Skip to main content
Log in

Development of a Recombinant Monospecific Anti-PLGF Bivalent Nanobody and Evaluation of it in Angiogenesis Modulation

  • Original Paper
  • Published:
Molecular Biotechnology Aims and scope Submit manuscript

Abstract

During the past two decades, tumor therapy based on monoclonal antibody has been found as a confident therapeutic approach in solid tumors and hematologic malignancies. Nanobodies are the smallest fragment of an antigen-binding domain in heavy chain-only antibody originated from the Camelidae family. Accordingly, they are being recently developed rapidly as diagnostic and therapeutic agents. In this regard, targeting of angiogenic factors like Placenta growth factor (PLGF) via nanobodies show a high effectiveness. In the current study, we developed a recombinant anti-PLGF bivalent nanobody based on the affinity enhancement mutant form of anti-PLGF nanobody to suppress the angiogenesis progression. Thereafter, the bivalent nanobody (bi-Nb) was cloned and then expressed into a bacterial system. Afterward, the purity was authorized using western blot assay and the affinity was assessed using ELISA. In this regard, proliferation, 3D capillary tube formation, and migration assays were employed as functional assays. The obtained data were analyzed using t-test and P < 0.05 was considered as statistically significant. The results indicate that the bivalent nanobody could inhibit proliferation, mobility, and formation of endothelial cell capillary-like structure. Moreover, the EC50 was estimated for endothelial cell’s proliferation and capillary tube’s formation to be about 100 ng/ml and 65 ng/ml, respectively. Migration of MCF-7 was inhibited as about 69%, rather than the control. Accumulation of data have shown that targeting of angiogenic factors like VEGF via monoclonal antibodies or nanobodies can be useful in the suppression of tumor progression. Also, the inhibition of PLGF with monoclonal antibody indicated that it is significant in angiogenesis suppression. However, due to intrinsic properties of nanobodies, they are suggested to be used. Since the small size is rapidly removed through liver or kidney system, so it is important to use bivalent or polymeric forms for extending the half-life. Our findings indicated that the inhibition of PLGF can prevent growth and proliferation of endothelial cells and tumor cells through the bivalent nanobody. So, it is suggested as a novel therapeutic agent for angiogenesis suppression.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

AMD:

Age-related macular degeneration

Ang:

Angiopoietin

bi-Nb:

Bivalent nanobody

bi-Nb-PLGF-pHEN6c:

Anti-PLGF bivalent nanobody-pHEN6c

CML:

Chronic myeloid leukemia

DMEM:

Dulbecco’s modified eagle medium

EGF:

Epidermal growth factor

ELISA:

Enzyme-linked immunosorbent assay

HGF:

Hepatocyte growth factor

HIF-1α:

Hypoxia-inducible factor-1 alpha

HRP-conjugated antibody:

Horse radish peroxidase-conjugated antibody

PBS:

Phosphate-buffered saline

PCR:

Polymerase chain reaction

PDGF:

Platelet-derived growth factor

PI3K:

Phosphatidylinositol-3 kinase

PLGF:

Placenta growth factor

PTM:

Post-translational modification

SDS-PAGE:

Sodium dodecyl sulfate–polyacrylamide gel electrophoresis

VEGF:

Vascular endothelial growth factor

VEGFR:

Vascular endothelial growth factor receptor

VHH:

Variable domain of the heavy chain of a heavy chain antibody

VWF:

Von Willebrand factor

MCF-7:

Michigan Cancer Foundation-7(a breast cancer cell line)

MTT:

3, 4, 5-Dimethylthiazol-2-yl-2–5-diphenyltetrazolium bromide

Ni–NTA:

Nickel ion-nitrilotriacetic acid

DMSO:

Dimethyl sulfoxide

FBS:

Fetal bovine serum

IgA:

Immunoglobulin A

IgG:

Immunoglobulin G

IPTG:

Isopropyl β-D-1 thiogalactopyranoside

TB medium:

Terrific broth medium

TMB:

3, 3′, 5, 5′-Tetramethyl benzidine

TNF-α:

Tumor necrosis factor alpha

HUVEC:

Human umbilical vein endothelial cell

DAB:

Diaminobenzidine

References

  1. Tetzlaff, F., & Fischer, A. (2018). Human endothelial cell spheroid-based sprouting angiogenesis assay in collagen. Bio-Protocol, 8(17), e2995.

