WO2017096124A1 - Heterodimeric vascular endothelial growth factor and use thereof - Google Patents

Heterodimeric vascular endothelial growth factor and use thereof Download PDF

Info

Publication number
WO2017096124A1
WO2017096124A1 PCT/US2016/064554 US2016064554W WO2017096124A1 WO 2017096124 A1 WO2017096124 A1 WO 2017096124A1 US 2016064554 W US2016064554 W US 2016064554W WO 2017096124 A1 WO2017096124 A1 WO 2017096124A1
Authority
WO
WIPO (PCT)
Prior art keywords
vegf
fusion protein
vegf1
cells
vegf121
Prior art date
Application number
PCT/US2016/064554
Other languages
French (fr)
Inventor
Alan Yueh-Luen LEE
Jui-Ling TSAI
Original Assignee
National Health Research Institutes
Kung, Hsing-Jien
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Health Research Institutes, Kung, Hsing-Jien filed Critical National Health Research Institutes
Priority to JP2018548648A priority Critical patent/JP6936808B2/en
Priority to US15/780,734 priority patent/US10851142B2/en
Priority to CN201680071128.4A priority patent/CN108699562B/en
Priority to EP16871555.5A priority patent/EP3384029B1/en
Publication of WO2017096124A1 publication Critical patent/WO2017096124A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • Angiogenesis is a critical rate-limiting process during tumor progression, which is induced by tilting the balance toward proangiogenic factors to drive vascular growth. Cancer cells in a microenvironment count on angiogenesis to supply oxygen and nutrients. Thus, agents targeting angiogenic pathways have been investigated as potential cancer drugs.
  • VEGF- A tumor-derived vascular endothelial growth factor A
  • VEGFR signaling VEGF-Trap
  • anti-VEGFR antibodies VEGF-Trap
  • anti-VEGFR antibodies VEGF- A monoclonal antibody Bevacizumab (Avastin)
  • Bevacizumab the first VEGF- A targeted antibody approved by the US FDA in 2004.
  • Tumors may develop resistance to antiangiogenic agents via adaptive responses, e.g., upregulating alternative proangiogenic signaling, co-opting normal peritumoral blood vessels, suppressing immune surveillance by recasting immune cells and bone-marrow-derived proangiogenic cells, and activating invasiveness phenotype.
  • adaptive responses are induced by intratumoral hypoxia that results from tumor vessel pruning and extensive suppression of angiogenesis.
  • a fusion protein that contains (i) a first vascular endothelial growth factor (VEGF) isoform, and (ii) a second VEGF isoform, and (iii) a dimerization domain between the first isoform and the second isoform.
  • the first isoform and the second isoform are selected from VEGF121 and VEGF1 6 5.
  • VEGF121 can have an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 2.
  • VEGFi 6 5 can have an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 4.
  • the dimerization domain contains two Fc regions and a linker between the two Fc regions.
  • the linker can be a flexible linker consisting of 15 to 30 amino acids.
  • the linker can be (Gly-Gly-Gly-Gly-Ser) n , n being 3, 4, 5, or 6.
  • the fusion protein includes, in the direction from the N-terminus to the C-terminus, VEGF121, one of the two Fc regions, the linker, the other of the two Fc regions, and VEGF1 6 5.
  • the fusion protein contains, in the direction from the N- terminus to the C-terminus, VEGF1 6 5, one of the two Fc regions, the linker, the other of the two Fc regions, and VEGF121.
  • the fusion protein can have an amino acid sequence that is at least 80% (e.g., identical to the sequence of SEQ ID NO: 11.
  • nucleic acid molecule that includes a nucleic acid sequence encoding the fusion protein described in this disclosure.
  • the nucleic acid sequence encodes an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 11.
  • a pharmaceutical composition that contains the fusion protein and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can be used to treat cancer or inhibit angiogenesis in a subject in need thereof.
  • FIG. 1 is a schematic representation of the VEGF isoforms.
  • FIG. 2 is a schematic representation of a VEGF121-VEGF1 6 5 fusion protein
  • FIG. 3 is a set of graphs showing the effect of the VEGF121-VEGF1 6 5 protein on cell proliferation.
  • a total of 1 x 10 4 3B-11 cells (A) and HCT-15 cells (B) were treated with the fusion protein (42, 83, 125 pM) in the presence of VEGF1 6 5 (222 pM).
  • 1 Control
  • 2 VEGF 165
  • 3 VEGF 165 + VEGF 12 i-VEGF 165 (42 pM)
  • FIG. 4 is a set of graphs showing the effect of the VEGF121-VEGF1 6 5 protein on tube formation.
  • 3B-11 (8 x 10 5 ) cells were inoculated on Matrigel and treated with VEGF121- VEGFies (42, 83, 125 pM) in the presence of VEGFi 65 (222 pM). Tube formation was quantified by counting the connected cells in randomly selected fields at lOOx magnification.
  • VEGF 121 -VEGF 165 plasmid was transfected into 3B-11 cells. Tube formation assay was carried out using the 3B- 11 cells. Data are presented as the means + SEM based on three independent experiments.
  • FIG. 5 is a graph showing the effect of the VEGFi 2 i-VEGFi 6 5 protein on 3B-11 cell migration.
  • VEGFi 2 i-VEGFi 6 5 plasmid was transfected into 3B-11 cells. Cell migration was determined using the transfected 3B-11 cells. Cell migration ability of 3B-11 cells was enhanced in the presence of VEGF1 6 5 (222 pM) but inhibited by the presence of the VEGFm- VEGF1 6 5 plasmid.
  • FIG. 6 is a set of graphs showing the effect of the VEGFi 2 i-VEGFi 6 5 protein on HCT- 15 cell migration.
  • HCT-15 cells were inoculated in Trans wellTM permeable inserts and treated with the VEGFi 2 i-VEGFi 65 protein (42, 83, 125 pM) or in the presence of VEGFi 65
  • VEGF 121 -VEGF 165 plasmid was transfected into HCT-15 cells. Cell migration was determined using the transfected HCT-15 cells.
  • FIG. 7 is a graph showing the effect of the VEGFi 2 i-VEGFi 6 5 protein on cell invasion.
  • HCT-15 cells were treated with different concentrations of VEGF i 2 i-VEGFi 65 (42, 83, 125 pM) in the presence of VEGF1 6 5 (222 pM).
  • Cell invasion was determined by the transwell chamber assay. After migrating for 48 h, the number of cells passing through the filter was counted after staining with crystal violet (original magnification: 400x).
  • VEGF 165 + VEGF 12 i-VEGF 165 (42 pM); 4: VEGF 165 + VEGF 12 i-VEGF 165 (83 pM); 5: VEGF 165 + VEGF 12 i-VEGF 165 (125 pM).
  • FIG. 8 is a set of graphs showing the effect of the VEGF121-VEGF1 6 5 protein in a xenograft tumorigenesis assay.
  • HCT-15 cells were injected subcutaneously into the dorsal flank of nude mice (1 site per mouse). Injected mice were examined every two days for tumor formation. Different concentrations of VEGF121-VEGF1 6 5 protein (10, 50, or
  • tumor volume 1 x w 2 x 0.52.
  • VEGF vascular endothelial growth factor
  • a fusion VEGF protein containing isoform VEGF121 and isoform VEGF1 6 5 linked by a dimerization domain.
  • the VEGF-A gene includes eight exons, which can give rise to alternatively spliced variants, i.e., VEGF121, VEGF145, VEGFi 65 , VEGFi 83 , VEGFi 89 , and VEGF 206 .
  • VEGF121 is a freely soluble and weakly acidic polypeptide that lacks a heparin- binding domain.
  • a VEGF 12 i nucleic acid sequence (SEQ ID NO: 1) and the amino acid sequence it encodes (SEQ ID NO: 2) are provided herewith.
  • the sequence of SEQ ID NO: 2 includes an N-terminal signal peptide, which is not present in the mature form of VEGF121.
  • VEGF1 6 5 contains basic amino acids and a heparin-binding domain that binds the
  • VEGF receptor to induce signal transduction and stimulate endothelial cell proliferation.
  • a VEGF1 6 5 nucleic acid sequence SEQ ID NO: 3
  • amino acid sequence it encodes SEQ ID NO: 4
  • the sequence of SEQ ID NO: 4 includes an N- terminal signal peptide, which is not present in the mature form of VEGF1 6 5.
  • the fusion VEGF further includes a dimerization domain positioned in between the two VEGF isoforms such that the fusion VEFG forms a heterodimer.
  • the dimerization domain consists of two Fc regions linked by a linker.
  • the Fc region can be a human IgG Fc region.
  • the Fc region can have the amino acid sequence of SEQ ID NO: 6, which is encoded by the nucleic acid sequence of SEQ ID NO: 5.
  • the linker between the two Fc regions can be any flexible linker known in the art.
  • the linker can have between 15 and 30 amino acids.
  • a flexible linker can be a Gly- and Ser- rich linker.
