary symptoms of LC predominantly include cough, hemoptysis, dyspnea, or chest pain, while extrapulmonary symptoms are constitutional symptoms, compression of neighboring structures, and paraneoplastic syndromes [3]. The diagnostic evaluation of LC includes, in addition to a physical examination, a chest X-ray as a first-line imaging study, and a computed tomography (CT) scan of the chest in cases of abnormal X-ray. Definite diagnosis always requires biopsy of the tumor mass. Advanced studies for LC staging involve a CT of the thorax and abdomen, brain imaging, and positron emission tomography (PET)-CT [4]. For patients with non-metastatic resectable NSCLC, surgical excision, followed by chemotherapy, represents the first-line therapy. Non-surgical patients are mostly treated with chemotherapy plus radiotherapy, and possibly immunotherapy, whereas molecular-targeted treatment may be offered in cases of targetable mutation [5]. Limited-stage SCLC always requires systemic therapy and concurrent radiotherapy, whereas chemotherapy with or without immunotherapy is the proposed therapeutic regime for extensive-stage SCLC [6].
Erythropoietin-producing hepatocellular receptors (EPHs) comprise the leading subfamily of receptor tyrosine kinases (RTKs), which bind the membrane-bound proteins, ephrins [7]. EPHs are categorized into two subgroups, EPHAs and EPHBs, depending on their structural homology and preferential binding affinities to their respective ligands, ephrin-A and ephrin-B [8,9]. Specifically, EPHA binds ephrin-A via a glycosylphosphatidylinositol anchor on a plasma membrane, whereas ephrin-B interacts with EPHB via a transmembrane domain [10]. In humans, nine EPHA receptors (EPHA1-8, 10), which interact with five ephrin-A ligands (ephrin-A1-5), along with five EPHB receptors (EPHB1-4, 6), which bind three ephrin-B ligands (ephrin-B1-3), have been defined [11]. The EPH–ephrin interaction prompts both forward signaling in the receptor-expressing cell, as well as backward signaling in the ephrin-bearing cell [12]. EPHs/ephrins show a wide expression in various cell types, and influence diverse physiological functions related to cell migration, cell–cell or cell–matrix interactions, or (lymph-) angiogenesis, thus potentially exerting both tumor-promoting and tumor-suppressive properties [12,13,14,15,16,17,18,19,20].
Over the past decade, a large number of original articles has been published on the role of different members of the EPH/ephrin system in the pathogenesis and progression of LC; moreover, these studies described their implication in (N)SCLC molecular pathways, immune evasion, metastasis, recurrence, and clinicopathological features, as well as patient survival and prognosis [21,22,23,24,25,26,27,28,29,30,31,32,33,34]. Recently, both Kou et al. and Anderton et al. extensively reviewed the implication of diverse EPHs/ephrins in LC and outlined the tumor-promoting effects of EPHA1, EPHA2, EPHA4, EPHA5, EPHA7, EPHB3, EPHB4, ephrin-A3, and ephrin-B2, as well as the tumor-suppressive effects of EPHA3, EPHB6, and ephrin-B3 [35,36]. Inspired by these observations, and given the current advancements in the field of targeted therapy, several study groups have recently attempted to investigate the feasibility of targeting the EPH/ephrin system in LC. The present review summarizes the results of all available relevant studies and presents newly developed agents that target the various members of the EPH/ephrin system in LC.

2. Available EPH/Ephrin-Targeting Agents in LC

Since their clinical introduction in 2001, tyrosine kinase inhibitors (TKIs) represent potent pharmacologic agents that target the active site of RTKs and, thereby, either reversibly or irreversibly, inhibit the phosphorylation of intracellular targets [37]. Imatinib became the first Food and Drug Administration (FDA)-approved TKI for the treatment of Philadelphia chromosome–positive chronic myeloid leukemia-targeting Bcr-Abl, thus not only revolutionizing treatment for these patients but also laying the foundation for the use of targeted therapeutics [38]. To date, the FDA has approved a large number of TKIs for mostly epidermal growth factor receptor (EGFR)-mutated NSCLC [39]. Given that EPHs represent a subgroup of RTKs, consequently, dasatinib, osimertinib, ensartinib, ALW–II–41-27, and XL647, as well as PKC 412, seem to represent promising TKIs for EPH-expressing LC.
Short interfering RNAs (siRNAs) comprise a class of regulatory small RNA molecules that modify the stability or translational efficacy of messenger RNAs (mRNAs), thus silencing any disease-related genes in a sequence-specific manner [40]. siRNAs are currently widely used in diverse types of cancers, such as pancreatic, breast, colorectal, ovarian, hepatocellular, gastric, and cervical cancer [41]. In LC, siRNAs have already shown promising effects in preclinical and early clinical evaluations of both SCLC and NSCLC [42], thus paving the way for their employment in EPH-expressing LC.
By precisely binding to antigens on the surface of cancer cells, monoclonal antibodies (mAbs) represent laboratory-produced proteins that provoke long-lasting anti-cancer immune responses [43]. In the same context, immunoliposomes can be generated by antibody coupling to the liposomal surface, thus facilitating active tissue targeting [44]. The FDA has, to date, approved several mAbs for the treatment of both SCLC and NSCLC [45]. EPHs/ephrins, in their capacity as membrane-bound proteins, may, consequently, also serve as feasible targets of mAbs or immunoliposomes in LC.
Albumin represents a highly soluble and stable drug delivery system with a long circulatory half-life and high intratumoral accumulation due to its enhanced permeability and retention effect [46]. After its approval in 2005, Abraxane became the first and sole FDA-approved paclitaxel–albumin nanoparticle for the treatment of metastatic breast cancer, locally advanced or metastatic NSCLC, and metastatic pancreatic adenocarcinoma [47]. In EPH/ephrin-expressing LC, EPH/ephrin-loaded albumin spheres could, hence, possibly represent a novel therapeutic alternative.
Core–shell nanoparticles allow for effective and sustained drug delivery to tumors by offering the core protection from the surrounding environment, a versatile surface for targeting group binding, and an enhanced nanostructure bioavailability [48]. In LC treatment, core–shell nanoparticles have been successfully employed for simultaneous chemotherapy and radiation sensitization [49,50]. Interestingly, EPHs/ephrins might act as targeting moieties on these nanoparticles and improve their therapeutic efficacy against LC cells.
Taspine is an alkaloid that acts as a potent cicatrizant [51]. In addition to its other various biological properties, taspine, along with its derivatives, exhibits profound anti-cancer effects in diverse cancer entities, including LC [52,53,54,55]. After Dai et al. described that taspine derivative 12k suppresses colorectal cancer cell growth by competitive targeting of ephrin-B2-related pathways, EPH/ephrin-expressing LC also arose as a feasible target of taspine anti-tumor treatment [56].
The main available therapeutics targeting the EPH/ephrin system in LC are depicted in Table 1.

