WO2017147820A1 - Dual-targeting drug carrier - Google Patents

Dual-targeting drug carrier Download PDF

Info

Publication number
WO2017147820A1
WO2017147820A1 PCT/CN2016/075329 CN2016075329W WO2017147820A1 WO 2017147820 A1 WO2017147820 A1 WO 2017147820A1 CN 2016075329 W CN2016075329 W CN 2016075329W WO 2017147820 A1 WO2017147820 A1 WO 2017147820A1
Authority
WO
WIPO (PCT)
Prior art keywords
target
vegf
rgd
egf
dual
Prior art date
Application number
PCT/CN2016/075329
Other languages
French (fr)
Chinese (zh)
Inventor
张正
蓝耿立
王信二
张顺福
李佳哲
詹佩嘉
Original Assignee
法玛科技顾问股份有限公司
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 法玛科技顾问股份有限公司 filed Critical 法玛科技顾问股份有限公司
Priority to PCT/CN2016/075329 priority Critical patent/WO2017147820A1/en
Priority to CN201680083086.6A priority patent/CN108699164B/en
Publication of WO2017147820A1 publication Critical patent/WO2017147820A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes

Definitions

  • the present invention relates to a novel target drug carrier, and in particular to a dual target drug carrier having two target molecules.
  • Cancer is the leading cause of death in the United States, and cancer mortality continues to increase. Cancer is the inability of cells to divide, grow and differentiate normally. The initial clinical manifestations of cancer are extremely uneven, with almost 70 cancers occurring in human organs and tissues, and some of these similar cancer types belong to many different molecular diseases. Unfortunately, some cancers may have no actual symptoms until late in the course of the disease, making treatment and prognosis extremely difficult.
  • Cancer treatment usually includes surgery, chemotherapy, and/or radiation therapy. All current therapies have serious side effects and reduce the quality of life. Most chemotherapeutic drugs act on both normal and cancerous tissues. Therefore, one of the challenges in treating cancerous tumors is to give cancer cells maximum killing while minimizing damage to healthy tissue. Depending on the drug (eg, intravenous) and nature of the route of administration (eg, its physical and pharmacokinetic properties), typically only a small fraction of the administered dose reaches the target cell, with the remainder acting on other tissues or rapidly disappearing.
  • drug eg, intravenous
  • nature of the route of administration eg, its physical and pharmacokinetic properties
  • Tumor neovascular endothelial cells express a large number of specific membrane proteins, including ⁇ v ⁇ 3, ⁇ v ⁇ 5, integrin, and vascular-related growth factors.
  • RGD short-chain peptides
  • tumor cells are highly unstable and variability, and are resistant to drugs during treatment. At present, various drugs and diagnostic agents are still slow and ineffective in improving the survival of cancer patients.
  • the present invention provides a novel vector that can be used to diagnose and treat cancer.
  • the dual target drug carrier of the invention is a fusion protein platform, which can effectively reduce the threshold and cost of the pharmaceutical technology.
  • the invention provides a dual target drug carrier comprising a first target molecule and a second target molecule.
  • the first or second target molecule specifically binds to vascular endothelial cells in tumor cells or/and tumor microenvironment
  • the first and second target molecules include, but are not limited to, arginine-glycine-aspartate (RGD), asparagine-glycine-arginine (NGR) ), cyclic NGR (cyclic asparagine-glycine-arginine), internalization of RGD (iRGD, internalization-arginine-glycine-aspartate), cystine-glycine-aspartate Acid-lysine-arginine-threonine-arginine-glycine-alanine (CGNKRTRGA), stomach Gastrin, bombesin, octreotide or a derivative thereof.
  • RGD arginine-glycine-aspartate
  • NGR asparagine-glycine-arginine
  • cyclic NGR cyclic asparagine-glycine-arginine
  • internalization of RGD iRGD, internalization-arginine-glycine-aspartate
  • Macromolecular peptides include, but are not limited to, epidermal growth factor (EGF), anti-EGFR (epidermal growth factor receptor) antibody, vascular endothelial growth factor (VEGF), anti-VEGFR ( Vascular Endothelial Growth Factor Receptor, anti-HER2 (human epidermal growth factor receptor 2) antibody, hepatocyte growth factor receptor (HGFR), anti-HIV - HGFR antibody, tumor necrosis factor (TNF) or anti-TNF antibody.
  • EGF epidermal growth factor
  • VEGF vascular endothelial growth factor
  • anti-HER2 human epidermal growth factor receptor 2
  • HGFR hepatocyte growth factor receptor
  • TNF tumor necrosis factor
  • the first target molecule is linked to the second target molecule by a linker.
  • the connector is a peptide having 5 to 20 amino acids.
  • the linker comprises, but is not limited to, GG, PGGGG or GGGGSGGGGS, wherein G represents glycine, P represents proline, and S represents serine.
  • a radioisotope is further included.
  • the invention further provides a pharmaceutical composition comprising the above dual target pharmaceutical carrier and a pharmaceutically acceptable carrier.
  • a liposome is further included.
  • nanoparticles are further included.
  • Figure 1A shows purified VEGF, RGD-VEGF, RGD4C-VEGF, EGF, SDS-PAGE electropherogram of RGD-EGF and RGD4C-EGF protein.