  • the linker can be (Gly-Gly-Gly-Gly-Ser) n (SEQ ID NO:7), n being an integer (e.g., 1, 2, 3, 4, 5, 6, 7, or 8).
  • the fusion protein can further include a signal peptide at the N-terminus.
  • the signal peptide can be the signal peptide endogenous to the VEGF isoforms.
  • the signal peptide can have the sequence of SEQ ID NO: 9 (encoded by the nucleic acid sequence of SEQ ID NO: 8).
  • the C-terminal VEGF isoform in the fusion protein may or may not include a signal peptide.
  • the fusion protein can include a C-terminal tag to facilitate isolation or identification of the fusion protein.
  • a C-terminal tag can be a poly(His) tag, HA tag, Myc tag, V5, or FLAG tag.
  • the fusion protein can contain, in the direction from the N-terminus to the C- terminus, VEGFi 6 5, an Fc region, a linker, an Fc region, and VEGF121.
  • the fusion protein can contain, in the direction from the N-terminus to the C-terminus, VEGF121, an Fc region, a linker, an Fc region, and VEGF1 6 5.
  • Each isoform can be linked to an Fc region directly or indirectly via a linker, which can be different or identical to the linker between the two Fc regions.
  • the fusion protein has an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 11, which is encoded by the sequence of SEQ ID NO: 10.
  • fusion protein e.g., recombinant technology
  • an expression construct encoding the protein can be generated and introduced into suitable host cells (e.g., mammalian cells).
  • suitable host cells e.g., mammalian cells.
  • the fusion protein expressed in the host cells can then be isolated.
  • the fusion protein can be mixed with a pharmaceutically acceptable carrier to form a pharmaceutical composition.
  • the composition can be administered to a subject in need thereof to treat cancer or inhibit angiogenesis.
  • the fusion protein can also be used in a combination therapy with other cancer treatments.
  • the fusion protein can also be conjugated to or encapsulated in moieties (e.g., lipids, carbohydrates, polymers or nanoparticles) designed to target the fusion protein to tumors and/or their associated vasculature.
  • composition can be formulated with a pharmaceutically acceptable carrier such as 5 a phosphate buffered saline, a bicarbonate solution, and/or an adjuvant.
  • a pharmaceutically acceptable carrier such as 5 a phosphate buffered saline, a bicarbonate solution, and/or an adjuvant.
  • compositions may be prepared as an injectable, liquid solution, emulsion, or another suitable formulation.
  • adjuvants include, but are not limited to, alum-precipitate, Freund's0 complete adjuvant, Freund's incomplete adjuvant, monophosphoryl-lipid A/trehalose
  • dicorynomycolate adjuvant water in oil emulsion containing Corynebacterium parvum and tRNA, and other substances that accomplish the task of increasing immune response by mimicking specific sets of evolutionarily conserved molecules including liposomes, lipopoly saccharide (LPS), molecular cages for antigen, components of bacterial cell walls, 5 and endocytosed nucleic acids such as double-stranded RNA, single-stranded DNA, and unmethylated CpG dinucleotide-containing DNA.
  • liposomes lipopoly saccharide (LPS)
  • LPS lipopoly saccharide
  • molecular cages for antigen components of bacterial cell walls
  • 5 and endocytosed nucleic acids such as double-stranded RNA, single-stranded DNA, and unmethylated CpG dinucleotide-containing DNA.
  • Other examples include cholera toxin, E. coli heat-labile enterotoxin, liposome,
  • the composition can also include a polymer that facilitates in vivo delivery.
  • parenterally e.g., subcutaneous injection, intravenous injection, or intramuscular injection.
  • Other routes of administration may also be used.
  • a skilled practitioner would be able to determine the appropriate dosage and route of administration.
  • Cancers that can be treated with the fusion protein include solid tumors such as 5 glioblastoma, colorectal cancer, lung cancer, renal cancer, liver cancer, kidney cancer,
  • neuroendocrine tumors breast cancer, esophageal cancer, gastrointestinal stromal tumors, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate cancer, stomach cancer, and head and neck cancer.
  • VEGF121-VEGF1 6 5 fused with two Fc regions of human IgGl as a powerful antiangiogenic modulator. It was found that the chimeric VEGF121-VEGF1 6 5 recombinant protein reduced tube formation of 3B-11 endothelial cells and inhibited invasiveness of HCT-15 cancer cells. Furthermore, we found that the VEGF121- VEGFies protein attenuated VEGFR2-HIF-la signaling through the PI3K-AKT-mTOR pathway in cancer cells. The data demonstrated that the chimeric VEGF121-VEGF1 6 5 protein antagonizes angiogenesis and HIF- ⁇ signaling, and suggested that it could combat drug resistance to antiangiogenic therapy.
  • a VEGF121-VEGF1 6 5 fusion was generated by fusing a human IgGl Fc nucleic acid sequence to the 3' terminus of a VEGF121 sequence and the 5' terminus of a VEGF1 6 5 sequence, respectively, and the two Fc sequences were connected by a linker sequence.
  • the VEGF121-VEGF1 6 5 fusion nucleic acid sequence was cloned into the pcDNA3.1 vector, yielding the expression vector for VEGF121-VEGF1 6 5.
  • the integrity of the final construct was confirmed by DNA sequencing.
  • the deduced protein includes a putative 26-aa signal peptide. See Fig. 2.
  • the plasmid containing the VEGF121-VEGF1 6 5 fusion gene was transfected into 293T cell line.
  • the expression and secretion of VEGF121-VEGF1 6 5 fusion protein were confirmed by Western blot.
  • the recombinant protein VEGF121-VEGF1 6 5 was expressed as a His-tag fusion protein in 293T cells and purified using nickel affinity chromatography. The purity and the molecular weight of the purified fusion protein were determined by Western blot (data not shown).
  • VEGF121-VEGF1 6 5 fusion protein inhibited cell proliferation induced by VEGF1 6 5
  • VEGF121-VEGF1 6 5 protein affected proliferation of VEGF1 6 5- stimulated 3B-11 cells, a convenient endothelial cell model for tube formation assay. See Zhou et al., Methods 2008, 44(2): 190-195. VEGF 165 -induced cell growth of 3B-11 was blocked in a concentration-dependent manner by VEGF121-VEGF1 6 5. See Fig. 3, panel A.
  • VEGF121-VEGF1 6 5 The effect of VEGF1 6 5 at a concentration of 222 pM on 3B-11 cell proliferation was easily inhibited by the recombinant VEGF121-VEGF1 6 5 at 42 pM, suggesting that the VEGF121- VEGF1 6 5 protein was able to efficiently block the activity of VEGFi 6 5-induced proliferation. Moreover, the VEGF121-VEGF1 6 5 protein exhibited similar potency at inhibiting VEGF1 6 5- induced growth of HCT-15 cancer cells. See FIG. 3, panel B.
  • VEGF121-VEGF1 6 5 exhibited similar activity in inhibiting cell proliferation of 3B-11 and HCT-15 cells in an autocrine manner when the plasmid of the fusion gene was transfected into these cells, although the inhibition was not statistically significant (data not shown).
  • the results suggest that VEGF121-VEGF1 6 5 inhibit the increase in cell number due to suppression of proliferation, not cytotoxicity.
  • VEGF121-VEGF1 6 5 inhibited cell transformation of HCT-15 in an autocrine manner (data not shown).
  • Our results indicate that the VEGF121-VEGF1 6 5 recombinant protein can block cell proliferation of 3B-11 as well as cell proliferation and transformation of colon cancer cell HCT-15 in autocrine and paracrine manners.
  • VEGF121-VEGF1 6 5 fusion protein inhibited tube formation induced by VEGF1 6 5
  • VEGF121-VEGF1 6 5 fusion protein inhibited cell migration
  • Cell migration is a critical process in angiogenesis and tumor metastasis.
  • Cell migration was examined by a gap-closure migration assay.
  • the VEGF121- VEGF1 6 5 protein significantly inhibited migration of 3B-11 cells in an autocrine manner. See Fig. 5.
  • the results showed that cell migration of HCT-15 induced by VEGF1 6 5 was inhibited by the addition of the VEGF121-VEGF1 6 5 protein in a concentration-dependent manner. See Fig. 6, panel A.
  • VEGF121-VEGF1 6 5 protein significantly inhibited cell migration in a paracrine (Fig. 6, A) and an autocrine manner (Fig. 6, B). These data suggested that VEGF121-VEGF1 6 5 chimeric protein can inhibit migration of endothelial cells and tumor cells.
  • VEGF121-VEGF1 6 5 fusion protein impaired tumor invasion
  • VEGF121-VEGF1 6 5 chimeric protein's effect on metastasis of tumor cells.