3. EPHA-Targeting Therapeutic Agents

A large number of original research articles has, to date, been published on EPHA-targeting treatment agents.
Bai et al. assessed multiple LC patient cohorts and defined EPHA mutation as an anti-programmed death-1 (PD-1) ligand 1 (PD-L1) efficacy predictor, given the prolonged progression-free survival (PFS) and overall survival (OS) of EPHAmut lung adenocarcinoma patients. Specifically, EPHAmut correlated with higher T-cell signatures and reduced transforming growth factor (TGF)-β signaling in those patients [57].
EPHA2 represents the most studied member of the EPH/ephrin family in LC therapy. So far, several study groups have reported on the association of EPHA2 with EGFR kinase inhibitors in LC. Li et al. reported strong EPHA2 enrichment by dasatinib [58], while Choi et al. not only confirmed that both gefitinib and erlotinib reciprocally regulated EPHA2 expression in TKI-sensitive LC cells, but also treated EGFR-mutant HCC827 cells with the recombinant EPHA2-Fc peptide, thus inhibiting anchorage-independent NSCLC cell growth [59]. Furthermore, Koch et al. employed kinase affinity purification and quantitative mass spectrometry and proposed high EPHA2 expression in gefitinib-resistant HCC827 LC cells. Gefitinib sensitivity was only restored after siRNA-mediated EPHA2 knock-down or dasatinib application, which lessened focal adhesion kinase (FAK) phosphorylation and cell migration [60]. Srivastava et al. showed that the EPHA2 TKI ensartinib may synergistically act with the roundabout guidance receptor 1 (ROBO1) ligand slit guidance ligand 2 (SLIT2) to attenuate the growth of squamous LC cells, which exercises its tumor-suppressive effect by either EPHA2 heterodimerizing or homodimer binding and inhibits AKT activity [61]. Interestingly, Amato et al. described EPHA2 overexpression in erlotinib-resistant LC cells, and suggested loss of EPHA2 after genetic ablation in vitro and gene targeting in vivo, to account for apoptosis induction, decreased proliferation, as well as LC growth repression. Notably, the use of the EPHA2 small-molecule inhibitor ALW-II-41-27 showed similar results [62].
Ishigaki et al. presented cell-cycle arrest through the dephosphorylation of retinoblastoma (Rb) as the underlying mechanism of the proliferation inhibition of EPHA2-expressing SCLC cells, after both genetic siRNA EPHA2 inhibition and ALW-II-41-27/dasatinib treatment [63]. Additionally, Kaminskyy et al. targeted EPHA2 expression with specific siRNA in NSCLC cell lines, and highlighted the amplification of apoptotic signaling after an EPHA2 knockdown combination with ionizing radiation due to the partial phosphorylation reduction of the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) [64].
Two independent research groups also investigated the plausibility of nanoparticle administration in EPHA2-expressing LC. Iyer et al. generated dual-stimuli nanoparticles (E-DSNPs) loaded with cisplatin and a radiation sensitizer and functionalized with anti-EPHA2 antibodies, which selectively targeted EPHA2-expressing NSCLC cells. E-DSNPs demonstrated a triggered release of the radiosensitizer during concurrent radiation therapy, followed by chemo-drug release upon glutathione exposure, thus downregulating the in vitro LC cell survival fraction [65]. Based on the assumption that the EPHA2–ephrin-A1 interaction attenuates NSCLC growth and survival, Lee et al. used albumin mesosphere-conjugated ephrin-A1 in vivo, and suggested apoptosis induction, along with tumor shrinkage, in mouse NSCLC xenograft models [66].
Gan et al. launched a phase I clinical trial of the anti-EPHA2 DS-8895a antibody in patients with advanced EPHA2-expressing epithelial cancers, one of whom was diagnosed with NSCLC. Although DS-8895a was well tolerated at the evaluated doses, the 89Zr trace-labelled infusion of DS-8895a demonstrated specific low-grade tumor uptake, while no objective tumor responses were noticed [67].
So far, two study groups have assessed the role of EPHA3 in LC treatment. Peng et al. explored the role of EPHA3 in multidrug resistance and observed EPHA3 overexpression to be associated with reduced phosphorylation of the PI3K/BMX/STAT3 signaling pathway, cell-cycle arrest, apoptosis induction, as well as chemosensitivity in SCLC [68]. Furthermore, Sos et al. grew EPHA3-amplified HCC515 NSCLC cells and demonstrated significant in vivo NSCLC shrinkage after dasatinib treatment [69].
Staquicini et al. created the anti-EPHA5 mAb 11C12 and suggested that 11C12 sensitized LC cells to radiotherapy both in vitro and in vivo, and showed a combined anti-proliferative/pro-senescence effect, which significantly prolonged OS [70].
By siRNA-mediated EPHA7 silencing, Li et al. managed to inhibit NSCLC cell proliferation, migration, and invasion; enhance B-cell lymphoma 2 (Bcl-2) -associated X protein, caspase-3, and phosphatase and tensin homolog (PTEN) expression levels; as well as downregulate phosphorylated-AKT levels [71].
Altogether, EPHA2, EPHA3, EPHA5, and EPHA7, seem to represent feasible targets for newly developed targeted treatment agents in (N)SCLC. Therapeutic up- or downregulation of EPHA2, EPHA3, EPHA5, and EPHA7, not only directly influence complex molecular pathways related to LC cell survival and progression, but also enhance the therapeutic efficacy of well-established treatment regimens (chemotherapy/radiotherapy/targeted therapy) for advanced LC.