  • Figure 1B is a SDS-PAGE electrophoresis map of marker, VEGF, RGD-VEGF, and RGD4C-VEGF, wherein M is marker, 1 is VEGF (15.3 Da), 2 is RGD-VEGF (15.6 Da), and 3 is RGD4C-VEGF ( 16.3Da).
  • FIGS. 2A-2F are binding curves/bar graphs of RGD-VEGF and RGD4C-VEGF and ⁇ v ⁇ 3, VEGFR1, VEGFR2.
  • RGD-EGF and RGD4C-EGF bind to ⁇ v ⁇ 3 and EGFR, respectively.
  • Figure 2A is a graph showing the binding of RGD-VEGF and RGD4C-VEGF to ⁇ v ⁇ 3.
  • Figure 2B is a bar graph of competition of RGDfV polypeptide with RGD-VEGF and RGD4C-VEGF in combination with ⁇ v ⁇ 3.
  • Figure 2C is a graph showing the binding of RGD-VEGF and RGD4C-VEGF to VEGFR1.
  • Figure 2D is a graph showing the binding of RGD-VEGF and RGD4C-VEGF to VEGFR2.
  • Figure 2E is a bar graph of competition of VEGFR2 antibodies by RGD-VEGF and RGD4C-VEGF in combination with VEGFR2.
  • Figure 2F is a graph showing the binding of RGD-EGF and RGD4C-EGF to ⁇ v ⁇ 3.
  • Figure 3A is a graph showing the binding of U87MG (expressing ⁇ v ⁇ 3 and EGFR) cells to RGD-VEGF and RGD4C-VEGF.
  • RGD-VEGF and RGD4C-VEGF can bind to ⁇ v ⁇ 3 and EGFR, respectively.
  • Figure 3B is a bar graph of binding of U87MG (expressing ⁇ v ⁇ 3 and EGFR) cells to RGD-VEGF and RGD4C-VEGF.
  • Figure 3C is a graph showing the binding of HUVEC (expressing VEGFR1, VEGFR2 and ⁇ v ⁇ 3) cells to RGD-VEGF, RGD4C-VEGF and NGR-VEGF.
  • RGD-VEGF, RGD4C-VEGF and NGR-VEGF can bind to express VEGFR1 Cells of VEGFR2 and ⁇ v ⁇ 3.
  • Figure 3D is a bar graph of binding of HUVEC (expressing VEGFR1, VEGFR2 and ⁇ v ⁇ 3) cells to RGD-VEGF, RGD4C-VEGF and NGR-VEGF.
  • Figures 4A-4D are cell adhesion assays. After coating RGD-VEGF and RGD4C-VEGF on the cell plate, the number of U87MG cells and HUVEC cells attached to the culture plate can be increased. If a large amount of RGD peptide chain is added to compete, the adsorption of cells can be reduced. 4A shows that RGD-VEGF and RGD4C-VEGF are applied to the cell plate to increase the number of U87MG cells attached to the culture plate, and FIG. 4B shows the amount of cells attached to the culture plate after the addition of the RGDfV polypeptide.
  • Figure 4C-4D shows that RGD-VEGF and RGD4C-VEGF bind to HUVEC cells, and then compete with RGDfV polypeptide or VEGFR2 recombinant protein, respectively, in terms of absorbance and percentage.
  • Figures 5A-5B show that RGD-EGF and RGD4C-EGF or RGD-VEGF and RGD4C-VEGF activate a signaling pathway downstream of EGFR or VEGFR, respectively.
  • the EGF or VEGF of the dual target drug carrier of the invention retains its original biological properties.
  • M of Figure 5A is Marker
  • C is the control group (no protein added)
  • E is EGF (25 nM)
  • RE is RGD-EGF (25 nM)
  • R4C-E is RGD4C-EGF (25 nM)
  • C of Figure 5B For the control group (no protein added), RV is RGD-VEGF, R4-V RGD4C-VEGF.
  • Figure 6 is a graph showing the stability of a radiolabeled fusion protein in a buffer solution (HEPES, 4 ° C) and serum (37 ° C).
  • the radiolabeled fusion protein was stored in Hepes buffer (4 ° C) and fetal bovine serum (37 ° C), and the radiochemical purity was still higher than 90% after 24 hours, indicating the radioactive In-111-labeled fusion protein.
  • the serum stability is excellent.
  • Fig. 7 is a fluorescent photograph.
  • the RGD-VEGF and RGD4C-VEGF marker recombinant proteins can bind to cells.
  • Figure 8 is a single photon and computed tomography (SPECT/CT) angiogram of radiolabeled recombinant protein.
  • U87MG subcutaneous tumor mice were injected with 111 In-DTPA (Diethylene triamine pentacetate aci)-EGF, 111 In-DTPA-RGD-EGF and 111 In-DTPA-RGD4C-EGF.
  • Small animal single photon and computed tomography (SPECT/CT) angiography was performed at 4, 8 and 24 hours.
  • SPECT/CT computed tomography
  • the image shows that after the In-111 marker recombinant protein is applied, the specific accumulation of the tumor in the tumor (expressed as the tumor/muscle accumulation ratio) increases with time, reaching a peak at 8 hours after the drug injection, at this time 111 In
  • the tumor/muscle accumulation ratio of -DTPA-RGD4C-EGF was 4.4, slightly higher than 3.6 of 111 In-DTPA-RGD-EGF, and much higher than 1.7 of 111 In-DTPA-EGF.
  • the invention provides a dual target drug carrier comprising a first target molecule and a second target molecule.