  • VEGF121-VEGF1 6 5 chimeric protein's effect on metastasis of tumor cells.
  • VEGF1 6 5 induced invasive capability as indicated by intensive penetration. See Fig. 7.
  • VEGFi 6 5-induced cell invasion was inhibited by the addition of VEGF 12 i-VEGF 165 in a concentration-dependent manner in HCT-15 cancer cells.
  • the number of penetrated cells was significantly decreased when treated with increased concentrations of the VEGF121-VEGF1 6 5 protein as compared with VEGF1 6 5 only. See Fig. 7.
  • VEGF121-VEGF1 6 5 attenuates HIF- ⁇ signaling to decrease the resistance to anti- angiogenic therapy.
  • VEGF121-VEGF1 6 5 attenuates HIF- ⁇ signaling to decrease the resistance to anti- angiogenic therapy.
  • Lon is upregulated by the hypoxia inducible factor- la (HIF-la) and involved in response to low oxygen availability, which adapt cancer cells to a hypoxic environment.
  • HIF-la hypoxia inducible factor- la
  • fusion protein influenced the expression of Lon protease.
  • the expression of HIF-la, VEGF1 6 5, and Lon was determined by Western blot analysis. The results showed that VEGFR2-HIF-la-
  • VEGFi 6 5/Lon signaling in HCT-15 cancer cells was activated by VEGF1 6 5 treatment and hypoxia stimulated by cobalt chloride (C0CI 2 ) (data not shown).
  • the signaling activation was inhibited by the addition of VEGF 121 -VEGF 165 in a concentration-dependent manner under normoxia and hypoxia conditions.
  • the recombinant VEGF 121 -VEGF 165 protein reduced the level of HIF- ⁇ , VEGF 165 , Lon, and phospho-VEGFR2 induced by VEGF 165 and/or hypoxia (data not shown).
  • VEGF 121 -VEGF 165 protein inhibited the activation of VEGFR2-HIF-la-VEGFi 6 5/Lon signaling through repressing PBK-AKT-mTOR pathway (data not shown), suggesting that the VEGF 121 -VEGF 165 protein overcame survival mechanism triggered by the PI3K-AKT-mTOR to VEGFR2-HIF-la- VEGFi 6 5/Lon axis under hypoxia.
  • VEGF 121 -VEGF 165 can inhibit autocrine VEGFR2-HIF-la-VEGFi 65 /Lon signaling through PBK-AKT-mTOR pathway in cancer cells.
  • VEGF 121 -VEGF 165 fusion protein impaired tumor growth in vivo
  • VEGF 12 i-VEGF 165 protein can arrest tube formation of endothelial cells and interfere with tumor cell growth, migration and invasion in vitro.
  • a xenograft tumorigenesis assay was performed to demonstrate the inhibitory effect of the VEGF 12 i-VEGF 165 protein on tumors in vivo.
  • BALB/c nude mice (6-8 weeks old) were used in the assay.
  • lxlO 6 HCT-15 cells suspended in 0.2 ml of Matrigel were injected subcutaneously into the dorsal flank of nude mice (1 site per mouse). The mice were examined every two days for tumor formation.
  • VEGF121- VEGF 165 protein 10, 50, or 250 ng/ml
  • PBS control a PBS control
  • the body weight of the mice treated with the VEGF 121 -VEGF 165 chimeric protein did not change significantly, suggesting that the protein was not toxic to the animals. See Fig. 8, panel A.
  • the chimeric protein reduced tumor growth in the mice in a dose-dependent manner.
  • Fig. 8, panel B See Fig. 8, panel B.
  • the study demonstrated that the VEGF 121 -VEGF 165 chimeric protein can suppress tumor growth in vivo.
  • 3B-11 cells were purchased from ATCC (#CRL-2160, Manassas, VA, USA). 3B-11 cells were maintained in DMEM and HCT-15 cells in RPMI-
  • FBS fetal bovine serum qualified; Invitrogen
  • PSA penicillin-streptomycin amphotericin B ; Biological industries, NY, USA
  • VEGF 121 -VEGF 165 recombinant proteins The plasmids were transfected into 293T cells using the Biomics transfection reagent as described in the instruction manual of the pcDNA3.1 vector helper- free system (Biomics). After incubation for 36 hours, the supernatant of the 293T cell culture was collected and purified on Ni-NTA resin, eluted with 250 mmol/L imidazole according to the instruction manual. The recombinant protein was concentrated by the Microcon Centrifugal Filter Unit (Millipore, Bedford, MA, USA). Finally, the purified protein was confirmed by 10% SDS-PAGE and Western Blotting.
  • Cell proliferation assay The proliferation of 3B-11 cells and HCT-15 cells was assessed using CCK-8 dye reduction assay (Enzo, USA). 3B-11 or HCT-15 cells were pre- treated with different concentrations of VEGF 12 i-VEGF 165 (42, 83, 125 pM) for 30 min, a commercial VEGF 165 was added (222 pM, Abeam, Cambridge, MA, USA), and then the cells were incubated for 24 hours. At the end of the treatment, 10 ⁇ of the CCK-8 solution was added to each well of the plate and the plate was incubated for 2 ⁇ 4 hours in the incubator. After shaking the plate for 10 seconds, cell viability was assessed by measuring the absorbance at 450 nm using a microplate reader. All measurements were performed three times. The ⁇ -test was used to compare groups. Data are presented as mean + SD.
  • Clonogenic assay is an in vitro transformation assay based on the ability of a single cell to grow into a colony. To examine this, the plasmid of
  • VEGF 121 -VEGF 165 was transfected into HCT-15 in a 10 cm dish overnight and treated with VEGF 165 recombinant protein 222 pM as a positive control.
  • the treated HCT-15 cells ( ⁇ lxlO 3 per well) were plated in 6-well plates and incubated in a 37 °C incubator. Fresh RPMI medium containing 10% FBS was added every 48 hours.
  • cells were washed twice with ice cold PBS, fixed with methanol for 10 minutes and then stained with 1% crystal violet in methanol for 15 minutes followed by washing with deionized water. Colonies with more than 50 cells were scored and counted under the microscope at 200x.
  • Cell migration assay was determined by gap closure assay. 3B-
  • 11 cells or HCT-15 cells were treated with different concentrations (42, 83, 125 pM) of the recombinant proteins for 16 h (37 °C, 5% CO 2 ). These cells were trypsinized and resuspended in serum- free DMEM or RPMI-1640 medium. A total of 8xl0 5 cells in 70 ⁇ serum-free DMEM or RPMI-1640 were seeded in medium in each well (8xl0 5 cells/ well) and incubated at 37 °C, 5% CO 2. Next day, the ibidi culture-insert (Applied Biophysics, USA) was gently removed by using sterile tweezers, and each well was then filled with 2 ml 0.1% FBS medium. Cell migration was monitored for 48 h by microscope.
  • Tube formation assay Corning Matrigel® Matrix (BD Biosciences, San Jose, CA, USA) solution was thawed on ice overnight and 50 ⁇ aliquots were coated onto a 96-well plate and incubated at 37°C for lh to solidify. 50 ⁇ of DMEM supplemented with 10% FBS medium containing about 8xl0 5 3B-11 cells was seeded onto the plated Matrigel Matrix and incubated at 37°C. These cells were treated as previously described. The assay was done in triplicate and was incubated at 37 °C with 5% CO 2 .
  • Cell invasion assay Cell invasion was evaluated using a transwell chamber (Corning Costar; Cambridge, MA, USA) equipped with a Matrigel-coated filter membrane (8 ⁇ pores). Briefly, the filters were pre-coated with 200 ⁇ g/ml basement membrane proteins (Matrigel; BD Biosciences, San Jose, CA, USA) and allowed to dry overnight at 37°C with 5% C0 2 . HCT-15 cells (8xl0 5 ) in FBS-free medium were seeded in the upper chambers, and lower wells were filled with 10% FBS medium. After incubation at 37°C for 48h, non- migratory cells on the upper side of the insert were removed with a cotton swab. The cells that had passed through the filter were fixed in methanol and stained with crystal violet.
  • HCT-15 cells were seeded onto a 10 mm dish at a density of 1.5xl0 6 cells in 10 ml medium for 24 h under normoxia and hypoxia (C0CI 2 , Cobalt dichloride; 150 ⁇ ) and treated as previously described.
  • Total protein concentrations were determined using a BCA protein assay. Equal quantities of total protein were resolved using 10% SDS-PAGE and electroblotted onto polyvinylidene fluoride membranes. Membranes were blocked with
  • Phospho-PI3K (Tyr458/ Tyrl99, #4228), phospho- AKT (Ser473, #4060), and phoshpo-mTOR (Ser2448, #2971) antibodies were obtained from Cell Signaling Technology (Beverly, MA, USA); HIF-la (#610958) antibodies was obtained from BD Biosciences (Franklin Lakes, NJ); phospho-VEGFR2 (Tyrl054/Tyrl059, ab5473), VEGF-165A (ab69479) antibodies were from Abeam (Cambridge, MA, USA); beta-actin antibody was fromfrom GeneTex (GTX109639, Hsinchu, Taiwan).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Vascular Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicinal Preparation (AREA)

