Next Article in Journal
Anti-Oxidative and Anti-Apoptotic Oligosaccharides from Pichia pastoris-Fermented Cress Polysaccharides Ameliorate Chromium-Induced Liver Toxicity
Previous Article in Journal
Novel Coumarin–Nucleobase Hybrids with Potential Anticancer Activity: Synthesis, In Vitro Cell-Based Evaluation, and Molecular Docking
Previous Article in Special Issue
Chirality and Rigidity in Triazole-Modified Peptidomimetics Interacting with Neuropilin-1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Vascular Endothelial Growth Factor (VEGF) and VEGF Receptor Inhibitors in Health and Disease

CiTCoM, UMR CNRS 8038, U1268 INSERM, UFR de Pharmacie, Faculté de Santé, Université Paris Cité, 75006 Paris, France
Pharmaceuticals 2024, 17(7), 959; https://doi.org/10.3390/ph17070959
Submission received: 11 July 2024 / Accepted: 16 July 2024 / Published: 18 July 2024
In this Special Issue of Pharmaceuticals, we present four reviews and seven original articles addressing recent aspects of research on Vascular Endothelial Growth Factors (VEGFs) and their receptors, from clinical practice to fundamental studies in new drug development. The VEGF family of cytokines constitutes the main pro-angiogenic and lymphangiogenic factors. In mammals, this family includes VEGF-A (or simply VEGF), VEGF-B, -C, -D, and placental growth factor (PlGF). Their intracellular effects are mediated by binding with three receptors, VEGFR-1/2/3, and two co-receptors, the neuropilins NRP-1/2. Over the past 20 years, pharmaceuticals targeting the VEGF/VEGFR axis (primarily VEGF-A/VEGFR-2) and its downstream signaling pathways have been successfully used to treat pathological angiogenesis, which plays a role in several cancers and ocular diseases. However, these treatments have their limitations, particularly with regard to their long-term efficiency and their route of administration [1,2].
In the VEGF/VEGFR axis, it is important to distinguish two biological targets and, therefore, two completely different types of pharmaceutical inhibitors. The first biological targets are extracellular: VEGF/VEGFR and VEGF/NRP interactions, which are protein/protein interaction interfaces, typically inhibited by large molecules. The second targets are the intracellular tyrosine kinase domains of VEGFRs and other downstream intracellular enzymes. These enzyme active sites are generally inhibited by small molecules.
In clinical practice, extracellular VEGF ligands are classed as macromolecules, such as receptor fragments and antibodies. The anti-VEGF antibody bevacizumab was approved to treat colorectal cancer in 2004 and has since been widely used to treat other types of cancer. It is prescribed to treat non-small cell lung cancer in combination with paclitaxel–carboplatin chemotherapy [3]. The development of angiogenesis inhibitors in this pathology is the subject of intense research [4]. It was found that recombinant endostatin interferes with the VEGF/VEGFR axis and creates a “vascular normalization” window. In light of this, combining endostatin and radiotherapy appears to be a promising approach in non-small cell lung cancer (reviewed in Contribution 1). For ocular diseases, recent clinical studies continue to accurately evaluate the effects of intravitreal bevacizumab and ranibizumab, particularly for neovascular Age-Related Macular Degeneration (nAMD) (reviewed in Contribution 2). These studies confirm that these treatments have beneficial effects for the vast majority of patients (Contribution 3) and patient-reported outcomes indicated a significant improvement in quality of life (Contribution 4). However, administration of these macromolecules requires repeated intravitreal injections, leading to patient discomfort and, potentially, non-compliance with treatment. Interestingly, recent research on inhibitors of extracellular VEGF/VEGFR and VEGF/NRP interactions has expanded to smaller molecular entities with specifically designed properties, such as monospecific [5,6] and bispecific nanobodies [7,8], peptides, and peptidomimetics (reviewed in Contribution 5). These new potential therapeutic molecules include peptide ligands of VEGF [9,10] or VEGFRs (Contribution 6 and reference [11]) and peptidomimetics targeting NRP-1 (Contribution 7 and references [12,13]). Moreover, aptamers have proven to be very useful for the detection of small concentrations of VEGF for diagnostic purposes, in addition to therapeutic applications [14]. The application of aptamers for early detection and treatment of ovarian cancer is reviewed in Contribution 8.
Small molecule inhibitors of the intracellular tyrosine kinase domains of the VEGFRs have been successfully developed for cancer treatment but have not yet reached the clinic for nAMD. These small molecules are generally less specific than macromolecules, and exhibit more side effects in systemic circulation [2,15]. For example, the triple VEGFR inhibitor tivozanib, which was recently approved to treat refractory advanced renal cell carcinoma, has side effects such as hypertension [16]. The underlying mechanism was studied in mice models in Contribution 9. On the other hand, the lower specificity of these small molecules allows multi-targeting, as demonstrated by chiauranib, an inhibitor of the VEGFR/Aurora B/CSF-1R kinases [17], and promising preclinical studies on transformed follicular lymphoma are presented in Contribution 10.
The VEGF/VEGFR axis also plays a role in several pathologies other than cancers and retinopathies, such as diabetic nephropathy [18]. The underlying mechanism and potential therapeutic applications were investigated in rat models of diabetes treated with suramin in Contribution 11.
We thank all authors and reviewers for their valuable contributions to this Special Issue, and we believe that this will trigger further studies to better understand the action of Pharmaceuticals targeting the key VEGF/VEGFR axis and develop a next generation of drugs, for improved patient outcomes.