  • the target molecule of the present invention is a peptide, antibody or protein.
  • the target molecules of the invention can be chemically modified to increase stability.
  • Chemically modified peptides or peptide analogs include any chemically functional equivalent that increases stability and/or potency either in vivo or in vitro.
  • a peptide analog refers to an amino acid derivative of any peptide.
  • Peptide analogs include, but are not limited to, modified branches upon synthesis, combined with unsaturated amino acids and/or derivatives thereof, cross-linking and other methods that increase the peptide identity.
  • a branched modification includes a modification of an amine group.
  • the target molecule can bind to the lesion tissue in vivo and in vitro, especially for vascular endothelial cells of tumor, cancer tissue/cell and tumor microenvironment. Therefore, when the target peptide is conjugated to a reporter molecule (bioimaging fluorescent molecule or radiation), the tumor target peptide can directly display the tumor location and contribute to the diagnosis of cancer.
  • Conjugation as used in the present invention means that two molecules that contribute to the treatment/diagnosis of a tumor, such as a tumor target peptide and a reporter molecule, are bound by sufficient affinity. Conjugation can be accomplished by covalent, non-covalent bonds or other forms of bonding, for example, coating.
  • the target molecule of the present invention may be a small molecule peptide and/or a macropeptide.
  • Small molecule peptides include, but are not limited to, lysine-glycine-aspartate (RGD), asparagine-glycine-arginine (NGR), cyclic NGR, internalization RGD (iRGD), cystine - glycine-aspartate-lysine-arginine-threonine-arginine-glycine-alanine (CGNKRTRGA), gastrin, bombesin, octreotide Octreotide) or a derivative thereof.
  • RGD lysine-glycine-aspartate
  • NGR asparagine-glycine-arginine
  • iRGD internalization RGD
  • gastrin bombe
  • Macromolecular peptides include, but are not limited to, epidermal growth factor (EGF), anti-EGFR antibodies, vascular endothelial growth factor (VEGF), anti-VEGFR antibodies, anti-HER2 antibodies, hepatocyte growth factor receptor (HGFR), Anti-HGFR antibody, tumor necrosis factor (TNF) or anti-TNF antibody.
  • EGF epidermal growth factor
  • VEGF vascular endothelial growth factor
  • HGFR hepatocyte growth factor receptor
  • TNF tumor necrosis factor
  • the target molecule can form a conjugate with an imaging substance or a radiotherapeutic substance (radiopharmaceutical or isotope).
  • Radioactive imaging materials include, but are not limited to, 122 I, 123 I, 124 I, 125 I, 131 I, 18 F, 75 Br, 76 Br, 76 Br, 77 Br, 211 At, 225 Ac, 177 Lu, 153 Sm , 186 Re, 188 Re, 67 Cu, 213 Bi, 212 Bi, 212 Pb and 67 Ga.
  • Radiopharmaceuticals include, but are not limited to, 111 In hydroxyquinoline, 131 I sodium iodide, 99m Tc phenyl bromide, 99m Tc red blood cells, 123 I sodium iodide, 99m Tc Exametazime, 99m Tc large particle polymeric albumin, 99m Tc-methylene phosphonate, 99m Tc Mertiatide, 99m Tc hydroxymethylene diphosphonate, 99m Tc triamine pentaacetic acid, 99m Tc Pertechnetate, 99m TcSestamibi, 99m Tc sulphur colloid, 99m Tc Tetrofosmin, 201 Tl and 133 Xe .
  • Radiation isotopes include, but are not limited to, 52 Fe, 52 mMn, 55 Co, 64 Cu, 67 Ga, 68 Ga, 99 mTc, 111 In, 123 I, 125 I, 131 I, 32 P, 47 Sc, 67 Cu, 90 Y, 109 Pd, 111 Ag, 149 Pm, 186 Re, 188 Re, 211 At, 212 Bi, 213 Bi, 105 Rh, 153 Sm, 177 Lu and 198 Au.
  • Non-gastrointestinal as used in the present invention includes subcutaneous, intradermal, intravenous, intramuscular, articular, arterial, synovial, focal or intracranial injections.
  • the "linker” as used in the present invention refers to a sequence of 2 to 50 synthetic amino acids, preferably 5 to 20 synthetic amino acids, which are linked to two peptide sequences, for example, to link two peptide structures.
  • the connector of the present invention can link two amino acid sequences through a peptide bond.
  • the linker peptide of the present invention is a linear sequence linking the first portion to the second portion.
  • the replaceable connector may be (GGGGS)n, (G)n, (EAAAK)n, (XP)n or (PAPAP)n, eg, GG, GGSG, GGGS, GGGGS, GGSGG, GGGSGG, GGGSGGG, GGGSGGGS, GGGSGGGGS, ASGG, GGGSASGG, SGCGS, GGGGSGGGG, GGSHG, SGCGGGS, and AACAA, wherein said G represents glycine; said S represents serine; said E represents glutamic acid; said A represents alanine; K represents lysine; P represents valine; said H represents histidine; and C represents cystine.
  • the dual target drug carrier of the present invention can carry a tumor therapeutic drug or It is a substance (such as a therapeutic radionuclide) that becomes a tumor target drug carrier.
  • a drug capable of molecular imaging can also be carried as a molecular imaging probe for tumor diagnosis.
  • the double target developer carrier of the invention has the effect of tumor diagnosis.