Abstract

A fusion protein, comprising: (i) a first vascular endothelial growth factor (VEGF) isoform, and (ii) a second VEGF isoform, and (iii) a dimerization domain between the first isoform and the second isoform, wherein the first isoform and the second isoform are selected from VEGF12i and VEGF165.

Description

HETERODIMERIC VASCULAR ENDOTHELIAL GROWTH FACTOR
AND USE THEREOF
CROSS-REFERENCE TO RELATED APPLICATION This application claims priority to U.S. Provisional Application No. 62/262,630, filed on December 3, 2015, the entire content of which is hereby incorporated by reference herein.
BACKGROUND
Angiogenesis is a critical rate-limiting process during tumor progression, which is induced by tilting the balance toward proangiogenic factors to drive vascular growth. Cancer cells in a microenvironment count on angiogenesis to supply oxygen and nutrients. Thus, agents targeting angiogenic pathways have been investigated as potential cancer drugs.
Initial efforts have primarily focused on targeting endothelial and tumor-derived vascular endothelial growth factor A (VEGF- A)/VEGFR signaling. Several different strategies have been designed to inhibit this signaling as monotherapy or adjuvant therapies, e.g., small molecules inhibitors of VEGFR signaling, VEGF-Trap, anti-VEGFR antibodies, and the anti- VEGF- A monoclonal antibody Bevacizumab (Avastin), the first VEGF- A targeted antibody approved by the US FDA in 2004.
However, a significant number of patients either do not respond to antiangiogenic agents or rapidly develop resistance to them. Tumors may develop resistance to antiangiogenic agents via adaptive responses, e.g., upregulating alternative proangiogenic signaling, co-opting normal peritumoral blood vessels, suppressing immune surveillance by recasting immune cells and bone-marrow-derived proangiogenic cells, and activating invasiveness phenotype. These adaptive responses are induced by intratumoral hypoxia that results from tumor vessel pruning and extensive suppression of angiogenesis.
It remains a major challenge to efficiently inhibit VEGF-A/VEGFR signaling and, at the same time, alleviate resistance to antiangiogenic therapy.
SUMMARY
In one aspect, provided herein is a fusion protein that contains (i) a first vascular endothelial growth factor (VEGF) isoform, and (ii) a second VEGF isoform, and (iii) a dimerization domain between the first isoform and the second isoform. The first isoform and the second isoform are selected from VEGF121 and VEGF165. VEGF121 can have an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 2. VEGFi65 can have an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 4.
In one embodiment, the dimerization domain contains two Fc regions and a linker between the two Fc regions. The linker can be a flexible linker consisting of 15 to 30 amino acids. For example, the linker can be (Gly-Gly-Gly-Gly-Ser)n, n being 3, 4, 5, or 6.
In one embodiment, the fusion protein includes, in the direction from the N-terminus to the C-terminus, VEGF121, one of the two Fc regions, the linker, the other of the two Fc regions, and VEGF165.
In another embodiment, the fusion protein contains, in the direction from the N- terminus to the C-terminus, VEGF165, one of the two Fc regions, the linker, the other of the two Fc regions, and VEGF121.
The fusion protein can have an amino acid sequence that is at least 80% (e.g., identical to the sequence of SEQ ID NO: 11.
In another aspect, provided herein is a nucleic acid molecule that includes a nucleic acid sequence encoding the fusion protein described in this disclosure. In one embodiment, the nucleic acid sequence encodes an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 11.
In yet another aspect, provided herein is a pharmaceutical composition that contains the fusion protein and a pharmaceutically acceptable carrier. The pharmaceutical composition can be used to treat cancer or inhibit angiogenesis in a subject in need thereof.
The details of one or more embodiments are set forth in the accompanying drawing and the description below. Other features, objects, and advantages of the embodiments will be apparent from the description and drawing, and from the claims. BRIEF DESCRIPTION OF DRAWINGS
FIG. 1 is a schematic representation of the VEGF isoforms.
FIG. 2 is a schematic representation of a VEGF121-VEGF165 fusion protein
FIG. 3 is a set of graphs showing the effect of the VEGF121-VEGF165 protein on cell proliferation. A total of 1 x 104 3B-11 cells (A) and HCT-15 cells (B) were treated with the fusion protein (42, 83, 125 pM) in the presence of VEGF165 (222 pM). Cell proliferation was measured by a CCK-8 kit (t-test, *P < 0.05, **P < 0.01, n = 3). 1: Control; 2: VEGF165; 3: VEGF165 + VEGF12i-VEGF165 (42 pM); 4: VEGF165 + VEGF12i-VEGF165 (83 pM);
5: VEGF165 + VEGF12i-VEGF165 (125 pM).
FIG. 4 is a set of graphs showing the effect of the VEGF121-VEGF165 protein on tube formation. (A) 3B-11 (8 x 105) cells were inoculated on Matrigel and treated with VEGF121- VEGFies (42, 83, 125 pM) in the presence of VEGFi65 (222 pM). Tube formation was quantified by counting the connected cells in randomly selected fields at lOOx magnification. 1: Control; 2: VEGF165; 3: VEGF165 + VEGF12i-VEGF165 (42 pM); 4: VEGF165 + VEGF121- VEGF165 (83 pM); 5: VEGF165 + VEGF12i-VEGF165 (125 pM). (B) VEGF121-VEGF165 plasmid was transfected into 3B-11 cells. Tube formation assay was carried out using the 3B- 11 cells. Data are presented as the means + SEM based on three independent experiments. 1: Control; 2: VEGF165 (222 pM); 3: VEGF121-VEGF165 plasmid. *P < 0.05, **P < 0.01, ***p < 0.001.
FIG. 5 is a graph showing the effect of the VEGFi2i-VEGFi65 protein on 3B-11 cell migration. VEGFi2i-VEGFi65 plasmid was transfected into 3B-11 cells. Cell migration was determined using the transfected 3B-11 cells. Cell migration ability of 3B-11 cells was enhanced in the presence of VEGF165 (222 pM) but inhibited by the presence of the VEGFm- VEGF165 plasmid.
FIG. 6 is a set of graphs showing the effect of the VEGFi2i-VEGFi65 protein on HCT- 15 cell migration. (A) HCT-15 cells were inoculated in Trans well™ permeable inserts and treated with the VEGFi2i-VEGFi65 protein (42, 83, 125 pM) or in the presence of VEGFi65
(222 pM). The distance between the gap was analyzed in Transwell™ permeable inserts. Untreated HCT-15 cells were used as controls. 1: Control; 2: VEGF165; 3: VEGF165 + VEGF121-VEGF165 (42 pM); 4: VEGF165 + VEGF121-VEGF165 (83 pM); 5: VEGF165 + VEGF121-VEGF165 (125 pM). (B) VEGF121-VEGF165 plasmid was transfected into HCT-15 cells. Cell migration was determined using the transfected HCT-15 cells. Cell migration ability of HCT-15 cells was enhanced in the presence of VEGF165 (222 pM) but inhibited by the presence of VEGF121-VEGF165 plasmid. i-test, *P < 0.05, **P < 0.01, ***p < 0.001, n = 3.
FIG. 7 is a graph showing the effect of the VEGFi2i-VEGFi65 protein on cell invasion. HCT-15 cells were treated with different concentrations of VEGF i2i-VEGFi65 (42, 83, 125 pM) in the presence of VEGF165 (222 pM). Cell invasion was determined by the transwell chamber assay. After migrating for 48 h, the number of cells passing through the filter was counted after staining with crystal violet (original magnification: 400x). 1: Control; 2: VEGF165; 3: VEGF165 + VEGF12i-VEGF165 (42 pM); 4: VEGF165 + VEGF12i-VEGF165 (83 pM); 5: VEGF165 + VEGF12i-VEGF165 (125 pM). i-test, *P < 0.05, **P < 0.01, *** p < 0.001.
FIG. 8 is a set of graphs showing the effect of the VEGF121-VEGF165 protein in a xenograft tumorigenesis assay. HCT-15 cells were injected subcutaneously into the dorsal flank of nude mice (1 site per mouse). Injected mice were examined every two days for tumor formation. Different concentrations of VEGF121-VEGF165 protein (10, 50, or
250 ng/ml) or a PBS control were directly injected into the tumors in the mice. (A) The body weight of injected mice was monitored. (B) Tumor volume was estimated from its length and width, as measured by a 6-inch-dial caliper, using the formula: tumor volume = 1 x w2 x 0.52.
DETAILED DESCRIPTION
It was surprisingly discovered that a heterodimeric vascular endothelial growth factor (VEGF) composed of two different VEGF isoforms reduced proliferation, migration, invasion, and tube formation in endothelial and cancer cells through competing with VEGF165 homodimer in a paracrine and an autocrine manner.
Therefore, described herein is a fusion VEGF protein containing isoform VEGF121 and isoform VEGF165 linked by a dimerization domain.
The VEGF-A gene includes eight exons, which can give rise to alternatively spliced variants, i.e., VEGF121, VEGF145, VEGFi65, VEGFi83, VEGFi89, and VEGF206. See Fig. 1 VEGF121 is a freely soluble and weakly acidic polypeptide that lacks a heparin- binding domain. A VEGF12i nucleic acid sequence (SEQ ID NO: 1) and the amino acid sequence it encodes (SEQ ID NO: 2) are provided herewith. The sequence of SEQ ID NO: 2 includes an N-terminal signal peptide, which is not present in the mature form of VEGF121.
VEGF165 contains basic amino acids and a heparin-binding domain that binds the
VEGF receptor to induce signal transduction and stimulate endothelial cell proliferation. A VEGF165 nucleic acid sequence (SEQ ID NO: 3) and the amino acid sequence it encodes (SEQ ID NO: 4) are provided herewith. The sequence of SEQ ID NO: 4 includes an N- terminal signal peptide, which is not present in the mature form of VEGF165.
The fusion VEGF further includes a dimerization domain positioned in between the two VEGF isoforms such that the fusion VEFG forms a heterodimer. In one embodiment, the dimerization domain consists of two Fc regions linked by a linker. The Fc region can be a human IgG Fc region. For example, the Fc region can have the amino acid sequence of SEQ ID NO: 6, which is encoded by the nucleic acid sequence of SEQ ID NO: 5.
The linker between the two Fc regions can be any flexible linker known in the art. The linker can have between 15 and 30 amino acids. A flexible linker can be a Gly- and Ser- rich linker. For example, the linker can be (Gly-Gly-Gly-Gly-Ser)n (SEQ ID NO:7), n being an integer (e.g., 1, 2, 3, 4, 5, 6, 7, or 8).
The fusion protein can further include a signal peptide at the N-terminus. The signal peptide can be the signal peptide endogenous to the VEGF isoforms. For example, the signal peptide can have the sequence of SEQ ID NO: 9 (encoded by the nucleic acid sequence of SEQ ID NO: 8). The C-terminal VEGF isoform in the fusion protein may or may not include a signal peptide.
