Skip to main content

Advertisement

Log in

Structure-Guided Molecular Engineering of a Vascular Endothelial Growth Factor Antagonist to Treat Retinal Diseases

  • 2020 CMBE Young Innovators issue
  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Background

Ocular neovascularization is a hallmark of retinal diseases including neovascular age-related macular degeneration and diabetic retinopathy, two leading causes of blindness in adults. Neovascularization is driven by the interaction of soluble vascular endothelial growth factor (VEGF) ligands with transmembrane VEGF receptors (VEGFR), and inhibition of the VEGF pathway has shown tremendous clinical promise. However, anti-VEGF therapies require invasive intravitreal injections at frequent intervals and high doses, and many patients show incomplete responses to current drugs due to the lack of sustained VEGF signaling suppression.

Methods

We synthesized insights from structural biology with molecular engineering technologies to engineer an anti-VEGF antagonist protein. Starting from the clinically approved decoy receptor protein aflibercept, we strategically designed a yeast-displayed mutagenic library of variants and isolated clones with superior VEGF affinity compared to the clinical drug. Our lead engineered protein was expressed in the choroidal space of rat eyes via nonviral gene delivery.

Results

Using a structure-informed directed evolution approach, we identified multiple promising anti-VEGF antagonist proteins with improved target affinity. Improvements were primarily mediated through reduction in dissociation rate, and structurally significant convergent sequence mutations were identified. Nonviral gene transfer of our engineered antagonist protein demonstrated robust and durable expression in the choroid of treated rats one month post-injection.

Conclusions

We engineered a novel anti-VEGF protein as a new weapon against retinal diseases and demonstrated safe and noninvasive ocular delivery in rats. Furthermore, our structure-guided design approach presents a general strategy for discovery of targeted protein drugs for a vast array of applications.

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.

Institutional subscriptions

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7

Similar content being viewed by others

Abbreviations

NVAMD:

Neovascular age-related macular degeneration

DR:

Diabetic retinopathy

VEGF:

Vascular endothelial growth factor

PDGF:

Platelet derived growth factor

VEGFR:

Vascular endothelial growth factor receptor

RTK:

Receptor tyrosine kinase

IgG1:

Immunoglobulin G1

AAV:

Adeno-associated virus

LV:

Lentivirus

HEK:

Human embryonic kidney

FPLC:

Fast protein liquid chromatography

HBS:

HEPES-buffered saline

BAP:

Biotin-acceptor protein

PDBePISA:

Protein data bank in Europe proteins, interfaces, structures, and assemblies

MACS:

Magnetic-activated cell sorting

SA:

Streptavidin

BSA:

Bovine serum albumin

EDTA:

Ethylenediaminetetraacetic acid

FACS:

Fluorescence-activated cell sorting

PBAE:

Poly(beta-amino ester)

THF:

Tetrahydrofuran

DMSO:

Dimethyl sulfoxide

PDI:

Polydispersity index

PTFE:

Polytetrafluoroethylene

GPC:

Gel permeation chromatography

DLS:

Dynamic light scattering

NTA:

Nanoparticle tracking analysis

PBS:

Phosphate-buffered saline

TEM:

Transmission electron microscopy

BLI:

Bio-layer interferometry

scFv:

Single-chain variable fragment

References

  1. Avery, R. L., et al. Systemic pharmacokinetics and pharmacodynamics of intravitreal aflibercept, bevacizumab, and ranibizumab. Retina 37:1847–1858, 2017.

    Google Scholar 

  2. Boder, E. T., and K. D. Wittrup. Yeast surface display for screening combinatorial polypeptide libraries. Nat. Biotechnol. 15:553–557, 1997.

    Google Scholar 

  3. Boyer, D. S., J. S. Heier, D. M. Brown, S. F. Francom, T. Ianchulev, and R. G. Rubio. A phase IIIb study to evaluate the safety of ranibizumab in subjects with neovascular age-related macular degeneration. Ophthalmology 116:1731–1739, 2009.

    Google Scholar 

  4. Brown, D. M., et al. Ranibizumab vs. verteporfin for neovascular age-related macular degeneration. N. Engl. J. Med. 355:1432–1444, 2006.

    Google Scholar 

  5. Brozzo, M. S., et al. Thermodynamic and structural description of allosterically regulated VEGFR-2 dimerization. Blood 119:1781–1788, 2012.

    Google Scholar 

  6. Campochiaro, P. A. Ocular neovascularization. J. Mol. Med. 91:311–321, 2013.

    Google Scholar 

  7. Campochiaro, P. A., et al. Lentiviral vector gene transfer of endostatin/angiostatin for macular degeneration (GEM) study. Hum. Gene Ther. 28:99–111, 2017.