  • EGF, RGD-EGF, RGD4C-EGF, VEGF, RGD-VEGF and RGD4C-VEGF genes were constructed in the expression vector of pET28a(+) with Nco I and Xho I restriction enzyme cleavage sites.
  • the C-terminus (ie, the carbon terminus) of RGD and RGD4C is ligated to the N-terminus (ie, the nitrogen terminus) of EGF or VEGF, and the C-terminus of EGF or VEGF has a His 6 flag.
  • the His 6 marker facilitates subsequent purification and analysis, wherein the gene sequences of RGD-EGF, RGD4C-EGF, RGD-VEGF, and RGD4C-VEGF comprise the linker GG.
  • the constructed pET28a(+) EGF, RGD-EGF, RGD4C-EGF, VEGF, RGD-VEGF and RGD4C-VEGF expression vectors were transfected into E. coli.DH5 ⁇ to produce and preserve vector DNA. The sequence was confirmed by DNA sequencing.
  • Example 1 The vector of Example 1 was sent to E. coli BL21 (DE3) by heat shock, and screened and confirmed by kanamycin (50 ⁇ g/mL).
  • the E. coli BL21 (DE3) strain carrying the protein expression vector was induced with 1 mM IPTG for 16 hours at 37 °C.
  • the bacterial solution was sterilized by a French press at a pressure of 30 PSI, centrifuged at 13,000 g for 30 minutes, and samples of the supernatant and the pellet were collected for SDS-PAGE electrophoresis analysis, and observed by SDS-PAGE. Performance to the target fusion protein. If it is in the form of an insoluble body, the bacteria are present in the form of an insoluble body.
  • Subsequent protein production receives a pellet after the disruption for protein purification.
  • the lysed target fusion protein was filtered through a 0.45 ⁇ m filter, followed by a 1:50 ratio of refolding buffer (20 mM Tris, 0.1 Mm GSSG, 1 mM GSH, 1 mM EDTA pH 8.0). Dilution refolding.
  • the target protein sample was slowly added to the refolding buffer, and the concentration of the target fusion protein after dilution was not more than 0.2 mg/ml, and the renaturation was carried out at 4 ° C for 12 hours, and then filtered through a 0.45 ⁇ m filter to remove the precipitate.
  • Target protein was carried out at 4 ° C for 12 hours, and then filtered through a 0.45 ⁇ m filter to remove the precipitate.
  • the protein refolding solution was passed through a nickel column at a flow rate of 5 mL/min.
  • the column was first washed with buffer A (20 mM Tris (pH 8.0), 500 mM NaCl, 20 mM imidazole, 0.5 mM PMSF), followed by washing, using buffer A and buffer B (20 mM Tris (pH 8.0), The ratio of the imidazole of the solution was adjusted in proportion to 500 mM NaCl, 500 mM imidazole, 0.5 mM PMSF) to wash out the target fusion protein. Since the collected target fusion protein still has other impurity proteins, the fusion protein was isolated using a gel column (HiPrap 26/60S-100).
  • EGF EGF
  • RGD-EGF NGR-EGF
  • RGD4C-EGF VEGF
  • RGD-VEGF RGD-VEGF
  • NGR-VEGF NGR-VEGF
  • RGD4C-VEGF target fusion proteins target fusion proteins to cells was analyzed by ELISA cell binding assay.
  • 25 ⁇ g/well of integrin ( ⁇ v ⁇ 3) receptor dissolved in PBS (calcium and magnesium ions) was placed in a 96well ELISA microplate, and cultured at 4 ° C for 12 hours, and then reacted with 3% BSA at 25 ° C for 1 hour.
  • the serially diluted target fusion protein was added to culture at 25 ° C for 2 hours, and after washing with PBS buffer (containing calcium and magnesium ions), the mouse anti-His-HRP antibody was used to detect the binding of the protein to the receptor, and the wavelength was measured. The absorbance at 450 nm is used to quantify the amount of protein bound to the receptor.
  • the obtained data using the prism software for nonlinear curve fitting can obtain the dissociation constant K d value of protein binding, which can be used as a comparison of the binding ability of the protein to different receptors.
  • the mouse anti-His-HRP antibody was used to detect binding of the protein to the receptor, and the absorbance at a wavelength of 450 nm was measured to quantify the amount of protein bound to the receptor.
  • the obtained data using the prism software for nonlinear curve fitting can obtain the dissociation constant Kd value of protein binding, which can be used as a comparison of the binding ability of the protein to different receptors.
  • the dissociation constant K d values of EGF, RGD-EGF, RGD4C-EGF and EGFR were 2.38, 20.35 and 14.33 nM, respectively.
  • the dissociation constant K d values of VEGF, RGD-VEGF, RGD4C-VEGF and Integrin were 0, 5.0 and 1.0, respectively.
  • VEGF, RGD-VEGF, VEGFR1 solution RGD4C-VEGF with a dissociation constant K d values of 15.3,11.3 and 11.4.
  • the dissociation constant K d values of VEGF, RGD-VEGF, RGD4C-VEGF and VEGFR2 were 22.2, 12.1 and 13.9, respectively.
  • various dual-target fusion proteins can bind to more than two receptors, with the ability to dual target.
  • RGD-EGF and RGD4C-EGF can bind to Integrin ( ⁇ v ⁇ 3) and EGFR, respectively, and RGD-VEGF and RGD4C-VEGF can bind to Integrin ( ⁇ v ⁇ 3), VEGFR1 and VEGFR2, respectively.