In addition, the fusion protein can include a C-terminal tag to facilitate isolation or identification of the fusion protein. Such tag can be a poly(His) tag, HA tag, Myc tag, V5, or FLAG tag.
The fusion protein can contain, in the direction from the N-terminus to the C- terminus, VEGFi65, an Fc region, a linker, an Fc region, and VEGF121. Alternatively, the fusion protein can contain, in the direction from the N-terminus to the C-terminus, VEGF121, an Fc region, a linker, an Fc region, and VEGF165. Each isoform can be linked to an Fc region directly or indirectly via a linker, which can be different or identical to the linker between the two Fc regions. In one embodiment, the fusion protein has an amino acid sequence that is at least 80% (e.g., at least 99%, 98%, 97%, 96%, 95%, 90%, or 85%) identical to the sequence of SEQ ID NO: 11, which is encoded by the sequence of SEQ ID NO: 10.
Conventional methods, e.g., recombinant technology, can be used to make the fusion protein. For example, an expression construct encoding the protein can be generated and introduced into suitable host cells (e.g., mammalian cells). The fusion protein expressed in the host cells can then be isolated.
The fusion protein can be mixed with a pharmaceutically acceptable carrier to form a pharmaceutical composition. The composition can be administered to a subject in need thereof to treat cancer or inhibit angiogenesis. The fusion protein can also be used in a combination therapy with other cancer treatments. The fusion protein can also be conjugated to or encapsulated in moieties (e.g., lipids, carbohydrates, polymers or nanoparticles) designed to target the fusion protein to tumors and/or their associated vasculature.
The composition can be formulated with a pharmaceutically acceptable carrier such as 5 a phosphate buffered saline, a bicarbonate solution, and/or an adjuvant. Suitable
pharmaceutical carriers and diluents, as well as pharmaceutical necessities for their use, are known in the art. This composition may be prepared as an injectable, liquid solution, emulsion, or another suitable formulation.
Examples of adjuvants include, but are not limited to, alum-precipitate, Freund's0 complete adjuvant, Freund's incomplete adjuvant, monophosphoryl-lipid A/trehalose
dicorynomycolate adjuvant, water in oil emulsion containing Corynebacterium parvum and tRNA, and other substances that accomplish the task of increasing immune response by mimicking specific sets of evolutionarily conserved molecules including liposomes, lipopoly saccharide (LPS), molecular cages for antigen, components of bacterial cell walls, 5 and endocytosed nucleic acids such as double-stranded RNA, single-stranded DNA, and unmethylated CpG dinucleotide-containing DNA. Other examples include cholera toxin, E. coli heat-labile enterotoxin, liposome, immune- stimulating complex (ISCOM),
immunostimulatory sequences oligodeoxynucleotide, and aluminum hydroxide. The composition can also include a polymer that facilitates in vivo delivery.
o An effective amount of the composition described above may be administered
parenterally, e.g., subcutaneous injection, intravenous injection, or intramuscular injection. Other routes of administration may also be used. A skilled practitioner would be able to determine the appropriate dosage and route of administration.
Cancers that can be treated with the fusion protein include solid tumors such as 5 glioblastoma, colorectal cancer, lung cancer, renal cancer, liver cancer, kidney cancer,
neuroendocrine tumors, breast cancer, esophageal cancer, gastrointestinal stromal tumors, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate cancer, stomach cancer, and head and neck cancer.
The specific example below is to be construed as merely illustrative, and not o limitative of the remainder of the disclosure in any way whatsoever. Without further
elaboration, it is believed that one skilled in the art can, based on the description herein, utilize the present disclosure to its fullest extent. All publications cited herein are hereby incorporated by reference in their entirety.
EXAMPLE
We generated a novel chimeric dimer of VEGF121-VEGF165 fused with two Fc regions of human IgGl as a powerful antiangiogenic modulator. It was found that the chimeric VEGF121-VEGF165 recombinant protein reduced tube formation of 3B-11 endothelial cells and inhibited invasiveness of HCT-15 cancer cells. Furthermore, we found that the VEGF121- VEGFies protein attenuated VEGFR2-HIF-la signaling through the PI3K-AKT-mTOR pathway in cancer cells. The data demonstrated that the chimeric VEGF121-VEGF165 protein antagonizes angiogenesis and HIF-Ια signaling, and suggested that it could combat drug resistance to antiangiogenic therapy.
Construction and characterization of a VEGF12i-VEGF165 fusion protein
A VEGF121-VEGF165 fusion was generated by fusing a human IgGl Fc nucleic acid sequence to the 3' terminus of a VEGF121 sequence and the 5' terminus of a VEGF165 sequence, respectively, and the two Fc sequences were connected by a linker sequence. The VEGF121-VEGF165 fusion nucleic acid sequence was cloned into the pcDNA3.1 vector, yielding the expression vector for VEGF121-VEGF165. The integrity of the final construct was confirmed by DNA sequencing. The deduced protein includes a putative 26-aa signal peptide. See Fig. 2. The plasmid containing the VEGF121-VEGF165 fusion gene was transfected into 293T cell line. The expression and secretion of VEGF121-VEGF165 fusion protein were confirmed by Western blot. We found a single band of -120 kDa for VEGF121-VEGF165 fusion protein in samples from cultured medium and cell lysates of transfected 293T cells (data not shown). The recombinant protein VEGF121-VEGF165 was expressed as a His-tag fusion protein in 293T cells and purified using nickel affinity chromatography. The purity and the molecular weight of the purified fusion protein were determined by Western blot (data not shown). These results indicated that VEGF121-VEGF165 formed a dimer covalently linked by IgGl Fc fragments and a polypeptide linker.
VEGF121-VEGF165 fusion protein inhibited cell proliferation induced by VEGF165
Since endothelial cell proliferation is required for early angiogenic response, we examined whether the VEGF121-VEGF165 protein affected proliferation of VEGF165- stimulated 3B-11 cells, a convenient endothelial cell model for tube formation assay. See Zhou et al., Methods 2008, 44(2): 190-195. VEGF165-induced cell growth of 3B-11 was blocked in a concentration-dependent manner by VEGF121-VEGF165. See Fig. 3, panel A. The effect of VEGF165 at a concentration of 222 pM on 3B-11 cell proliferation was easily inhibited by the recombinant VEGF121-VEGF165 at 42 pM, suggesting that the VEGF121- VEGF165 protein was able to efficiently block the activity of VEGFi65-induced proliferation. Moreover, the VEGF121-VEGF165 protein exhibited similar potency at inhibiting VEGF165- induced growth of HCT-15 cancer cells. See FIG. 3, panel B. We also found that VEGF121- VEGF165 exhibited similar activity in inhibiting cell proliferation of 3B-11 and HCT-15 cells in an autocrine manner when the plasmid of the fusion gene was transfected into these cells, although the inhibition was not statistically significant (data not shown). The results suggest that VEGF121-VEGF165 inhibit the increase in cell number due to suppression of proliferation, not cytotoxicity. Furthermore, VEGF121-VEGF165 inhibited cell transformation of HCT-15 in an autocrine manner (data not shown). Our results indicate that the VEGF121-VEGF165 recombinant protein can block cell proliferation of 3B-11 as well as cell proliferation and transformation of colon cancer cell HCT-15 in autocrine and paracrine manners.
VEGF121-VEGF165 fusion protein inhibited tube formation induced by VEGF165
Later stages of angiogenesis require morphological alterations of endothelial cells, which result in lumen formation. We examined tube formation in vitro in the presence of the VEGF121-VEGF165 protein. An in vitro tube formation assay was employed by using 3B-11 endothelial cells that were induced to invade a three-dimensional collagen gel where they formed a network of capillary-like tubes. See Zhou et al., Methods 2008, 44(2): 190-195.
The results showed that 3B-11 cells could form a tube network under normal condition, and VEGF165 increases numbers of tube formation. See Fig. 4, panel A. However, the numbers of tube-like structure formation in 3B-11 cells were inhibited by the addition of the VEGF121-VEGF165 protein in a concentration-dependent manner. See Fig. 4, panel A. Furthermore, the VEGF121-VEGF165 protein significantly inhibited VEGFi65-induced tube formation in a paracrine manner (Fig. 4, A) and in an autocrine manner (Fig. 4, B). These results demonstrated that the VEGF121-VEGF165 chimeric protein can inhibit VEGF165- induced angiogenesis in vitro.
VEGF121-VEGF165 fusion protein inhibited cell migration Cell migration is a critical process in angiogenesis and tumor metastasis. We examined whether cell migration was affected by the VEGF121-VEGF165 chimeric protein. Cell migration was examined by a gap-closure migration assay. Consistently, the VEGF121- VEGF165 protein significantly inhibited migration of 3B-11 cells in an autocrine manner. See Fig. 5. In addition, the results showed that cell migration of HCT-15 induced by VEGF165 was inhibited by the addition of the VEGF121-VEGF165 protein in a concentration-dependent manner. See Fig. 6, panel A. The VEGF121-VEGF165 protein significantly inhibited cell migration in a paracrine (Fig. 6, A) and an autocrine manner (Fig. 6, B). These data suggested that VEGF121-VEGF165 chimeric protein can inhibit migration of endothelial cells and tumor cells.
VEGF121-VEGF165 fusion protein impaired tumor invasion
To validate VEGF121-VEGF165 chimeric protein's effect on metastasis of tumor cells, we examined the effect of the protein on cell invasion by using the Transwell assay. In the absence of VEGF121-VEGF165, VEGF165 induced invasive capability as indicated by intensive penetration. See Fig. 7. However, VEGFi65-induced cell invasion was inhibited by the addition of VEGF12i-VEGF165 in a concentration-dependent manner in HCT-15 cancer cells.
The number of penetrated cells was significantly decreased when treated with increased concentrations of the VEGF121-VEGF165 protein as compared with VEGF165 only. See Fig. 7.