Conflicts of Interest

The author declares no conflicts of interest.

List of Contributions

  • Cunningham, C.; Bolcaen, J.; Bisio, A.; Genis, A.; Strijdom, H.; Vandevoorde, C. Recombinant Endostatin as a Potential Radiosensitizer in the Treatment of Non-Small Cell Lung Cancer. Pharmaceuticals 2023, 16, 219. https://doi.org/10.3390/ph16020219.
  • Hang, A.; Feldman, S.; Amin, A.P.; Ochoa, J.A.R.; Park, S.S. Intravitreal Anti-Vascular Endothelial Growth Factor Therapies for Retinal Disorders. Pharmaceuticals 2023, 16, 1140. https://doi.org/10.3390/ph16081140.
  • Korva-Gurung, I.; Kubin, A.M.; Ohtonen, P.; Hautala, N. Visual Outcomes of Anti-VEGF Treatment on Neovascular Age-Related Macular Degeneration: A Real-World Population-Based Cohort Study. Pharmaceuticals 2023, 16, 927. https://doi.org/10.3390/ph16070927.
  • Almuiña-Varela, P.; García-Quintanilla, L.; Rodríguez-Cid, M.J.; Gil-Martínez, M.; Abraldes, M.J.; Gómez-Ulla, F.; Estany-Gestal, A.; Alcántara-Espinosa, J.M.; Fernández-Rodríguez, M.; Fernández-Ferreiro, A. Relationships between Patient-Reported Outcome Measures and Clinical Measures in Naïve Neovascular Age-Related Macular Degeneration Patients Treated with Intravitreal Ranibizumab. Pharmaceuticals 2024, 17, 157. https://doi.org/10.3390/ph17020157.
  • Di Stasi, R.; De Rosa, L.; D’Andrea, L.D. Structure-Based Design of Peptides Targeting VEGF/VEGFRs. Pharmaceuticals 2023, 16, 851. https://doi.org/10.3390/ph16060851.
  • Namjoo, M.; Ghafouri, H.; Assareh, E.; Aref, A.R.; Mostafavi, E.; Hamrahi Mohsen, A.; Balalaie, S.; Broussy, S.; Asghari, S.M. A VEGFB-Based Peptidomimetic Inhibits VEGFR2-Mediated PI3K/Akt/mTOR and PLCγ/ERK Signaling and Elicits Apoptotic, Antiangiogenic, and Antitumor Activities. Pharmaceuticals 2023, 16, 906. https://doi.org/10.3390/ph16060906.
  • Fedorczyk, B.; Redkiewicz, P.; Matalińska, J.; Piast, R.; Kosson, P.; Wieczorek, R. Chirality and Rigidity in Triazole-Modified Peptidomimetics Interacting with Neuropilin-1. Pharmaceuticals 2024, 17, 190. https://doi.org/10.3390/ph17020190.
  • Mishra, Y.; Chattaraj, A.; Mishra, V.; Ranjan, A.; Tambuwala, M.M. Aptamers Versus Vascular Endothelial Growth Factor (VEGF): A New Battle against Ovarian Cancer. Pharmaceuticals 2023, 16, 849. https://doi.org/10.3390/ph16060849.
  • Alanazi, W.A.; Alanazi, A.S.; El-Nagar, D.M.; Aljuraybah, A.M.; Alsanea, S.; Alharbi, M. Mechanism Underlying Triple VEGFR Inhibitor Tivozanib-Induced Hypertension in Mice Model. Pharmaceuticals 2023, 16, 295. https://doi.org/10.3390/ph16020295.
  • Tang, Y.; Zhong, M.; Pan, G.; Tan, J.; Xie, C.; Jiang, Y.; Yao, J.; Shan, W.; Lin, J.; Huang, J.; et al. Preclinical Studies of Chiauranib Show It Inhibits Transformed Follicular Lymphoma through the VEGFR2/ERK/STAT3 Signaling Pathway. Pharmaceuticals 2023, 16, 15. https://doi.org/10.3390/ph16010015.
  • Chyła-Danił, G.; Sałaga-Zaleska, K.; Kreft, E.; Krzesińska, A.; Herman, S.; Kuchta, A.; Sakowicz-Burkiewicz, M.; Lenartowicz, M.; Jankowski, M. Suramin Affects the Renal VEGF-A/VEGFR Axis in Short-Term Streptozotocin-Induced Diabetes. Pharmaceuticals 2023, 16, 470. https://doi.org/10.3390/ph16030470.