  • This example uses MDA-MB468 (highly expressed EGFR), MDA-MB231 (moderately expressed EGFR), MCF-7 (low performance), HT1080 (expressed APN), U87MG (expressed ⁇ v ⁇ 3) and HUVEC (expressed VEGFR and ⁇ v ⁇ 3) Cell lines to analyze the specific binding of the dual-target fusion protein to different receptors.
  • MDA-MB468, MDA-MB231, MCF-7, HT1080, U87MG and HUVEC cells were seeded in a 96-well microplate at a dose of 20,000 cells/well, and cultured at 37 ° C for 12 hours. After the cells were attached, they were fixed with 4% paraformaldehyde hyde precooled at 4 ° C, and then incubated at room temperature for 15 minutes. After the fixed cells were incubated with 3% FBS at 25 ° C for 1 hour, serially diluted target fusion proteins were added and incubated at 25 ° C for 1 hour.
  • the mouse anti-His-HRP antibody was used to detect the binding of the protein to the cells, and then the TMP of the HRP was used for color measurement.
  • the absorbance at a wavelength of 450 nm was used for quantify the amount of protein bound to the cells.
  • VEGF, RGD-VEGF, RGD4C- VEGF, NGR-VEGF and VEGFR intergrin solution on HUVEC cells with a dissociation constant K d values of 12.7,11.9,6.4 and 9.4nM.
  • RGD-EGF, RGD4C-EGF, RGD-VEGF, and RGD4C-VEGF can bind to cells expressing the corresponding receptor.
  • This example uses an ECM cell adhesion analysis system to analyze the adhesion characteristics of cells.
  • Different concentrations of the target fusion protein were coated per well in a 96-well microplate and incubated at 16 ° C for 12 hours. After the protein was attached, 3% BSA was added and incubated at 25 ° C for 1 hour.
  • Tumor cells U87MG, 5 ⁇ 10 5 cells/well
  • Tumor cells that had been starved for one hour were added to a 96-well microplate, and after incubation at 37 ° C for 2 hours, they were washed with D-PBS to remove cells not bound to the protein.
  • MTT was added and incubated at 37 ° C for 1 hour.
  • the surviving cells metabolized the MTT agent to formazan blue-violet crystals.
  • the wax was dissolved in DMSO to detect the absorbance at a wavelength of 570 nm.
  • the cell attachment characteristics were calculated by the following formula.
  • FIGS 4A-D RGD-VEGF and RGD4C-VEGF were applied to the cell disk After that, the number of U87MG cells and HUVEC cells attached to the culture plate can be increased, and if a large amount of RGD peptide chain is added to compete, the adsorption of cells can be reduced.
  • Figure 4A shows that RGD-VEGF and RGD4C-VEGF are coated on the cell plate to increase the amount of U87MG cells attached to the culture plate
  • Figure 4B shows the amount of cells attached to the plate after the addition of the RGDfV polypeptide.
  • Figure 4C-4D shows that RGD-VEGF and RGD4C-VEGF bind to HUVEC cells, and then compete with RGDfV polypeptide or VEGFR2 recombinant protein, respectively, in terms of absorbance and percentage.
  • the fusion protein activates the cellular receptor and its downstream signaling molecule phosphorylation.
  • Various human tumor cells were cultured with 25nM target fusion protein at 37 ° C for 60 minutes to collect intracellular protein extracts, and the phosphorylation reaction of cell receptors and their downstream signal molecules was analyzed by Western blotting method.
  • RGD-EGF and RGD4C-EGF were co-cultured with MDA-MB468 cells expressing EGFR; or RGD-VEGF and RGD4C-VEGF were co-cultured with HUVEC vascular endothelial cells expressing VEGFR1 and VEGFR2.
  • the signaling molecules downstream of EGFR (Fig. 5A) or VEGFR (Fig. 5B) were analyzed.
  • RGD-EGF and RGD4C-EGF or RGD-VEGF and RGD4C-VEGF can activate the signaling pathway downstream of EGFR or VEGFR, respectively.
  • the results show that the double target molecule of the present invention not only has the ability to bind to a relatively biomarker molecule, but a large peptide molecule such as EGF or VEGF retains its original biological properties.
  • p-SCN-Bn-DTPA 2-(4-Iso thiocyanatobenzyl-diethylenetriaminepentaacetic acid, p-SCN-Bn-DTPA) is dissolved in HEPES buffer solution, and RGD-EGF is added.
  • RGD4C-EGF was reacted at room temperature for 1 hour.
  • the unreacted small molecule reactant was removed by colloidal chromatography (Sephadax G25) to obtain a radioactive marker precursor protein such as DTPA-RGD-EGF and DTPA-RGD4C-EGF.
  • HEPES buffer solution An appropriate amount of HEPES buffer solution and radioactive ( 111 InCl 3 , 67 GaCl 3 , 90 YCl 3 and 177 LuCl 3 , etc.) were added to the above-mentioned fusion protein solution bound to DTPA, and reacted at room temperature for 1 hour. Excess EDTA was added to chelate the radioactive metal ions not bound to the protein, and purified by membrane filtration centrifugation and colloidal chromatography. The activity was measured and the radiochemical purity of the product was analyzed by radioactive thin layer analysis. The stability of the fusion protein in buffer solution (HEPES, 4 ° C) and serum (37 ° C).