    Google Scholar 

  8. CATT Research Group. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. N. Engl. J. Med. 364:1897–1908, 2011.

    Google Scholar 

  9. Chao, G., W. L. Lau, B. J. Hackel, S. L. Sazinsky, S. M. Lippow, and K. D. Wittrup. Isolating and engineering human antibodies using yeast surface display. Nat. Protoc. 1:755–768, 2006.

    Google Scholar 

  10. Daien, V., et al. Incidence and outcomes of infectious and noninfectious endophthalmitis after intravitreal injections for age-related macular degeneration. Ophthalmology 125:66–74, 2018.

    Google Scholar 

  11. Diabetic Retinopathy Clinical Research Network. Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema. N. Engl. J. Med. 372:1193–1203, 2015.

    Google Scholar 

  12. Ding, K., et al. AAV8-vectored suprachoroidal gene transfer produces widespread ocular transgene expression. J. Clin. Invest. 129:4901–4911, 2019.

    Google Scholar 

  13. Dixon, J. A., S. C. N. Oliver, J. L. Olson, and N. Mandava. VEGF trap-eye for the treatment of neovascular age-related macular degeneration. Expert Opin. Investig. Drugs. 18:1573–1580, 2009.

    Google Scholar 

  14. Falavarjani, K. G., and Q. D. Nguyen. Adverse events and complications associated with intravitreal injection of anti-VEGF agents: a review of literature. Eye 27:787–794, 2013.

    Google Scholar 

  15. Heier, J. S., et al. Intravitreous injection of AAV2-sFLT01 in patients with advanced neovascular age-related macular degeneration: a phase 1, open-label trial. Lancet 390:50–61, 2017.

    Google Scholar 

  16. Ho, C. C. M., et al. “Velcro” engineering of high affinity CD47 ectodomain as signal regulatory protein α (SIRPα) antagonists that enhance antibody-dependent cellular phagocytosis. J. Biol. Chem. 290:12650–12663, 2015.

    Google Scholar 

  17. Holz, F. G., et al. Multi-country real-life experience of anti-vascular endothelial growth factor therapy for wet age-related macular degeneration. Br. J. Ophthalmol. 99:220–226, 2015.

    Google Scholar 

  18. Holz, F. G., S. Schmitz-Valckenberg, and M. Fleckenstein. Recent developments in the treatment of age-related macular degeneration. J. Clin. Invest. 124:1430–1438, 2014.

    Google Scholar 

  19. Jager, R. D., W. F. Mieler, and J. W. Miller. Age-related macular degeneration. N. Engl. J. Med 358:2606–2617, 2008.

    Google Scholar 

  20. Kieran, M. W., R. Kalluri, and Y.-J. Cho. The VEGF pathway in cancer and disease: responses, resistance, and the path forward. Cold Spring Harb. Perspect. Med. 2:1–17, 2012.

    Google Scholar 

  21. Klein, R., et al. Prevalence of age-related macular degeneration in 4 racial/ethnic groups in the multi-ethnic study of atherosclerosis. Ophthalmology 113:373–380, 2006.

    Google Scholar 

  22. Klein, R., B. E. Klein, S. E. Moss, M. D. Davis, and D. L. DeMets. The Wisconsin epidemiologic study of diabetic retinopathy. II. Prevalence and risk of diabetic retinopathy when age at diagnosis is less than 30 years. Arch. Ophthalmol. 102:520–526, 1984.

    Google Scholar 

  23. Klein, R., B. E. K. Klein, S. C. Tomany, S. M. Meuer, and G.-H. Huang. Ten-year incidence and progression of age-related maculopathy: the Beaver Dam eye study. Ophthalmology 109:1767–1779, 2002.

    Google Scholar 

  24. Kotterman, M. A., L. Yin, J. M. Strazzeri, J. G. Flannery, W. H. Merigan, and D. V. Schaffer. Antibody neutralization poses a barrier to intravitreal adeno-associated viral vector gene delivery to non-human primates. Gene Ther. 22:116–126, 2015.

    Google Scholar 

  25. Krissinel, E., and K. Henrick. Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 372:774–797, 2007.

    Google Scholar 

  26. Krohl, P. J., S. D. Ludwig, and J. B. Spangler. Emerging technologies in protein interface engineering for biomedical applications. Curr. Opin. Biotech. 60:82–88, 2019.

    Google Scholar 

  27. Kwak, N., N. Okamoto, J. M. Wood, and P. A. Campochiaro. VEGF is major stimulator in model of choroidal neovascularization. Invest. Ophthalmol. Vis. Sci. 41:3158–3164, 2000.