4. EPHB-Targeting Therapeutic Agents

EPHB4 represents the most studied EPHB in LC treatment. Ferguson et al. first described that the combination of siRNA-mediated EPHB4 knockdown with topoisomerase I inhibition diminished SCLC cell viability in vitro, while, when co-applied with paclitaxel, inducing tumor regression in vivo [72]. In NSCLC, EPHB4 was found to be significantly downregulated by osimertinib [73].
Yoon et al. underlined that EPHB6 mutation induces cell adhesion-mediated paclitaxel resistance via EPHA2 and CDH11 expression in LC and proposed ALW-II-41-27 therapy to suppress EPHB6 (Q926R)-induced CDH11 expression and focal adhesion formation [74].
All in all, therapeutic downregulation of EPHB4 and EPHB6 seems to specifically improve paclitaxel efficacy in LC.

5. Ephrin-Targeting Therapeutic Agents

Ephrin-A1, -A2, -B2, and -B3, have, to date, been suggested as feasible targets of treatment agents in LC.
In 2011, Lee et al. published their first research paper on targeted LC therapy using ephrin-A1-loaded albumin microspheres and underlined that albumin microspheres exhibited low toxicity for A549 cells, as well as rapid phagocytosis after incubation. Of note, these microspheres effectively reduced FAK expression, NSCLC cell migration, and tumor growth in matrigels [75]. Two years later, the same study group published their second article on the targeted delivery of highly stable, low-cytotoxicity let-7a microRNA-encapsulated ephrin-A1-conjugated liposomal nanoparticles in LC, which resulted in repressed Ras expression, as well as reduced NSCLC proliferation, migration, and tumor growth [76]. By achieving this breakthrough, Lee and coworkers reached a milestone and they still remain the first and only research group to have employed RNA-based therapeutics targeting the EPH/ephrin system in LC treatment. In this context, Murugan et al. recently published their comprehensive review article on up- and downregulated microRNAs in LC, and outlined the current advancements in the development of relevant microRNA-based therapeutics [77]. Importantly, the authors focused on the compelling need to ensure tumor tissue-specific delivery, given the unwanted off-target effects of microRNA-based suppression and replacement therapy, and accentuated the fact that, to date, except for the numerous microRNA delivery strategies in preclinical stages, there are only two registered clinical trials but no FDA-approved microRNA LC therapeutics [77].
Huang et al. generated the ephrin-A2-targeted taxane liposomal prodrug 2. This immunoliposome displayed an equilibrium dissociation constant toward the extracellular ephrin-A2 receptor domain, while its application in the NSCLC xenograft model A549 resulted in a profound tumor regression [78].
In addition, Dai et al. reported that the taspine derivative 12k repressed the ephrin-B2 and its PDZ protein, thus impairing vascular endothelial growth factor receptor (VEGFR)-2/-3 expression and suppressing A549 cell migration [79].
Finally, siRNA-mediated ephrin-B3 silencing led to decreased NSCLC U-1810 cell proliferation, as well as a morphological change from a small-round to a flattened-elongated appearance [80].
Taken altogether, ephrin-A1, and -A2, seem to represent ideal targets for drug delivery in LC, while pharmacologic downregulation of ephrin-B2, and -B3, directly suppresses LC cell proliferation and migration.
The effects of different therapeutic agents on the EPH/ephrin system in LC are summarized in Table 2.

6. Conclusions

Given its high prevalence and mortality, LC has long been the focus of diverse studies on the development of potent anti-cancer drugs. Despite the therapeutic advancements over the past 20 years, which have undoubtedly revolutionized anti-LC treatment, 5-year relative survival rates amount to 26% and 7% for NSCLC and SCLC, respectively, for all Surveillance, Epidemiology, and End Results (SEER) stages combined [81]. Consequently, the generation and establishment of effective novel therapeutic regimens are of utmost importance. In this context, the EPH/ephrin system seems to represent an achievable and most promising target for anti-LC treatment, and numerous research groups have already outlined the feasibility of EPH/ephrin targeting in other cancer entities [12,15,16,18]. In this review, we were able to identify EPHA2, EPHA3, EPHA5, EPHA7, EPHB4, EPHB6, ephrin-A1, ephrin-A2, ephrin-B2, and ephrin-B3 as potential targets of novel therapeutic agents, alone or in combination with well-established therapies (Figure 1 and Figure 2), and to demonstrate that their pharmaceutical up- or downregulation may lead to profound (N)SCLC suppression both in vitro and in vivo. Most significantly, EPH/ephrin targeting either restituted or improved LC cell sensitivity to chemo-, radio-, and targeted therapy, thus minimizing the quota of LC patients not profiting from already-FDA-approved treatments, which experts have sufficient experience with. Unfortunately, the only clinical trial incorporating a patient with advanced NSCLC could not describe objective anti-tumor effects after anti-EPHA2 DS-8895a antibody administration, hence questioning the results of the preclinical studies. As such, further clinical studies using larger patient collectives need to be conducted in order to verify the clinical utility and safety of EPH/ephrin-targeting agents in LC. Ideally, more emphasis should be given on the more aggressive SCLC subtype, which correlates with a worse prognosis and OS [81]. In summary, EPHs/ephrins seem to represent novel auspicious targets in modern LC therapy.

Author Contributions

Literature analysis and conceptualization, I.P. and S.T.; original draft preparation and writing, I.P.; art work, I.P.; review and supervision, I.K., K.S.D., K.V., K.M. (Kleita Michaelidou), K.M. (Konstantinos Mavridis), S.A. and S.T. All authors have read and agreed to the published version of the manuscript.