These results indicated that the invasion of cancer cells was markedly suppressed by the addition of the VEGF121-VEGF165 protein, suggesting a role of the protein in suppressing cancer metastasis.
VEGF12i-VEGF165 chimeric protein attenuated autocrine VEGFR2-HIF-la- VEGF165/Lon signaling through PI3K-AKT-mTOR pathway
To check whether VEGF121-VEGF165 attenuates HIF-Ια signaling to decrease the resistance to anti- angiogenic therapy, we first examined the effect of VEGF121-VEGF165 on the VEGFR2-HIF-la-VEGFi65 axis in tumor cells. Lon is upregulated by the hypoxia inducible factor- la (HIF-la) and involved in response to low oxygen availability, which adapt cancer cells to a hypoxic environment. We examined whether the fusion protein influenced the expression of Lon protease. The expression of HIF-la, VEGF165, and Lon was determined by Western blot analysis. The results showed that VEGFR2-HIF-la-
VEGFi65/Lon signaling in HCT-15 cancer cells was activated by VEGF165 treatment and hypoxia stimulated by cobalt chloride (C0CI2) (data not shown). However, the signaling activation was inhibited by the addition of VEGF121-VEGF165 in a concentration-dependent manner under normoxia and hypoxia conditions. The recombinant VEGF121-VEGF165 protein reduced the level of HIF-Ια, VEGF165, Lon, and phospho-VEGFR2 induced by VEGF165 and/or hypoxia (data not shown). Mechanically, the VEGF121-VEGF165 protein inhibited the activation of VEGFR2-HIF-la-VEGFi65/Lon signaling through repressing PBK-AKT-mTOR pathway (data not shown), suggesting that the VEGF121-VEGF165 protein overcame survival mechanism triggered by the PI3K-AKT-mTOR to VEGFR2-HIF-la- VEGFi65/Lon axis under hypoxia. These data suggest that VEGF121-VEGF165 can inhibit autocrine VEGFR2-HIF-la-VEGFi65/Lon signaling through PBK-AKT-mTOR pathway in cancer cells.
VEGF121-VEGF165 fusion protein impaired tumor growth in vivo
Our data demonstrated that the chimeric VEGF12i-VEGF165 protein can arrest tube formation of endothelial cells and interfere with tumor cell growth, migration and invasion in vitro. A xenograft tumorigenesis assay was performed to demonstrate the inhibitory effect of the VEGF12i-VEGF165 protein on tumors in vivo. BALB/c nude mice (6-8 weeks old) were used in the assay. lxlO6 HCT-15 cells suspended in 0.2 ml of Matrigel were injected subcutaneously into the dorsal flank of nude mice (1 site per mouse). The mice were examined every two days for tumor formation. Different concentrations of the VEGF121- VEGF165 protein (10, 50, or 250 ng/ml) or a PBS control were directly injected into the tumors in the mice. See Fig. 8, panel B. The body weight of the mice was monitored. See Fig. 8, panel A. The mice were then sacrificed by CO2 euthanasia.
The body weight of the mice treated with the VEGF121-VEGF165 chimeric protein did not change significantly, suggesting that the protein was not toxic to the animals. See Fig. 8, panel A. The chimeric protein reduced tumor growth in the mice in a dose-dependent manner. See Fig. 8, panel B. Thus, the study demonstrated that the VEGF121-VEGF165 chimeric protein can suppress tumor growth in vivo.
Materials and Methods
Cell lines and cell cultures: 3B-11 cells were purchased from ATCC (#CRL-2160, Manassas, VA, USA). 3B-11 cells were maintained in DMEM and HCT-15 cells in RPMI-
1640 supplemented medium with 10% (v/v) heat-inactivated FBS (fetal bovine serum qualified; Invitrogen), 1% PSA (penicillin-streptomycin amphotericin B ; Biological industries, NY, USA) in a 37°C humidified incubator with 5% C02.
Purification of VEGF121-VEGF165 recombinant proteins: The plasmids were transfected into 293T cells using the Biomics transfection reagent as described in the instruction manual of the pcDNA3.1 vector helper- free system (Biomics). After incubation for 36 hours, the supernatant of the 293T cell culture was collected and purified on Ni-NTA resin, eluted with 250 mmol/L imidazole according to the instruction manual. The recombinant protein was concentrated by the Microcon Centrifugal Filter Unit (Millipore, Bedford, MA, USA). Finally, the purified protein was confirmed by 10% SDS-PAGE and Western Blotting.
Cell proliferation assay: The proliferation of 3B-11 cells and HCT-15 cells was assessed using CCK-8 dye reduction assay (Enzo, USA). 3B-11 or HCT-15 cells were pre- treated with different concentrations of VEGF12i-VEGF165 (42, 83, 125 pM) for 30 min, a commercial VEGF165 was added (222 pM, Abeam, Cambridge, MA, USA), and then the cells were incubated for 24 hours. At the end of the treatment, 10 μΐ of the CCK-8 solution was added to each well of the plate and the plate was incubated for 2~4 hours in the incubator. After shaking the plate for 10 seconds, cell viability was assessed by measuring the absorbance at 450 nm using a microplate reader. All measurements were performed three times. The Γ-test was used to compare groups. Data are presented as mean + SD.
Colony formation assay: Clonogenic assay is an in vitro transformation assay based on the ability of a single cell to grow into a colony. To examine this, the plasmid of
VEGF121-VEGF165 was transfected into HCT-15 in a 10 cm dish overnight and treated with VEGF165 recombinant protein 222 pM as a positive control. Next day, the treated HCT-15 cells (~ lxlO3 per well) were plated in 6-well plates and incubated in a 37 °C incubator. Fresh RPMI medium containing 10% FBS was added every 48 hours. At the end of the 14th day, cells were washed twice with ice cold PBS, fixed with methanol for 10 minutes and then stained with 1% crystal violet in methanol for 15 minutes followed by washing with deionized water. Colonies with more than 50 cells were scored and counted under the microscope at 200x.
Cell migration assay: Cell migration assay was determined by gap closure assay. 3B-
11 cells or HCT-15 cells were treated with different concentrations (42, 83, 125 pM) of the recombinant proteins for 16 h (37 °C, 5% CO2). These cells were trypsinized and resuspended in serum- free DMEM or RPMI-1640 medium. A total of 8xl05 cells in 70 μΐ serum-free DMEM or RPMI-1640 were seeded in medium in each well (8xl05 cells/ well) and incubated at 37 °C, 5% CO2. Next day, the ibidi culture-insert (Applied Biophysics, USA) was gently removed by using sterile tweezers, and each well was then filled with 2 ml 0.1% FBS medium. Cell migration was monitored for 48 h by microscope.
Tube formation assay: Corning Matrigel® Matrix (BD Biosciences, San Jose, CA, USA) solution was thawed on ice overnight and 50 μΐ aliquots were coated onto a 96-well plate and incubated at 37°C for lh to solidify. 50 μΐ of DMEM supplemented with 10% FBS medium containing about 8xl05 3B-11 cells was seeded onto the plated Matrigel Matrix and incubated at 37°C. These cells were treated as previously described. The assay was done in triplicate and was incubated at 37 °C with 5% CO2. Images of the formation of capillary-like structures were obtained after 2 h with a computer-assisted microscope (Olympus, Tokyo, Japan) at 200x magnification. Tubular structures were quantified by manually counting the numbers of connected cells in randomly selected fields at 200x magnification. Total tube numbers of network formation were counted.
Cell invasion assay: Cell invasion was evaluated using a transwell chamber (Corning Costar; Cambridge, MA, USA) equipped with a Matrigel-coated filter membrane (8 μιη pores). Briefly, the filters were pre-coated with 200 μg/ml basement membrane proteins (Matrigel; BD Biosciences, San Jose, CA, USA) and allowed to dry overnight at 37°C with 5% C02. HCT-15 cells (8xl05) in FBS-free medium were seeded in the upper chambers, and lower wells were filled with 10% FBS medium. After incubation at 37°C for 48h, non- migratory cells on the upper side of the insert were removed with a cotton swab. The cells that had passed through the filter were fixed in methanol and stained with crystal violet.
Randomly selected fields on the lower side of the photograph under microscopy were counted.
Immunoblotting: HCT-15 cells were seeded onto a 10 mm dish at a density of 1.5xl06 cells in 10 ml medium for 24 h under normoxia and hypoxia (C0CI2, Cobalt dichloride; 150 μΜ) and treated as previously described. Total protein concentrations were determined using a BCA protein assay. Equal quantities of total protein were resolved using 10% SDS-PAGE and electroblotted onto polyvinylidene fluoride membranes. Membranes were blocked with
5% skimmed milk and probed overnight at 4°C with primary antibodies. Membranes were then probed with the appropriate HRP-conjugated secondary antibodies (GeneTex, Hsinchu, Taiwan) and the immunoreactive bands were visualized using an enhanced chemiluminescence method (Bio-Rad, Hercules, CA, USA). Antibodies used in this study were purchased or produced as indicated. Antibody to human Lon was produced as described previously. See Wang et al., Cancer Sci 2010, 101(12):2612-2620; and Cheng et al., Cell death & disease 2013, 4, e681. Phospho-PI3K (Tyr458/ Tyrl99, #4228), phospho- AKT (Ser473, #4060), and phoshpo-mTOR (Ser2448, #2971) antibodies were obtained from Cell Signaling Technology (Beverly, MA, USA); HIF-la (#610958) antibodies was obtained from BD Biosciences (Franklin Lakes, NJ); phospho-VEGFR2 (Tyrl054/Tyrl059, ab5473), VEGF-165A (ab69479) antibodies were from Abeam (Cambridge, MA, USA); beta-actin antibody was fromfrom GeneTex (GTX109639, Hsinchu, Taiwan).
Statistical methods: Parametric Student's t test was used in this study to judge the significance of difference between conditions of interest. In general, a P value of <0.05 was considered as statistically significant (Student's t test, *p < 0.05, **p < 0.01, and ***p < 0.001). OTHER EMBODIMENTS
All of the features disclosed in this specification may be combined in any
combination. Each feature disclosed in this specification may be replaced by an alternative feature serving the same, equivalent, or similar purpose. Thus, unless expressly stated otherwise, each feature disclosed is only an example of a generic series of equivalent or similar features.
From the above description, one skilled in the art can easily ascertain the essential characteristics of the described embodiments, and without departing from the spirit and scope thereof, can make various changes and modifications of the embodiments to adapt it to various usages and conditions. Thus, other embodiments are also within the claims.