  • radioactive 111 InCl 3 , 67 GaCl 3 , 90 YCl 3 and 177 LuCl 3 , etc.
  • Tumor cells or vascular cells were cultured in a 12-well cell culture dish at 37 ° C for one day in a CO 2 incubator.
  • the medium in the culture plate was removed, and the cells were washed with 1 mL of PBS, and then VEGF, RGD-VEGF and RGD4C-VEGF were separately added, and the cells were allowed to stand at 37 ° C for 2 hours in a CO 2 incubator.
  • Tumor mice (tumor size about 50-100 mm 3 ) were injected with 500 ⁇ Ci of radiolabeled recombinant protein in the tail vein, respectively.
  • Small animal SPECT/CT angiography was performed at 1, 2, 4, 8, 24, 48 and 72 hours after the injection. After the end of the angiography, the image was processed by software, and the tumor to muscle ratio was calculated by ROI (region of interest) analysis to evaluate the dynamic accumulation of the drug in the living body of the animal.
  • ROI region of interest
  • Computed tomography angiography.
  • the image shows that after the In-111 marker recombinant protein is applied, the specific accumulation of the tumor in the tumor (expressed as the tumor/muscle accumulation ratio) increases with time, reaching a peak at 8 hours after the drug injection, at this time 111 In
  • the tumor/muscle accumulation ratio of -DTPA-RGD4C-EGF was 4.4, slightly higher than 3.6 of 111 In-DTPA-RGD-EGF, and much higher than 1.7 of 111 In-DTPA-EGF.
  • RGD4C-EGF and RGD-EGF fusion proteins have dual targeting ability, and the accumulation of tumors is significantly higher than that of EGF recombinant protein with only single target ability;
  • the sequence of RGD4C-EGF fusion protein has better binding ability to integrin ⁇ v ⁇ 3, which makes it accumulate in tumors slightly better than RGD-EGF fusion protein with linear RGD sequence.

Abstract

Provided is a dual-targeting drug carrier, comprising a first targeting molecule and a second targeting molecule. The targeting molecule comprises a peptide, an antibody, or a protein. The targeting molecule is able to bind to a specific receptor or a specific protein or glycoprotein. The target molecule can also conjugate with a radiation therapy material to form a conjugate.

Description

双重标靶药物载体Double target drug carrier 技术领域Technical field
本发明涉及一种新颖的标靶药物载体,且特别涉及一种具有2种标靶分子的双重标靶药物载体。The present invention relates to a novel target drug carrier, and in particular to a dual target drug carrier having two target molecules.
背景技术Background technique
癌症是造成美国死亡的主要原因,癌症死亡率持续增加。癌症是细胞无法正常分裂,生长和分化。癌症最初的临床表现极不平均,在人体器官和组织上几乎可产生超过70种的癌症,且其中一些类似的癌症类型属于多个不相同的分子疾病。不幸的是,一些癌症可能无实际上的症状,直到在病程晚期,因此在治疗及预后上极为困难。Cancer is the leading cause of death in the United States, and cancer mortality continues to increase. Cancer is the inability of cells to divide, grow and differentiate normally. The initial clinical manifestations of cancer are extremely uneven, with almost 70 cancers occurring in human organs and tissues, and some of these similar cancer types belong to many different molecular diseases. Unfortunately, some cancers may have no actual symptoms until late in the course of the disease, making treatment and prognosis extremely difficult.
癌症治疗通常包括手术、化疗及/或放射治疗。目前所有的疗法都会发生严重的副作用,且降低生活质量。大多数的化疗药物会同时作用于正常和癌变组织。因此,在治疗癌性肿瘤的挑战之一是,给予癌细胞最大化的杀伤,同时尽量减少对健康组织的伤害。根据给药途径的药物(例如,静脉内)和性质(例如,它的物理和药物动力学性质),通常只有一小部分的投与量到达目标细胞,其余则作用于其他组织或迅速消失。Cancer treatment usually includes surgery, chemotherapy, and/or radiation therapy. All current therapies have serious side effects and reduce the quality of life. Most chemotherapeutic drugs act on both normal and cancerous tissues. Therefore, one of the challenges in treating cancerous tumors is to give cancer cells maximum killing while minimizing damage to healthy tissue. Depending on the drug (eg, intravenous) and nature of the route of administration (eg, its physical and pharmacokinetic properties), typically only a small fraction of the administered dose reaches the target cell, with the remainder acting on other tissues or rapidly disappearing.
为了提高传送效率,降低毒性非癌细胞,已有使药物递送至人体内特异性位点的各种方式。例如,使用单株抗体治疗癌症。抗 体提供目标选择性,但仍然存在昂贵以及与非目标细胞相互作用的问题。In order to improve delivery efficiency and reduce toxic non-cancerous cells, there have been various ways of delivering drugs to specific sites in the human body. For example, the use of monoclonal antibodies to treat cancer. Resistance The body provides target selectivity, but there are still problems with expensive and interaction with non-target cells.