References

  1. Cao, Y.; Langer, R.; Ferrara, N. Targeting angiogenesis in oncology, ophthalmology and beyond. Nat. Rev. Drug Discov. 2023, 22, 476–495. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, L.; Liu, W.Q.; Broussy, S.; Han, B.; Fang, H. Recent advances of anti-angiogenic inhibitors targeting VEGF/VEGFR axis. Front. Pharmacol. 2023, 14, 1307860. [Google Scholar] [CrossRef] [PubMed]
  3. Sandler, A.; Gray, R.; Perry, M.C.; Brahmer, J.; Schiller, J.H.; Dowlati, A.; Lilenbaum, R.; Johnson, D.H. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. New Engl. J. Med. 2006, 355, 2542–2550. [Google Scholar] [CrossRef] [PubMed]
  4. Manzo, A.; Montanino, A.; Carillio, G.; Costanzo, R.; Sandomenico, C.; Normanno, N.; Piccirillo, M.C.; Daniele, G.; Perrone, F.; Rocco, G.; et al. Angiogenesis inhibitors in NSCLC. Int. J. Mol. Sci. 2017, 18, 2021. [Google Scholar] [CrossRef] [PubMed]
  5. Karami, E.; Sabatier, J.M.; Behdani, M.; Irani, S.; Kazemi-Lomedasht, F. A nanobody-derived mimotope against VEGF inhibits cancer angiogenesis. J. Enzym. Inhib. Med. Chem. 2020, 35, 1233–1239. [Google Scholar] [CrossRef] [PubMed]
  6. Karami, E.; Naderi, S.; Roshan, R.; Behdani, M.; Kazemi-Lomedasht, F. Targeted therapy of angiogenesis using anti-VEGFR2 and anti-NRP-1 nanobodies. Cancer Chemother. Pharmacol. 2022, 89, 165–172. [Google Scholar] [CrossRef] [PubMed]
  7. Le Tourneau, C.; Becker, H.; Claus, R.; Elez, E.; Ricci, F.; Fritsch, R.; Silber, Y.; Hennequin, A.; Tabernero, J.; Jayadeva, G.; et al. Two phase I studies of BI 836880, a vascular endothelial growth factor/angiopoietin-2 inhibitor, administered once every 3 weeks or once weekly in patients with advanced solid tumors. ESMO Open 2022, 7, 100576. [Google Scholar] [CrossRef] [PubMed]
  8. Hofmann, I.; Baum, A.; Hofmann, M.H.; Trapani, F.; Reichel-Voda, C.; Ehrensperger, D.; Aichinger, M.; Ebner, F.; Budano, N.; Schweifer, N.; et al. Pharmacodynamic and antitumor activity of BI 836880, a dual Vascular Endothelial Growth Factor and Angiopoietin 2 inhibitor, alone and combined with Programmed Cell Death Protein-1 inhibition. J. Pharmacol. Exp. Ther. 2023, 384, 331–342. [Google Scholar] [CrossRef] [PubMed]
  9. Guryanov, I.; Korzhikov-Vlakh, V.; Bhattacharya, M.; Biondi, B.; Masiero, G.; Formaggio, F.; Tennikova, T.; Urtti, A. Conformationally constrained peptides with high affinity to the vascular endothelial growth factor. J. Med. Chem. 2021, 64, 10900–10907. [Google Scholar] [CrossRef] [PubMed]
  10. Ye, X.; Gaucher, J.F.; Hu, H.; Wang, L.; Broussy, S. Dimer peptide ligands of Vascular Endothelial Growth Factor: Optimizing linker length for high affinity and antiangiogenic activity. J. Med. Chem. 2023, 66, 9753–9765. [Google Scholar] [CrossRef] [PubMed]