Funding

This project has received funding from the Special Account for Research Grants, University of Crete (Grant No. 11026).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Key Statistics for Lung Cancer; American Cancer Society: Atlanta, GA, USA, 2022.
  2. What Is Lung Cancer? American Cancer Society: Atlanta, GA, USA, 2019.
  3. Signs and Symptoms of Lung Cancer; American Cancer Society: Atlanta, GA, USA, 2019.
  4. Tests for Lung Cancer; American Cancer Society: Atlanta, GA, USA, 2022.
  5. Treatment Choices for Non-Small Cell Lung Cancer, by Stage; American Cancer Society: Atlanta, GA, USA, 2022.
  6. Treatment Choices for Small Cell Lung Cancer, by Stage; American Cancer Society: Atlanta, GA, USA, 2021.
  7. Eph Nomenclature Committee. Unified nomenclature for Eph family receptors and their ligands, the ephrins. Cell 1997, 90, 403–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Wei, Q.; Liu, J.; Wang, N.; Zhang, X.; Jin, J.; Chin-Sang, I.; Zheng, J.; Jia, Z. Structures of an Eph receptor tyrosine kinase and its potential activation mechanism. Acta Crystallogr. Sect. D Biol. Crystallogr. 2014, 70, 3135–3143. [Google Scholar] [CrossRef] [PubMed]
  9. Kania, A.; Klein, R. Mechanisms of ephrin-Eph signalling in development, physiology and disease. Nat. Rev. Mol. Cell Biol. 2016, 17, 240–256. [Google Scholar] [CrossRef] [PubMed]
  10. Yamada, T.; Yuasa, M.; Masaoka, T.; Taniyama, T.; Maehara, H.; Torigoe, I.; Yoshii, T.; Shinomiya, K.; Okawa, A.; Sotome, S. After repeated division, bone marrow stromal cells express inhibitory factors with osteogenic capabilities, and EphA5 is a primary candidate. Bone 2013, 57, 343–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Shiuan, E.; Chen, J. Eph Receptor Tyrosine Kinases in Tumor Immunity. Cancer Res. 2016, 76, 6452–6457. [Google Scholar] [CrossRef] [Green Version]
  12. Psilopatis, I.; Pergaris, A.; Vrettou, K.; Tsourouflis, G.; Theocharis, S. The EPH/Ephrin System in Gynecological Cancers: Focusing on the Roots of Carcinogenesis for Better Patient Management. Int. J. Mol. Sci. 2022, 23, 3249. [Google Scholar] [CrossRef]
  13. Coulthard, M.G.; Duffy, S.; Down, M.; Evans, B.; Power, M.; Smith, F.; Stylianou, C.; Kleikamp, S.; Oates, A.; Lackmann, M.; et al. The role of the Eph-ephrin signalling system in the regulation of developmental patterning. Int. J. Dev. Biol. 2002, 46, 375–384. [Google Scholar]
  14. Rudno-Rudzinska, J.; Kielan, W.; Frejlich, E.; Kotulski, K.; Hap, W.; Kurnol, K.; Dzierżek, P.; Zawadzki, M.; Hałoń, A. A review on Eph/ephrin, angiogenesis and lymphangiogenesis in gastric, colorectal and pancreatic cancers. Chin. J. Cancer Res. 2017, 29, 303–312. [Google Scholar] [CrossRef] [Green Version]
  15. Pergaris, A.; Danas, E.; Goutas, D.; Sykaras, A.G.; Soranidis, A.; Theocharis, S. The Clinical Impact of the EPH/Ephrin System in Cancer: Unwinding the Thread. Int. J. Mol. Sci. 2021, 22, 8412. [Google Scholar] [CrossRef]
  16. Papadakos, S.P.; Petrogiannopoulos, L.; Pergaris, A.; Theocharis, S. The EPH/Ephrin System in Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 2761. [Google Scholar] [CrossRef]
  17. Masaoutis, C.; Georgantzoglou, N.; Sarantis, P.; Theochari, I.; Tsoukalas, N.; Bobos, M.; Alexandrou, P.; Pergaris, A.; Rontogianni, D.; Theocharis, S. Ephrin Receptors (Ephs) Expression in Thymic Epithelial Tumors: Prognostic Implications and Future Therapeutic Approaches. Diagnostics 2021, 11, 2265. [Google Scholar] [CrossRef] [PubMed]
  18. Hadjimichael, A.C.; Pergaris, A.; Kaspiris, A.; Foukas, A.F.; Kokkali, S.; Tsourouflis, G.; Theocharis, S. The EPH/Ephrin System in Bone and Soft Tissue Sarcomas’ Pathogenesis and Therapy: New Advancements and a Literature Review. Int. J. Mol. Sci. 2022, 23, 5171. [Google Scholar] [CrossRef] [PubMed]
  19. Pasquale, E.B. Eph receptors and ephrins in cancer: Bidirectional signalling and beyond. Nat. Rev. Cancer 2010, 10, 165–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Goutas, D.; Pergaris, A.; Goutas, N.; Theocharis, S. Utilizing Exosomal-EPHs/Ephrins as Biomarkers and as a Potential Platform for Targeted Delivery of Therapeutic Exosomes. Int. J. Mol. Sci. 2022, 23, 3551. [Google Scholar] [CrossRef]
  21. Zhao, W.; Liu, L.; Li, X.; Xu, S. EphA10 drives tumor progression and immune evasion by regulating the MAPK/ERK cascade in lung adenocarcinoma. Int. Immunopharmacol. 2022, 110, 109031. [Google Scholar] [CrossRef]
  22. Zhao, X.; Chen, Y.; Sun, X.; He, Z.; Wu, T.; Wu, C.; Chen, J.; Wang, J.; Diao, K.; Liu, X. Oncogenic EFNA4 Amplification Promotes Lung Adenocarcinoma Lymph Node Metastasis. Cancers 2022, 14, 4226. [Google Scholar] [CrossRef]
  23. Yiminniyaze, R.; Zhang, X.; Zhu, N.; Wang, J.; Li, C.; Wumaier, G.; Zhou, D.; Li, J.; Xia, J.; Zhang, Y.; et al. EphrinA3 is a key regulator of malignant behaviors and a potential prognostic factor in lung adenocarcinoma. Cancer Med. 2022. [Google Scholar] [CrossRef]