近年来,致力于寻找肿瘤新生血管系统的特定目标物以发展新型的标靶治疗药物。肿瘤新生血管内皮细胞会大量表现多种特异性膜蛋白,包括αvβ3、αvβ5、整合素(integrin)、血管相关生长因子等。此外,过去研究发现短链胜肽(RGD、NGR)对于肿瘤血管新生内皮细胞具有极高的辨识度。In recent years, efforts have been made to find specific targets for tumor angiogenesis to develop novel target therapeutic drugs. Tumor neovascular endothelial cells express a large number of specific membrane proteins, including αvβ3, αvβ5, integrin, and vascular-related growth factors. In addition, past studies have found that short-chain peptides (RGD, NGR) have a very high recognition of tumor angiogenesis endothelial cells.
然而,肿瘤细胞具有高度不稳定性及变异性,且治疗过程中会对药物产生的抗药性。目前各种药物及诊断试剂对于改善癌症患者的存活仍缓慢且效果不彰。However, tumor cells are highly unstable and variability, and are resistant to drugs during treatment. At present, various drugs and diagnostic agents are still slow and ineffective in improving the survival of cancer patients.
发明内容Summary of the invention
有鉴于上述先前技术所存在的问题,本发明提供一种可用于诊断及治疗癌症的新颖载体。本发明的双重标靶药物载体为一种融合蛋白平台,能有效降低制药技术门坎与成本。In view of the problems with the prior art described above, the present invention provides a novel vector that can be used to diagnose and treat cancer. The dual target drug carrier of the invention is a fusion protein platform, which can effectively reduce the threshold and cost of the pharmaceutical technology.
本发明提供一种双重标靶药物载体,包括第一标靶分子以及第二标靶分子。The invention provides a dual target drug carrier comprising a first target molecule and a second target molecule.
在本发明的实施例中,其中所述第一或第二标靶分子专一性地结合至肿瘤细胞或/和肿瘤微环境中的血管内皮细胞In an embodiment of the invention, wherein the first or second target molecule specifically binds to vascular endothelial cells in tumor cells or/and tumor microenvironment
在本发明的实施例中,其中所述第一及第二标靶分子包括,但不限于,精氨酸-甘氨酸-天冬氨酸(RGD)、天冬酰胺-甘氨酸-精氨酸(NGR)、环状NGR(环状天冬酰胺-甘氨酸-精氨酸)、内化RGD(iRGD,内化-精氨酸-甘氨酸-天冬氨酸)、胱氨酸-甘氨酸-天门冬酰氨酸-赖氨酸-精氨酸-苏氨酸-精氨酸-甘氨酸-丙氨酸(CGNKRTRGA)、胃 泌素(gastrin)、蛙皮素(bombesin)、奥曲肽(octreotide)或其衍生物。大分子胜肽(peptide)包括,但不限于,表皮生长因子(EGF)、抗-EGFR(epidermal growth factor receptor,上表皮生长因子受体)抗体、血管内皮生长因子(VEGF)、抗-VEGFR(Vascular Endothelial Growth Factor Receptor,血管内皮细胞生长因子受体)抗体、抗-HER2(第二型人类表皮生长因子接受体,human epidermal growth factor receptor 2)抗体、肝细胞生长因子受体(HGFR)、抗-HGFR抗体、肿瘤坏死因子(TNF)或抗-TNF抗体。In an embodiment of the invention, wherein the first and second target molecules include, but are not limited to, arginine-glycine-aspartate (RGD), asparagine-glycine-arginine (NGR) ), cyclic NGR (cyclic asparagine-glycine-arginine), internalization of RGD (iRGD, internalization-arginine-glycine-aspartate), cystine-glycine-aspartate Acid-lysine-arginine-threonine-arginine-glycine-alanine (CGNKRTRGA), stomach Gastrin, bombesin, octreotide or a derivative thereof. Macromolecular peptides include, but are not limited to, epidermal growth factor (EGF), anti-EGFR (epidermal growth factor receptor) antibody, vascular endothelial growth factor (VEGF), anti-VEGFR ( Vascular Endothelial Growth Factor Receptor, anti-HER2 (human epidermal growth factor receptor 2) antibody, hepatocyte growth factor receptor (HGFR), anti-HIV - HGFR antibody, tumor necrosis factor (TNF) or anti-TNF antibody.
在本发明的实施例中,其中所述第一标靶分子以连接符与所述第二标靶分子连结。In an embodiment of the invention, wherein the first target molecule is linked to the second target molecule by a linker.
在本发明的实施例中,其中所述连接符为具有5至20个氨基酸的胜肽。In an embodiment of the invention, wherein the connector is a peptide having 5 to 20 amino acids.
在本发明的实施例中,其中所述连接符包括,但不限于,GG、PGGGG或GGGGSGGGGS,其中G系代表甘胺酸、P代表脯胺酸、S代表丝胺酸。In an embodiment of the invention, wherein the linker comprises, but is not limited to, GG, PGGGG or GGGGSGGGGS, wherein G represents glycine, P represents proline, and S represents serine.
在本发明的实施例中,更包括放射线同位素。In an embodiment of the invention, a radioisotope is further included.
本发明更提供一种医药组成物,包括上述双重标靶药物载体以及药学上可接受的载体。The invention further provides a pharmaceutical composition comprising the above dual target pharmaceutical carrier and a pharmaceutically acceptable carrier.
在本发明的实施例中,更包括微脂体。In an embodiment of the invention, a liposome is further included.
在本发明的实施例中,更包括纳米微粒。In an embodiment of the invention, nanoparticles are further included.