    Google Scholar 

  28. MacDonald, D. A., et al. Aflibercept exhibits VEGF binding stoichiometry distinct from bevacizumab and does not support formation of immune-like complexes. Angiogenesis 19:389–406, 2016.

    Google Scholar 

  29. Markovic-Mueller, S., et al. Structure of the full-length VEGFR-1 extracellular domain in complex with VEGF-A. Structure 25:341–352, 2017.

    Google Scholar 

  30. Pandey Arvind, K., et al. Mechanisms of VEGF (Vascular Endothelial Growth Factor) inhibitor-associated hypertension and vascular disease. Hypertension 71:e1–e8, 2018.

    Google Scholar 

  31. Papadopoulos, N., et al. Binding and neutralization of vascular endothelial growth factor (VEGF) and related ligands by VEGF Trap, ranibizumab and bevacizumab. Angiogenesis 15:171–185, 2012.

    Google Scholar 

  32. Patel, S. R., D. E. Berezovsky, B. E. McCarey, V. Zarnitsyn, H. F. Edelhauser, and M. R. Prausnitz. Targeted administration into the suprachoroidal space using a microneedle for drug delivery to the posterior segment of the eye. Invest. Ophthalmol. Vis. Sci. 53:4433–4441, 2012.

    Google Scholar 

  33. Patel, S. R., A. S. P. Lin, H. F. Edelhauser, and M. R. Prausnitz. Suprachoroidal drug delivery to the back of the eye using hollow microneedles. Pharm. Res. 28:166–176, 2011.

    Google Scholar 

  34. Pennington, K. L., and M. M. DeAngelis. Epidemiology of age-related macular degeneration (AMD): associations with cardiovascular disease phenotypes and lipid factors. Eye Vis. (Lond) 3:1–20, 2016.

    Google Scholar 

  35. Rakoczy, E. P., et al. Gene therapy with recombinant adeno-associated vectors for neovascular age-related macular degeneration: 1 year follow-up of a phase 1 randomised clinical trial. Lancet 386:2395–2403, 2015.

    Google Scholar 

  36. Raman, S., et al. Structure-guided design fine-tunes pharmacokinetics, tolerability, and antitumor profile of multispecific frizzled antibodies. Proc. Natl. Acad. Sci. USA 116:6812–6817, 2019.

    Google Scholar 

  37. Reichel, F. F., et al. AAV8 can induce innate and adaptive immune response in the primate eye. Mol. Ther. 25:2648–2660, 2017.

    Google Scholar 

  38. Rosenfeld, P. J., et al. Ranibizumab for neovascular age-related macular degeneration. N. Engl. J. Med. 355:1419–1431, 2006.

    Google Scholar 

  39. Saaddine, J. B., K. M. V. Narayan, and F. Vinicor. Vision loss: a public health problem? Ophthalmology 110:253–254, 2003.

    Google Scholar 

  40. Schlothauer, T., et al. Novel human IgG1 and IgG4 Fc-engineered antibodies with completely abolished immune effector functions. Protein Eng. Des. Sel. 29:457–466, 2016.

    Google Scholar 

  41. Shen, J., et al. Suprachoroidal gene transfer with nonviral nanoparticles. Sci. Adv. 6:1–10, 2020.

    Google Scholar 

  42. Shmueli, R. B., J. C. Sunshine, Z. Xu, E. J. Duh, and J. J. Green. Gene delivery nanoparticles specific for human microvasculature and macrovasculature. Nanomedicine 8:1200–1207, 2012.

    Google Scholar 

  43. Silva, D.-A., et al. De novo design of potent and selective mimics of IL-2 and IL-15. Nature 565:186–191, 2019.

    Google Scholar 

  44. Singer, M. A., et al. HORIZON: an open-label extension trial of ranibizumab for choroidal neovascularization secondary to age-related macular degeneration. Ophthalmology 119:1175–1183, 2012.

    Google Scholar 

  45. Sunshine, J. C., S. B. Sunshine, I. Bhutto, J. T. Handa, and J. J. Green. Poly(β-Amino Ester)-nanoparticle mediated transfection of retinal pigment epithelial cells in vitro and in vivo. PLoS ONE 7:e37543, 2012.

    Google Scholar 

  46. Tzeng, S. Y., L. J. Higgins, M. G. Pomper, and J. J. Green. Biomaterial-mediated cancer-specific DNA delivery to liver cell cultures using synthetic poly(beta-amino ester)s. J. Biomed. Mater. Res. 101A:1837–1845, 2013.

    Google Scholar 

  47. Vitt, U. A., S. Y. Hsu, and A. J. W. Hsueh. Evolution and classification of cystine knot-containing hormones and related extracellular signaling molecules. Mol. Endocrinol. 15:681–694, 2001.