  24. Deng, M.; Tong, R.; Zhang, Z.; Wang, T.; Liang, C.; Zhou, X.; Hou, G. EFNA3 as a predictor of clinical prognosis and immune checkpoint therapy efficacy in patients with lung adenocarcinoma. Cancer Cell Int. 2021, 21, 535. [Google Scholar] [CrossRef]
  25. Wang, L.; Peng, Q.; Sai, B.; Zheng, L.; Xu, J.; Yin, N.; Feng, X.; Xiang, J. Ligand-independent EphB1 signaling mediates TGF-beta-activated CDH2 and promotes lung cancer cell invasion and migration. J. Cancer 2020, 11, 4123–4131. [Google Scholar] [CrossRef] [Green Version]
  26. Volz, C.; Breid, S.; Selenz, C.; Zaplatina, A.; Golfmann, K.; Meder, L.; Dietlein, F.; Borchmann, S.; Chatterjee, S.; Siobal, M.; et al. Inhibition of Tumor VEGFR2 Induces Serine 897 EphA2-Dependent Tumor Cell Invasion and Metastasis in NSCLC. Cell Rep. 2020, 31, 107568. [Google Scholar] [CrossRef]
  27. Li, J.; Sun, Y.; Wang, X.; Wang, J.; Zhu, Y. The expressions of EphB4 and ephrinB2 in lung adenocarcinomas: A high level of the EphB4 protein is associated with lymph node metastasis. Int. J. Clin. Exp. Pathol. 2019, 12, 3447–3452. [Google Scholar] [PubMed]
  28. Zhao, C.; Wang, A.; Lu, F.; Chen, H.; Fu, P.; Zhao, X.; Chen, H. Overexpression of junctional adhesion molecule-A and EphB2 predicts poor survival in lung adenocarcinoma patients. Tumour Biol. 2017, 39, 1010428317691000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Efazat, G.; Novak, M.; Kaminskyy, V.O.; De Petris, L.; Kanter, L.; Juntti, T.; Bergman, P.; Zhivotovsky, B.; Lewensohn, R.; Hååg, P.; et al. Ephrin B3 interacts with multiple EphA receptors and drives migration and invasion in non-small cell lung cancer. Oncotarget 2016, 7, 60332–60347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Giaginis, C.; Tsoukalas, N.; Bournakis, E.; Alexandrou, P.; Kavantzas, N.; Patsouris, E.; Theocharis, S. Ephrin (Eph) receptor A1, A4, A5 and A7 expression in human non-small cell lung carcinoma: Associations with clinicopathological parameters, tumor proliferative capacity and patients’ survival. BMC Clin. Pathol. 2014, 14, 8. [Google Scholar] [CrossRef] [Green Version]
  31. Saintigny, P.; Peng, S.; Zhang, L.; Sen, B.; Wistuba, I.I.; Lippman, S.M.; Girard, L.; Minna, J.D.; Heymach, J.V.; Johnson, F.M. Global evaluation of Eph receptors and ephrins in lung adenocarcinomas identifies EphA4 as an inhibitor of cell migration and invasion. Mol. Cancer Ther. 2012, 11, 2021–2032. [Google Scholar] [CrossRef] [Green Version]
  32. Li, G.; Ji, X.D.; Gao, H.; Zhao, J.S.; Xu, J.F.; Sun, Z.J.; Deng, Y.Z.; Shi, S.; Feng, Y.X.; Zhu, Y.Q.; et al. EphB3 suppresses non-small-cell lung cancer metastasis via a PP2A/RACK1/Akt signalling complex. Nat. Commun. 2012, 3, 667. [Google Scholar] [CrossRef] [Green Version]
  33. Ishikawa, M.; Miyahara, R.; Sonobe, M.; Horiuchi, M.; Mennju, T.; Nakayama, E.; Kobayashi, M.; Kikuchi, R.; Kitamura, J.; Imamura, N.; et al. Higher expression of EphA2 and ephrin-A1 is related to favorable clinicopathological features in pathological stage I non-small cell lung carcinoma. Lung Cancer 2012, 76, 431–438. [Google Scholar] [CrossRef] [Green Version]
  34. Bulk, E.; Yu, J.; Hascher, A.; Koschmieder, S.; Wiewrodt, R.; Krug, U.; Timmermann, B.; Marra, A.; Hillejan, L.; Wiebe, K.; et al. Mutations of the EPHB6 receptor tyrosine kinase induce a pro-metastatic phenotype in non-small cell lung cancer. PLoS ONE 2012, 7, e44591. [Google Scholar] [CrossRef] [Green Version]
  35. Anderton, M.; van der Meulen, E.; Blumenthal, M.J.; Schafer, G. The Role of the Eph Receptor Family in Tumorigenesis. Cancers 2021, 13, 206. [Google Scholar] [CrossRef]
  36. Kou, C.J.; Kandpal, R.P. Differential Expression Patterns of Eph Receptors and Ephrin Ligands in Human Cancers. Biomed Res. Int. 2018, 2018, 7390104. [Google Scholar] [CrossRef] [Green Version]
  37. Pottier, C.; Fresnais, M.; Gilon, M.; Jerusalem, G.; Longuespee, R.; Sounni, N.E. Tyrosine Kinase Inhibitors in Cancer: Breakthrough and Challenges of Targeted Therapy. Cancers 2020, 12, 731. [Google Scholar] [CrossRef] [PubMed]
  38. Jeon, J.Y.; Sparreboom, A.; Baker, S.D. Kinase Inhibitors: The Reality Behind the Success. Clin. Pharmacol. Ther. 2017, 102, 726–730. [Google Scholar] [CrossRef] [PubMed]
  39. Noor, Z.S.; Cummings, A.L.; Johnson, M.M.; Spiegel, M.L.; Goldman, J.W. Targeted Therapy for Non-Small Cell Lung Cancer. Semin. Respir. Crit. Care Med. 2020, 41, 409–434. [Google Scholar] [CrossRef]
  40. Sun, B.K.; Tsao, H. Small RNAs in development and disease. J. Am. Acad. Dermatol. 2008, 59, 725–737, quiz 38–40. [Google Scholar] [CrossRef] [PubMed]
  41. Tian, Z.; Liang, G.; Cui, K.; Liang, Y.; Wang, Q.; Lv, S.; Cheng, X.; Zhang, L. Insight Into the Prospects for RNAi Therapy of Cancer. Front. Pharmacol. 2021, 12, 644718. [Google Scholar] [CrossRef] [PubMed]