    Article  CAS  Google Scholar 

  2. Nishida, N., et al. (2006). Angiogenesis in cancer. Vascular Health and Risk Management, 2(3), 213–219.

    Article  CAS  Google Scholar 

  3. De Falco, S. (2012). The discovery of placenta growth factor and its biological activity. Experimental & Molecular Medicine, 44, 1.

    Article  CAS  Google Scholar 

  4. Apicella, I., et al. (2018). Full functional knockout of placental growth factor by knockin with an inactive variant able to heterodimerize with VEGF-A. Cell Reports, 23(12), 3635–3646.

    Article  CAS  Google Scholar 

  5. Tarallo, V., et al. (2010). A placental growth factor variant unable to recognize vascular endothelial growth factor (VEGF) receptor-1 inhibits VEGF-dependent tumor angiogenesis via heterodimerization. Cancer Research, 70(5), 1804–1813.

    Article  CAS  Google Scholar 

  6. Lazzara, F., et al. (2019). Aflibercept regulates retinal inflammation elicited by high glucose via the PlGF/ERK pathway. Biochemical Pharmacology, 168, 341–351.

    Article  CAS  Google Scholar 

  7. Lazzara, F., et al. (2020). Stabilization of HIF-1α in human retinal endothelial cells modulates expression of miRNAs and proangiogenic growth factors. Frontiers in Pharmacology, 11(1063), e01063.

    Google Scholar 

  8. Albonici, L., et al. (2019). Multifaceted role of the placental growth factor (PlGF) in the antitumor immune response and cancer progression. International Journal of Molecular Sciences, 20(12), 2970.

    Article  CAS  Google Scholar 

  9. Pagani, E., et al. (2016). Placenta growth factor and neuropilin-1 collaborate in promoting melanoma aggressiveness. International Journal of Oncology, 48(4), 1581–1589.

    Article  CAS  Google Scholar 

  10. Martinsson-Niskanen, T., et al. (2011). Monoclonal antibody TB-403: A first-in-human, phase I, double-blind, dose escalation study directed against placental growth factor in healthy male subjects. Clinical Therapeutics, 33(9), 1142–1149.

    Article  CAS  Google Scholar 

  11. Dewerchin, M., & Carmeliet, P. (2012). PlGF: A multitasking cytokine with disease-restricted activity. Cold Spring Harbor Perspectives in Medicine, 2(8), a011056.

    Article  CAS  Google Scholar 

  12. Arbabi-Ghahroudi, M. (2017). Camelid single-domain antibodies: Historical perspective and future outlook. Frontiers in Immunology, 8, 01589.

    Article  CAS  Google Scholar 

  13. Leow, C. H., et al. (2018). The development of single domain antibodies for diagnostic and therapeutic applications. Antibody Engineering, 175.

  14. Bannas, P., Hambach, J., & Koch-Nolte, F. (2017). Nanobodies and nanobody-based human heavy chain antibodies as antitumor therapeutics. Frontiers in Immunology, 8, 01603.

    Article  CAS  Google Scholar 

  15. Van Audenhove, I., & Gettemans, J. (2016). Nanobodies as versatile tools to understand, diagnose, visualize and treat cancer. Ebiomedicine, 8, 40–48.

    Article  Google Scholar 

  16. Hu, Y. Z., Liu, C. X., & Muyldermans, S. (2017). Nanobody-based delivery systems for diagnosis and targeted tumor therapy. Frontiers in Immunology, 8, 01442.

    Article  CAS  Google Scholar 

  17. Kijanka, M., et al. (2015). Nanobody-based cancer therapy of solid tumors. Nanomedicine, 10(1), 161–174.

    Article  CAS  Google Scholar 

  18. Kontermann, R. E. (2009). Strategies to extend plasma half-lives of recombinant antibodies. Biodrugs, 23(2), 93–109.

    Article  CAS  Google Scholar 

  19. Klein, J. S., et al. (2014). Design and characterization of structured protein linkers with differing flexibilities. Protein Engineering Design & Selection, 27(10), 325–330.