Claims

WHAT IS CLAIMED IS:
1. A fusion protein, comprising:
(i) a first vascular endothelial growth factor (VEGF) isoform, and
(ii) a second VEGF isoform, and
(iii) a dimerization domain between the first isoform and the second isoform.
wherein the first isoform and the second isoform are selected from VEGFm and VEGF165.
2. The fusion protein of claim 1, wherein the dimerization domain contains two Fc regions.
3. The fusion protein of claim 2, further comprising a linker between the two Fc regions.
4. The fusion protein of claim 3, wherein the fusion protein contains, in the direction from the N-terminus to the C-terminus, VEGF121, one of the two Fc regions, the linker, the other of the two Fc regions, and VEGF165.
5. The fusion protein of claim 3, wherein the fusion protein contains, in the direction from the N-terminus to the C-terminus, VEGF165, one of the two Fc regions, the linker, the other of the two Fc regions, and VEGF121.
6. The fusion protein of any of claims 3-5, wherein the linker is a flexible linker consisting of 15 to 30 amino acids.
7. The fusion protein of claim 6, wherein the linker is (Gly-Gly-Gly-Gly-Ser)ni wherein n is 3.
8. The fusion protein of claim 7, wherein each of the Fc regions is a human IgGl Fc region.
9. The fusion protein of claim 8, further comprising a peptide tag.
10. The fusion protein of claim 9, wherein the tag is a C-terminal 6X-His tag.
11. The fusion protein of claim 10, further comprising an N-terminal signal peptide.
12. The fusion protein of claim 11, wherein the fusion protein has an amino acid sequence that is at least 90% identical to the sequence of SEQ ID NO: 11.
13. The fusion protein of claim 12, wherein the fusion protein has the sequence of SEQ ID NO: 11.
14. A nucleic acid molecule, comprising a nucleic acid sequence that encodes the fusion protein of any of claims 1-13.
15. A host cell, comprising the nucleic acid molecule of claim 14.
16. A pharmaceutical composition, comprising the fusion protein of any of claims 1-13 and a pharmaceutically acceptable carrier.
17. A method of treating a cancer in a subject, the method comprising administering to a subject in need thereof the composition of claim 16.
PCT/US2016/064554 2015-12-03 2016-12-02 Heterodimeric vascular endothelial growth factor and use thereof WO2017096124A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2018548648A JP6936808B2 (en) 2015-12-03 2016-12-02 Fusion proteins, nucleic acid molecules, host cells, pharmaceutical compositions, and how to use the pharmaceutical compositions
US15/780,734 US10851142B2 (en) 2015-12-03 2016-12-02 Heterodimeric vascular endothelial growth factor and use thereof
CN201680071128.4A CN108699562B (en) 2015-12-03 2016-12-02 Hetero-dimer type vascular endothelial growth factor and application thereof
EP16871555.5A EP3384029B1 (en) 2015-12-03 2016-12-02 Heterodimeric vascular endothelial growth factor and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562262630P 2015-12-03 2015-12-03
US62/262,630 2015-12-03