  42. Khan, P.; Siddiqui, J.A.; Lakshmanan, I.; Ganti, A.K.; Salgia, R.; Jain, M.; Batra, S.K.; Nasser, M.W. RNA-based therapies: A cog in the wheel of lung cancer defense. Mol. Cancer 2021, 20, 54. [Google Scholar] [CrossRef]
  43. Zahavi, D.; Weiner, L. Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34. [Google Scholar] [CrossRef]
  44. Paszko, E.; Senge, M.O. Immunoliposomes. Curr. Med. Chem. 2012, 19, 5239–5277. [Google Scholar] [CrossRef]
  45. Drugs Approved for Lung Cancer; National Cancer Institute: Bethesda, MD, USA, 2022.
  46. Cho, H.; Jeon, S.I.; Ahn, C.H.; Shim, M.K.; Kim, K. Emerging Albumin-Binding Anticancer Drugs for Tumor-Targeted Drug Delivery: Current Understandings and Clinical Translation. Pharmaceutics 2022, 14, 728. [Google Scholar] [CrossRef]
  47. Kundranda, M.N.; Niu, J. Albumin-bound paclitaxel in solid tumors: Clinical development and future directions. Drug Des. Dev. Ther. 2015, 9, 3767–3777. [Google Scholar] [CrossRef] [Green Version]
  48. Wang, X.; Li, H.; Chen, G. Core-shell nanoparticles for cancer imaging and therapy. In Core-Shell Nanostructures for Drug Delivery and Theranostics; Focarete, M.L., Tampieri, A., Eds.; Woodhead Publishing: Cambridge, UK, 2018; pp. 143–175. [Google Scholar]
  49. Yu, H.; Wang, Y.; Wang, S.; Li, X.; Li, W.; Ding, D.; Gong, X.; Keidar, M.; Zhang, W. Paclitaxel-Loaded Core-Shell Magnetic Nanoparticles and Cold Atmospheric Plasma Inhibit Non-Small Cell Lung Cancer Growth. ACS Appl. Mater. Interfaces 2018, 10, 43462–43471. [Google Scholar] [CrossRef] [PubMed]
  50. Menon, J.U.; Kuriakose, A.; Iyer, R.; Hernandez, E.; Gandee, L.; Zhang, S.; Takahashi, M.; Zhang, Z.; Saha, D.; Nguyen, K.T. Dual-Drug Containing Core-Shell Nanoparticles for Lung Cancer Therapy. Sci. Rep. 2017, 7, 13249. [Google Scholar] [CrossRef] [Green Version]
  51. Vaisberg, A.J.; Milla, M.; Planas, M.C.; Cordova, J.L.; De Agusti, E.R.; Ferreyra, R.; Mustiga, M.C.; Carlin, L.; Hammond, G.B. Taspine is the cicatrizant principle in Sangre de Grado extracted from Croton lechleri. Planta Med. 1989, 55, 140–143. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, X.; Zhang, H.; Qi, G.; Gu, X.; Zhao, Y.; Zhang, J. TPD7 inhibits the non-small cell lung cancer HCC827 cell growth by regulating EGFR signalling pathway. J. Chemother. 2022, 34, 110–116. [Google Scholar] [CrossRef] [PubMed]
  53. Lu, W.; Dai, B.; Ma, W.; Zhang, Y. A novel taspine analog, HMQ1611, inhibits growth of non-small cell lung cancer by inhibiting angiogenesis. Oncol. Lett. 2012, 4, 1109–1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Montopoli, M.; Bertin, R.; Chen, Z.; Bolcato, J.; Caparrotta, L.; Froldi, G. Croton lechleri sap and isolated alkaloid taspine exhibit inhibition against human melanoma SK23 and colon cancer HT29 cell lines. J. Ethnopharmacol. 2012, 144, 747–753. [Google Scholar] [CrossRef]
  55. Zhan, Y.; Zhang, Y.; Liu, C.; Zhang, J.; Smith, W.W.; Wang, N.; Chen, Y.; Zheng, L.; He, L. A novel taspine derivative, HMQ1611, inhibits breast cancer cell growth via estrogen receptor alpha and EGF receptor signaling pathways. Cancer Prev. Res. 2012, 5, 864–873. [Google Scholar] [CrossRef] [Green Version]
  56. Dai, B.; Wang, W.; Ma, Y.; Liu, R.; Zhang, Y. A taspine derivative supresses Caco-2 cell growth by competitively targeting EphrinB2 and regulating its pathway. Oncol. Rep. 2016, 36, 1526–1534. [Google Scholar] [CrossRef] [Green Version]
  57. Bai, H.; Duan, J.; Li, C.; Xie, W.; Fang, W.; Xu, Y.; Wang, G.; Wan, R.; Sun, J.; Xu, J.; et al. EPHA mutation as a predictor of immunotherapeutic efficacy in lung adenocarcinoma. J. Immunother. Cancer 2020, 8, e001315. [Google Scholar] [CrossRef]
  58. Li, J.; Rix, U.; Fang, B.; Bai, Y.; Edwards, A.; Colinge, J.; Bennett, K.L.; Gao, J.; Song, L.; Eschrich, S.; et al. A chemical and phosphoproteomic characterization of dasatinib action in lung cancer. Nat. Chem. Biol. 2010, 6, 291–299. [Google Scholar] [CrossRef] [Green Version]
  59. Choi, K.; Creighton, C.J.; Stivers, D.; Fujimoto, N.; Kurie, J.M. Transcriptional profiling of non-small cell lung cancer cells with activating EGFR somatic mutations. PLoS ONE 2007, 2, e1226. [Google Scholar] [CrossRef] [PubMed]
  60. Koch, H.; Busto, M.E.; Kramer, K.; Medard, G.; Kuster, B. Chemical Proteomics Uncovers EPHA2 as a Mechanism of Acquired Resistance to Small Molecule EGFR Kinase Inhibition. J. Proteome Res. 2015, 14, 2617–2625. [Google Scholar] [CrossRef] [PubMed]
  61. Srivastava, S.; Pang, K.M.; Iida, M.; Nelson, M.S.; Liu, J.; Nam, A.; Wang, J.; Mambetsariev, I.; Pillai, R.; Mohanty, A.; et al. Activation of EPHA2-ROBO1 Heterodimer by SLIT2 Attenuates Non-canonical Signaling and Proliferation in Squamous Cell Carcinomas. iScience 2020, 23, 101692. [Google Scholar] [CrossRef] [PubMed]