  11. De Rosa, L.; Capasso, D.; Diana, D.; Stefania, R.; Di Stasi, R.; Fattorusso, R.; D’Andrea, L.D. Metabolic and conformational stabilization of a VEGF-mimetic beta-hairpin peptide by click-chemistry. Eur. J. Med. Chem. 2021, 222, 113575. [Google Scholar] [CrossRef] [PubMed]
  12. Puszko, A.K.; Sosnowski, P.; Rignault-Bricard, R.; Hermine, O.; Hopfgartner, G.; Pulka-Ziach, K.; Lepelletier, Y.; Misicka, A. Urea-peptide hybrids as VEGF-A(165)/NRP-1 complex inhibitors with improved receptor affinity and biological properties. Int. J. Mol. Sci. 2021, 22, 72. [Google Scholar] [CrossRef]
  13. Puszko, A.K.; Sosnowski, P.; Hermine, O.; Hopfgartner, G.; Lepelletier, Y.; Misicka, A. Structure-activity relationship studies and biological properties evaluation of peptidic NRP-1 ligands: Investigation of N-terminal cysteine importance. Bioorg. Med. Chem. 2023, 94, 117482. [Google Scholar] [CrossRef] [PubMed]
  14. Riccardi, C.; Napolitano, E.; Platella, C.; Musumeci, D.; Melone, M.A.B.; Montesarchio, D. Anti-VEGF DNA-based aptamers in cancer therapeutics and diagnostics. Med. Res. Rev. 2021, 41, 464–506. [Google Scholar] [CrossRef] [PubMed]
  15. Poor, S.H.; Weissgerber, G.; Adams, C.M.; Bhatt, H.; Browning, D.J.; Chastain, J.; Ciulla, T.A.; Ferriere, M.; Gedif, K.; Glazer, L.C.; et al. A randomized, double-masked, multicenter trial of topical acrizanib (LHA510), a tyrosine kinase VEGF-receptor inhibitor, in treatment-experienced subjects with neovascular age-related macular degeneration. Am. J. Ophthalmol. 2022, 239, 180–189. [Google Scholar] [CrossRef] [PubMed]
  16. Salgia, N.J.; Zengin, Z.B.; Pal, S.K. Tivozanib in renal cell carcinoma: A new approach to previously treated disease. Ther. Adv. Med. Oncol. 2020, 12, 1758835920923818. [Google Scholar] [CrossRef] [PubMed]
  17. Zhou, Y.; Shan, S.; Li, Z.B.; Xin, L.J.; Pan, D.S.; Yang, Q.J.; Liu, Y.P.; Yue, X.P.; Liu, X.R.; Gao, J.Z.; et al. CS2164, a novel multi-target inhibitor against tumor angiogenesis, mitosis and chronic inflammation with anti-tumor potency. Cancer Sci. 2017, 108, 469–477. [Google Scholar] [CrossRef] [PubMed]
  18. Majumder, S.; Advani, A. VEGF and the diabetic kidney: More than too much of a good thing. J. Diabetes Complicat. 2017, 31, 273–279. [Google Scholar] [CrossRef] [PubMed]
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

Broussy, S. Vascular Endothelial Growth Factor (VEGF) and VEGF Receptor Inhibitors in Health and Disease. Pharmaceuticals 2024, 17, 959. https://doi.org/10.3390/ph17070959

AMA Style

Broussy S. Vascular Endothelial Growth Factor (VEGF) and VEGF Receptor Inhibitors in Health and Disease. Pharmaceuticals. 2024; 17(7):959. https://doi.org/10.3390/ph17070959

Chicago/Turabian Style

Broussy, Sylvain. 2024. "Vascular Endothelial Growth Factor (VEGF) and VEGF Receptor Inhibitors in Health and Disease" Pharmaceuticals 17, no. 7: 959. https://doi.org/10.3390/ph17070959

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