Publications (1)

Publication Number Publication Date
WO2017096124A1 true WO2017096124A1 (en) 2017-06-08

Family

ID=58797876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/064554 WO2017096124A1 (en) 2015-12-03 2016-12-02 Heterodimeric vascular endothelial growth factor and use thereof

Country Status (6)

Country Link
US (1) US10851142B2 (en)
EP (1) EP3384029B1 (en)
JP (1) JP6936808B2 (en)
CN (1) CN108699562B (en)
TW (1) TWI746491B (en)
WO (1) WO2017096124A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019173334A1 (en) * 2018-03-05 2019-09-12 The Schepens Eye Research Institute, Inc. Engineered vegf variants for retinal neuroprotection, promotion of axon growth and axon regeneration

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030027751A1 (en) * 2001-04-10 2003-02-06 Genvec, Inc. VEGF fusion proteins
US20090252729A1 (en) * 2007-05-14 2009-10-08 Farrington Graham K Single-chain Fc (scFc) regions, binding polypeptides comprising same, and methods related thereto

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002081520A2 (en) * 2001-04-06 2002-10-17 Maxygen Holdings Ltd. Single chain dimeric polypeptides derived from the vegf family
CN101070349B (en) * 2007-05-22 2010-10-13 山西康宝生物制品股份有限公司 Fusion protein with function of selective killing endothelial cells in tumor neogenetic blood vessels and use thereof
CL2008002782A1 (en) * 2007-09-21 2009-07-31 Genentech Inc Neutralizing anti-bv8 antibody; composition comprising it; and its use to treat tumors in humans previously treated with a vascular endothelial growth factor antagonist.
CN101469024A (en) * 2007-12-27 2009-07-01 上海汇康生物科技有限公司 Calreticulin- VEGFR2 fusion protein and use
WO2010083495A2 (en) * 2009-01-18 2010-07-22 The Board Of Trustees Of The Leland Stanford Junior University Polypeptides targeting vascular endothelial growth factor receptor-2 and alpha v beta 3 integrin
CN101935351A (en) * 2010-01-14 2011-01-05 中国药科大学 Human vascuoar endothelial cell growth factor analogue
CN102924604A (en) * 2012-11-01 2013-02-13 中国人民解放军军事医学科学院生物工程研究所 Recombined vessel endothelium growth factor receptor, encoding genes thereof and application

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030027751A1 (en) * 2001-04-10 2003-02-06 Genvec, Inc. VEGF fusion proteins
US20090252729A1 (en) * 2007-05-14 2009-10-08 Farrington Graham K Single-chain Fc (scFc) regions, binding polypeptides comprising same, and methods related thereto

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BOESEN, TP ET AL.: "Single-chain Vascular Endothelial Growth Factor Variant with Antagonist Activity.", THE JOURNAL OF BIOLOGICAL CHEMISTRY ., vol. 277, no. 43, 31 July 2002 (2002-07-31), pages 40335 - 40341, XP 055283795 *
CHENG ET AL., CELL DEATH & DISEASE, vol. 4, 2013, pages e681
SIEMEISTER, G ET AL.: "An Antagonistic Vascular Endothelial Growth Factor (VEGF) Variant Inhibits VEGF-Stimulated Receptor Autophosphorylation and Proliferation of Human Endothelial Cells.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, vol. 95, no. 8, 14 April 1998 (1998-04-14), pages 4625 - 4629, XP 000876737 *
SIEMEISTER, G ET AL.: "The Alpha-Helical Domain Near the Amino Terminus Is Essential for Dimerization of Vascular Endothelial Growth Factor.", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 273, no. 18, 1 May 1998 (1998-05-01), pages 11115 - 11120, XP 055387988 *
TSAI, JL ET AL.: "The Novel VEGF121-VEGF165 Fusion Attenuates Angiogenesis and Drug Resistance via Targeting VEGFR2-HIF-1a-VEGF165/Lon Signaling Through PI3K-AKT-mTOR Pathway.", CURRENT CANCER DRUG TARGETS., vol. 16, no. 3, 23 February 2016 (2016-02-23), pages 1 - 13, XP055546292 *
WANG ET AL., CANCER SCI, vol. 101, no. 12, 2010, pages 2612 - 2620
ZHOU ET AL., METHODS, vol. 44, no. 2, 2008, pages 190 - 195

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019173334A1 (en) * 2018-03-05 2019-09-12 The Schepens Eye Research Institute, Inc. Engineered vegf variants for retinal neuroprotection, promotion of axon growth and axon regeneration

Also Published As

Publication number Publication date
CN108699562A (en) 2018-10-23
JP6936808B2 (en) 2021-09-22
EP3384029A4 (en) 2019-04-17
JP2019500900A (en) 2019-01-17
US20180355004A1 (en) 2018-12-13
EP3384029B1 (en) 2022-01-05
TW201726715A (en) 2017-08-01
US10851142B2 (en) 2020-12-01
TWI746491B (en) 2021-11-21
CN108699562B (en) 2022-04-22
EP3384029A1 (en) 2018-10-10

Similar Documents

Publication Publication Date Title
JP6018648B2 (en) Method of using chimeric coiled-coil (double-coil) molecules
KR20140065352A (en) Bispecific antibody targeting vegfr-2 and dll4, and pharmaceutical composition comprising thereof
KR101397088B1 (en) A fusion protein for inhibiting both angiogenesis and tumor cell proliferation, and composition comprising the same
JP2013502421A (en) Method of using CD44 fusion protein for cancer treatment
ES2647568T3 (en) Cancer treatment
US20170306006A1 (en) Human notch1 decoys
US9504738B2 (en) Immunogenic epitopes, peptidomimetics, and anti-peptide antibodies, and methods of their use
Tsai et al. The novel VEGF121-VEGF165 fusion attenuates angiogenesis and drug resistance via targeting VEGFR2-HIF-1α-VEGF165/Lon signaling through PI3K-AKT-mTOR pathway
Yang et al. Modification of IL-24 by tumor penetrating peptide iRGD enhanced its antitumor efficacy against non-small cell lung cancer
US10851142B2 (en) Heterodimeric vascular endothelial growth factor and use thereof
WO2014191608A1 (en) Peptides derived from connexin 43, and pharmaceutical composition for the treatment of cancer
CN109504698A (en) A kind of method and purposes of the Chimeric antigen receptor targeting full humanization mesothelin
US11192920B2 (en) Peptides for binding to CD44v6 and use thereof
Dortet et al. CD44-independent activation of the Met signaling pathway by HGF and InlB
US20220378946A1 (en) Cell penetrating peptides and uses thereof
CN110627905B (en) VEGF (vascular endothelial growth factor) -EGFR (epidermal growth factor receptor) -targeted bifunctional fusion protein and application thereof
WO2012130141A1 (en) Method and composition for enhancing target cells uptake of therapeutic agents
CN109790225B (en) Chimeric proteins for targeting dsRNA
ES2940360T3 (en) Polypeptides capable of inhibiting the binding between leptin and neuropilin-1
CN109504697A (en) A kind of method and purposes of the Chimeric antigen receptor of targeting humanized mesothelin
WO2023139156A1 (en) Peptides and pharmaceutical composition for the treatment of tumours
CN115786270A (en) Engineered macrophages and their use in the treatment of fibrotic diseases
KR20130134628A (en) Composition for prevention and treatment of influenza a viral diseases
CN112063587A (en) Application of super antigen or variant thereof in preparation of CAR immune cells
Moss Engineering Novel Angiopoietin Receptor Ligands

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16871555

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2018548648

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2016871555

Country of ref document: EP