  62. Amato, K.R.; Wang, S.; Tan, L.; Hastings, A.K.; Song, W.; Lovly, C.M.; Meador, C.B.; Ye, F.; Lu, P.; Balko, J.M.; et al. EPHA2 Blockade Overcomes Acquired Resistance to EGFR Kinase Inhibitors in Lung Cancer. Cancer Res. 2016, 76, 305–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ishigaki, H.; Minami, T.; Morimura, O.; Kitai, H.; Horio, D.; Koda, Y.; Fujimoto, E.; Negi, Y.; Nakajima, Y.; Niki, M.; et al. EphA2 inhibition suppresses proliferation of small-cell lung cancer cells through inducing cell cycle arrest. Biochem. Biophys. Res. Commun. 2019, 519, 846–853. [Google Scholar] [CrossRef] [PubMed]
  64. Kaminskyy, V.O.; Haag, P.; Novak, M.; Végvári, Á.; Arapi, V.; Lewensohn, R.; Viktorsson, K. EPHA2 Interacts with DNA-PKcs in Cell Nucleus and Controls Ionizing Radiation Responses in Non-Small Cell Lung Cancer Cells. Cancers 2021, 13, 1010. [Google Scholar] [CrossRef]
  65. Iyer, R.; Ramachandramoorthy, H.; Nguyen, T.; Xu, C.; Fu, H.; Kotadia, T.; Chen, B.; Hong, Y.; Saha, D.; Nguyen, K.T. Lung Cancer Targeted Chemoradiotherapy via Dual-Stimuli Responsive Biodegradable Core-Shell Nanoparticles. Pharmaceutics 2022, 14, 1525. [Google Scholar] [CrossRef]
  66. Lee, H.Y.; Mohammed, K.A.; Kaye, F.; Moudgil, B.M.; Nasreen, N. EphA2 targeted intratumoral therapy for non-small cell lung cancer using albumin mesospheres. Am. J. Transl. Res. 2017, 9, 3293–3303. [Google Scholar]
  67. Gan, H.K.; Parakh, S.; Lee, F.T.; Tebbutt, N.C.; Ameratunga, M.; Lee, S.T.; O’Keefe, G.J.; Gong, S.J.; Vanrenen, C.; Caine, J.; et al. A phase 1 safety and bioimaging trial of antibody DS-8895a against EphA2 in patients with advanced or metastatic EphA2 positive cancers. Investig. New Drugs 2022, 40, 747–755. [Google Scholar] [CrossRef]
  68. Peng, J.; Wang, Q.; Liu, H.; Ye, M.; Wu, X.; Guo, L. EPHA3 regulates the multidrug resistance of small cell lung cancer via the PI3K/BMX/STAT3 signaling pathway. Tumour Biol. 2016, 37, 11959–11971. [Google Scholar] [CrossRef] [Green Version]
  69. Sos, M.L.; Michel, K.; Zander, T.; Weiss, J.; Frommolt, P.; Peifer, M.; Li, D.; Ullrich, R.; Koker, M.; Fischer, F.; et al. Predicting drug susceptibility of non-small cell lung cancers based on genetic lesions. J. Clin. Investig. 2009, 119, 1727–1740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Staquicini, F.I.; Qian, M.D.; Salameh, A.; Dobroff, A.S.; Edwards, J.K.; Cimino, D.F.; Moeller, B.J.; Kelly, P.; Nunez, M.I.; Tang, X.; et al. Receptor tyrosine kinase EphA5 is a functional molecular target in human lung cancer. J. Biol. Chem. 2015, 290, 7345–7359. [Google Scholar] [CrossRef] [PubMed]
  71. Li, R.; Sun, Y.; Jiang, A.; Wu, Y.; Li, C.; Jin, M.; Yan, H.; Jin, H. Knockdown of ephrin receptor A7 suppresses the proliferation and metastasis of A549 human lung cancer cells. Mol. Med. Rep. 2016, 13, 3190–3196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ferguson, B.D.; Liu, R.; Rolle, C.E.; Tan, Y.H.C.; Krasnoperov, V.; Kanteti, R.; Tretiakova, M.S.; Cervantes, G.M.; Hasina, R.; Hseu, R.D.; et al. The EphB4 receptor tyrosine kinase promotes lung cancer growth: A potential novel therapeutic target. PLoS ONE 2013, 8, e67668. [Google Scholar] [CrossRef] [Green Version]
  73. Nanamiya, R.; Saito-Koyama, R.; Miki, Y.; Inoue, C.; Asavasupreechar, T.; Abe, J.; Sato, I.; Sasano, H. EphB4 as a Novel Target for the EGFR-Independent Suppressive Effects of Osimertinib on Cell Cycle Progression in Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2021, 22, 8522. [Google Scholar] [CrossRef]
  74. Yoon, S.; Choi, J.H.; Kim, S.J.; Lee, E.J.; Shah, M.; Choi, S.; Woo, H.G. EPHB6 mutation induces cell adhesion-mediated paclitaxel resistance via EPHA2 and CDH11 expression. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef] [Green Version]
  75. Lee, H.Y.; Mohammed, K.A.; Peruvemba, S.; Goldberg, E.P.; Nasreen, N. Targeted lung cancer therapy using ephrinA1-loaded albumin microspheres. J. Pharm. Pharmacol. 2011, 63, 1401–1410. [Google Scholar] [CrossRef]
  76. Lee, H.Y.; Mohammed, K.A.; Kaye, F.; Sharma, P.; Moudgil, B.M.; Clapp, W.L.; Nasreen, N. Targeted delivery of let-7a microRNA encapsulated ephrin-A1 conjugated liposomal nanoparticles inhibit tumor growth in lung cancer. Int. J. Nanomed. 2013, 8, 4481–4494. [Google Scholar]
  77. Murugan, D.; Rangasamy, L. A perspective to weaponize microRNAs against lung cancer. Noncoding RNA Res. 2022, 8, 18–32. [Google Scholar] [CrossRef]
  78. Huang, Z.R.; Tipparaju, S.K.; Kirpotin, D.B.; Pien, C.; Kornaga, T.; O Noble, C.; Koshkaryev, A.; Tran, J.; Kamoun, W.S.; Drummond, D.C. Formulation optimization of an ephrin A2 targeted immunoliposome encapsulating reversibly modified taxane prodrugs. J. Control. Release 2019, 310, 47–57. [Google Scholar] [CrossRef]