    Google Scholar 

  48. Weiskopf, K., et al. Engineered SIRPα variants as immunotherapeutic adjuvants to anticancer antibodies. Science 341:88–91, 2013.

    Google Scholar 

  49. Wrenbeck, E. E., M. S. Faber, and T. A. Whitehead. Deep sequencing methods for protein engineering and design. Curr. Opin. Struct. Biol. 45:36–44, 2017.

    Google Scholar 

  50. Xiong, W., et al. AAV cis-regulatory sequences are correlated with ocular toxicity. Proc. Natl. Acad. Sci. USA 116:5785–5794, 2019.

    Google Scholar 

  51. Yang, J., et al. Comparison of binding characteristics and in vitro activities of three inhibitors of vascular endothelial growth factor A. Mol. Pharm. 11:3421–3430, 2014.

    Google Scholar 

  52. Yorston, D. Anti-VEGF drugs in the prevention of blindness. Community Eye Health 27:44–46, 2014.

    Google Scholar 

Download references

Acknowledgments

The authors thank Patrick James Krohl for technical assistance with the project. The authors also acknowledge the NIH (R01EY031097, R01CA228133), the E. Matilda Ziegler Foundation for the Blind, the Louis B. Thalheimer Translational Fund, and Research to Prevent Blindness (Dr. H. James and Carole Free Catalyst Award and unrestricted grant) for support.

Conflict of interest

The authors declare that they have no conflicts of interest.

Ethical Approval

All animals were treated in accordance with the Association for Research in Vision and Ophthalmology Statement for Use of Animals in Ophthalmic and Vision Research, and protocols were reviewed and approved by the Johns Hopkins University Animal Care and Use Committee. No human studies were carried out by the authors for this article.

Author information

Authors and Affiliations

Authors

Contributions

RK, AZ, JS, SYT, LRA, and PRS designed, conducted, and analyzed experiments. PAC, JJG, and JBS conceptualized the study, supervised all research, interpreted data, and provided funding. RK, PRS, and JBS wrote the manuscript with input from co-authors.

Corresponding author

Correspondence to Jamie B. Spangler.

Additional information

Associate Editor Scott Simon oversaw the review of this article.

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

12195_2020_641_MOESM1_ESM.pptx

Supplementary Figure 1: VEGF-A affinity characterization of receptor variants. Bio-layer interferometry-based kinetic titrations of soluble Fc-fused aflibercept (a), Fc-fused VEGFR-1 domains 2 and 3 (VEGFR-1-Fc) (b), and Fc-fused VEGFR-2 domains 2 and 3 (VEGFR-2-Fc) (c) against immobilized VEGF-A. Fivefold titrations of soluble protein were used starting at 100 nM for aflibercept and VEGFR-1-Fc and starting at 250 nM for VEGFR-2-Fc. Binding constants were determined using Octet Data Analysis HT Software and are presented in Table 1.

Supplementary Figure 2: VEGF-A affinity characterization of enhanced-affinity aflibercept variants. Bio-layer interferometry-based kinetic titrations of soluble Fc-fused aflibercept variants C5 (a), D4 (b), and F3 (c) against immobilized VEGF-A. Fivefold dilutions of soluble protein were used starting at 100 nM for all aflibercept variants. Binding constants were measured using Octet Data Analysis HT Software and are presented in Table 2.

Supplementary Figure 3: DNA encapsulation in NPs. PBAE/DNA NPs were made by diluting DNA in 25 mM sodium acetate buffer at pH 5 (NaAc), and mixing with diluted PBAE at increasing polymer-to-DNA mass ratios (w/w). After 10 min of incubation for NP formation, sucrose was added, and the NPs were then diluted 1:11 (v/v) in additional NaAc (a) or PBS (b). Samples were mixed with 30% glycerol as a loading buffer at a 1:5 ratio (v/v) of loading buffer to NPs, then loaded into a 1% agarose gel with 1 µg/mL ethidium bromide. Each well contained 110 ng DNA. The gel was run for 30 min under 80 V, then visualized by UV exposure. DNA was completely bound in the NPs at 2 w/w or higher at pH 5, and even after dilution in PBS, DNA was completely bound at 5 w/w or higher.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kureshi, R., Zhu, A., Shen, J. et al. Structure-Guided Molecular Engineering of a Vascular Endothelial Growth Factor Antagonist to Treat Retinal Diseases. Cel. Mol. Bioeng. 13, 405–418 (2020). https://doi.org/10.1007/s12195-020-00641-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-020-00641-0

Keywords

Navigation