    Article  CAS  Google Scholar 

  20. Chen, X. Y., Zaro, J. L., & Shen, W. C. (2013). Fusion protein linkers: Property, design and functionality. Advanced Drug Delivery Reviews, 65(10), 1357–1369.

    Article  CAS  Google Scholar 

  21. Movahedi, K., et al. (2012). Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Research, 72(16), 4165–4177.

    Article  CAS  Google Scholar 

  22. Scully, M., et al. (2019). Caplacizumab treatment for acquired thrombotic thrombocytopenic purpura. New England Journal of Medicine, 380(4), 335–346.

    Article  CAS  Google Scholar 

  23. Fleischmann, R., et al. (2011). A multiple ascending dose/proof of concept study of ATN-103 (ozoralizumab) in rheumatoid arthritis subjects on a background of methotrexate. Arthritis and Rheumatism, 63(10), S1033–S1034.

    Google Scholar 

  24. Ebrahimi, Z., et al. (2018). Rational affinity enhancement of fragmented antibody by ligand-based affinity improvement approach. Biochemical and Biophysical Research Communications, 506(3), 653–659.

    Article  CAS  Google Scholar 

  25. Arezumand, R., et al. (2016). Identification and characterization of a novel nanobody against human placental growth factor to modulate angiogenesis. Molecular Immunology, 78, 183–192.

    Article  CAS  Google Scholar 

  26. Beatty, J. D., Beatty, B. G., & Vlahos, W. G. (1987). Measurement of monoclonal antibody affinity by non-competitive enzyme immunoassay. Journal of Immunological Methods, 100(1–2), 173–179.

    Article  CAS  Google Scholar 

  27. Cabral, T., et al. (2017). Retinal and choroidal angiogenesis: A review of new targets. International Journal of Retina and Vitreous, 3(1), 31.

    Article  Google Scholar 

  28. Kong, D. H., et al. (2017). A review of anti-angiogenic targets for monoclonal antibody cancer therapy. International Journal of Molecular Science, 18(8), 1786.

    Article  CAS  Google Scholar 

  29. Ceci, C., et al. (2020). Role of VEGFs/VEGFR-1 signaling and its inhibition in modulating tumor invasion: Experimental evidence in different metastatic cancer models. International Journal of Molecular Science, 21(4), 1388.

    Article  CAS  Google Scholar 

  30. Giurdanella, G., et al. (2015). Aflibercept, bevacizumab and ranibizumab prevent glucose-induced damage in human retinal pericytes in vitro, through a PLA2/COX-2/VEGF-A pathway. Biochemical Pharmacology, 96(3), 278–287.

    Article  CAS  Google Scholar 

  31. Spadiut, O., et al. (2014). Microbials for the production of monoclonal antibodies and antibody fragments. Trends in Biotechnology, 32(1), 54–60.

    Article  CAS  Google Scholar 

  32. Sadeghi, A., et al. (2019). Development of a mono-specific anti-VEGF bivalent nanobody with extended plasma half-life for treatment of pathologic neovascularization. Drug Testing and Analysis, 12, 92.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This project was financially supported by North Khorasan University of Medical Sciences .and National Institute for medical research development (Nimad).

Funding

This project was financially supported by North Khorasan University of Medical Sciences, Bojnurd, Iran.

Author information

Authors and Affiliations

Authors

Contributions

RA: management the research group and analysis the result and writing the manuscript. AN: perform the assays and initial analysis and writing the manuscript. KM: help to performing the angiogenesis assays. AHK: writing the draft of manuscript. HNAA: writing the draft of manuscript.

Corresponding author

Correspondence to Roghaye Arezumand.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nikooharf, A., Arezumand, R., Mansouri, K. et al. Development of a Recombinant Monospecific Anti-PLGF Bivalent Nanobody and Evaluation of it in Angiogenesis Modulation. Mol Biotechnol 62, 580–588 (2020). https://doi.org/10.1007/s12033-020-00275-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12033-020-00275-7

Keywords

Navigation