  79. Dai, B.; Ma, Y.; Yang, T.; Wang, W.; Zhang, Y. Taspine derivative 12k suppressed A549 cell migration through the Wnt/beta-catenin and EphrinB2 signaling pathway. Biomed. Pharmacother. 2017, 87, 102–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Stahl, S.; Branca, R.M.; Efazat, G.; Ruzzene, M.; Zhivotovsky, B.; Lewensohn, R.; Viktorsson, K.; Lehtiö, J. Phosphoproteomic profiling of NSCLC cells reveals that ephrin B3 regulates pro-survival signaling through Akt1-mediated phosphorylation of the EphA2 receptor. J. Proteome Res. 2011, 10, 2566–2578. [Google Scholar] [CrossRef] [PubMed]
  81. Lung Cancer Survival Rates; American Cancer Society: Atlanta, GA, USA, 2022.
Figure 1. The role of the EPH/ephrin system in LC. Created with BioRender.com.
Figure 1. The role of the EPH/ephrin system in LC. Created with BioRender.com.
Ijms 24 00093 g001
Figure 2. Treatment agents targeting the EPH/ephrin system in LC.
Figure 2. Treatment agents targeting the EPH/ephrin system in LC.
Ijms 24 00093 g002
Table 1. Different therapeutics targeting the EPH/ephrin system in LC.
Table 1. Different therapeutics targeting the EPH/ephrin system in LC.
Targeting StrategyMechanism of Action
Small-molecule drugsTyrosine kinase inhibitors
Regulatory small RNA moleculesShort interfering RNAs
Monoclonal antibodiesAnti-EPH/ephrin monoclonal antibodies, antibody–drug conjugations
ImmunoliposomesAntibodies coupled to the liposomal surface
NanoparticlesAlbumin microspheres, core–shell nanoparticles
Natural compoundsTaspine derivatives
Table 2. Effects of diverse therapeutic agents on the EPH/ephrin system in LC.
Table 2. Effects of diverse therapeutic agents on the EPH/ephrin system in LC.
Targeted EPH/EphrinTherapeutic AgentMechanism of
Action
References
EPHA2TKIs
  • EPHA2
Enrichment;
  • Reciprocal EPHA2
expression
regulation;
  • FAK
phosphorylation reduction;
  • AKT activity downregulation;
  • Cell migration inhibition;
  • Apoptosis
Induction;
  • Decreased
Proliferation;
  • Tumor growth repression.
[58,60,61,62]
EPHA2-Fc peptide
  • Anchorage-
independent NSCLC cell growth
inhibition.
[59]
siRNA
  • Rb dephosphorylation;
  • Cell-cycle
Arrest;
  • Partial DNA-PKcs
phosphorylation; reduction;
  • Apoptotic
signaling
amplification.
[63,64]
E-DSNPs
  • Triggered
release of the radiosensitizer during
concurrent
radiation
therapy;
  • Chemo-drug release upon glutathione
Exposure;
  • In vitro LC cell survival
fraction downregulation.
[65]
Albumin mesosphere-conjugated ephrin-A1
  • Apoptosis
Induction;
  • NSCLC in vivo tumor
Shrinkage.
[66]
DS-8895a
  • Low-grade
tumor uptake.
[67]
EPHA3Dasatinib
  • NSCLC in vivo tumor
Shrinkage.
[69]
EPHA511C12
  • Sensitization to
Radiotherapy;
  • Anti-proliferative/pro-
senescence
effect;
  • Prolonged OS.
[70]
EPHA7siRNA
  • Inhibition of NSCLC cell proliferation, migration, and invasion;
  • Enhancement of Bcl-2-
associated X protein, caspase-3, and PTEN
expression
levels;
  • Phosphorylated-AKT level downregulation.
[71]
EPHB4siRNA
  • Reduced SCLC cell viability in vitro;
  • Tumor
regression in vivo.
[72]
Osimertinib
  • EPHB4 downregulation.
[73]
EPHB6ALW-II-41-27
  • EPHB6 (Q926R)-
induced CDH11
expression downregulation;
  • Suppressed
focal adhesion formation.
[74]
Ephrin-A1Ephrin-A1-loaded albumin
microspheres
  • Reduced FAK expression;
  • NSCLC cell migration
Inhibition;
  • Tumor growth repression.
[75]
let-7a microRNA-encapsulated ephrin-A1-conjugated liposomal nanoparticles
  • Repressed Ras expression;
  • Reduced NSCLC
proliferation;
  • Cell migration inhibition;
  • Tumor growth arrest.
[76]
Ephrin-A2Ephrin-A2-targeted taxane
liposomal prodrug 2
  • In vivo tumor
regression.
[78]
Ephrin-B212k
  • VEGFR-2/-3 downregulation;
  • Cell migration inhibition.
[79]
Ephrin-B3siRNA
  • Decreased cell proliferation;
  • Morphological changes.
[80]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Psilopatis, I.; Karniadakis, I.; Danos, K.S.; Vrettou, K.; Michaelidou, K.; Mavridis, K.; Agelaki, S.; Theocharis, S. May EPH/Ephrin Targeting Revolutionize Lung Cancer Treatment? Int. J. Mol. Sci. 2023, 24, 93. https://doi.org/10.3390/ijms24010093

AMA Style

Psilopatis I, Karniadakis I, Danos KS, Vrettou K, Michaelidou K, Mavridis K, Agelaki S, Theocharis S. May EPH/Ephrin Targeting Revolutionize Lung Cancer Treatment? International Journal of Molecular Sciences. 2023; 24(1):93. https://doi.org/10.3390/ijms24010093

Chicago/Turabian Style

Psilopatis, Iason, Ioannis Karniadakis, Konstantinos Stylianos Danos, Kleio Vrettou, Kleita Michaelidou, Konstantinos Mavridis, Sofia Agelaki, and Stamatios Theocharis. 2023. "May EPH/Ephrin Targeting Revolutionize Lung Cancer Treatment?" International Journal of Molecular Sciences 24, no. 1: 93. https://doi.org/10.3390/ijms24010093

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

Article Metrics

Back to TopTop