具体实施方式detailed description
图1A为纯化VEGF、RGD-VEGF、RGD4C-VEGF、EGF、 RGD-EGF、RGD4C-EGF蛋白的SDS-PAGE电泳图。Figure 1A shows purified VEGF, RGD-VEGF, RGD4C-VEGF, EGF, SDS-PAGE electropherogram of RGD-EGF and RGD4C-EGF protein.
图1B为marker、VEGF、RGD-VEGF、RGD4C-VEGF的SDS-PAGE电泳图,其中M为marker、1为VEGF(15.3Da)、2为RGD-VEGF(15.6Da)以及3为RGD4C-VEGF(16.3Da)。Figure 1B is a SDS-PAGE electrophoresis map of marker, VEGF, RGD-VEGF, and RGD4C-VEGF, wherein M is marker, 1 is VEGF (15.3 Da), 2 is RGD-VEGF (15.6 Da), and 3 is RGD4C-VEGF ( 16.3Da).
图2A-2F为RGD-VEGF及RGD4C-VEGF与αvβ3、VEGFR1、VEGFR2的结合曲线/柱状图。RGD-EGF及RGD4C-EGF可分别与αvβ3及EGFR结合。图2A为RGD-VEGF及RGD4C-VEGF与αvβ3的结合曲线图。图2B为RGD-VEGF及RGD4C-VEGF与αvβ3结合后以RGDfV多肽竞争之柱状图。图2C为RGD-VEGF及RGD4C-VEGF与VEGFR1的结合曲线图。图2D为RGD-VEGF及RGD4C-VEGF与VEGFR2的结合曲线图。2A-2F are binding curves/bar graphs of RGD-VEGF and RGD4C-VEGF and αvβ3, VEGFR1, VEGFR2. RGD-EGF and RGD4C-EGF bind to αvβ3 and EGFR, respectively. Figure 2A is a graph showing the binding of RGD-VEGF and RGD4C-VEGF to αvβ3. Figure 2B is a bar graph of competition of RGDfV polypeptide with RGD-VEGF and RGD4C-VEGF in combination with αvβ3. Figure 2C is a graph showing the binding of RGD-VEGF and RGD4C-VEGF to VEGFR1. Figure 2D is a graph showing the binding of RGD-VEGF and RGD4C-VEGF to VEGFR2.
图2E为RGD-VEGF及RGD4C-VEGF与VEGFR2结合后以VEGFR2抗体竞争之柱状图。图2F为RGD-EGF及RGD4C-EGF与αvβ3的结合曲线图。Figure 2E is a bar graph of competition of VEGFR2 antibodies by RGD-VEGF and RGD4C-VEGF in combination with VEGFR2. Figure 2F is a graph showing the binding of RGD-EGF and RGD4C-EGF to αvβ3.
图3A为U87MG(表现αvβ3及EGFR)细胞与RGD-VEGF及RGD4C-VEGF的结合曲图。RGD-VEGF及RGD4C-VEGF可分别与表现αvβ3及EGFR结合。Figure 3A is a graph showing the binding of U87MG (expressing αvβ3 and EGFR) cells to RGD-VEGF and RGD4C-VEGF. RGD-VEGF and RGD4C-VEGF can bind to αvβ3 and EGFR, respectively.
图3B为U87MG(表现αvβ3及EGFR)细胞与RGD-VEGF及RGD4C-VEGF的结合柱状图。Figure 3B is a bar graph of binding of U87MG (expressing αvβ3 and EGFR) cells to RGD-VEGF and RGD4C-VEGF.
图3C为HUVEC(表现VEGFR1、VEGFR2与αvβ3)细胞与RGD-VEGF、RGD4C-VEGF及NGR-VEGF的结合曲线图。RGD-VEGF、RGD4C-VEGF及NGR-VEGF可结合至表现VEGFR1、 VEGFR2与αvβ3的细胞。Figure 3C is a graph showing the binding of HUVEC (expressing VEGFR1, VEGFR2 and αvβ3) cells to RGD-VEGF, RGD4C-VEGF and NGR-VEGF. RGD-VEGF, RGD4C-VEGF and NGR-VEGF can bind to express VEGFR1 Cells of VEGFR2 and αvβ3.
图3D为HUVEC(表现VEGFR1、VEGFR2与αvβ3)细胞与RGD-VEGF、RGD4C-VEGF及NGR-VEGF的结合柱状图。Figure 3D is a bar graph of binding of HUVEC (expressing VEGFR1, VEGFR2 and αvβ3) cells to RGD-VEGF, RGD4C-VEGF and NGR-VEGF.
图4A-4D为细胞黏附试验。RGD-VEGF及RGD4C-VEGF涂覆于细胞盘后,可增加U87MG细胞及HUVEC细胞贴附于培养盘上的数量,若加入大量RGD胜肽链竞争,可减少细胞的吸附。其中图4A为RGD-VEGF及RGD4C-VEGF涂覆于细胞盘后,可增加U87MG细胞贴附于培养盘上的数量,而图4B为添加RGDfV多肽后,细胞贴附于培养盘上的数量即受到抑制,图4C-4D为RGD-VEGF及RGD4C-VEGF与HUVEC细胞结合后,再分别以RGDfV多肽或VEGFR2重组蛋白竞争,分别以吸光值及百分比表示。Figures 4A-4D are cell adhesion assays. After coating RGD-VEGF and RGD4C-VEGF on the cell plate, the number of U87MG cells and HUVEC cells attached to the culture plate can be increased. If a large amount of RGD peptide chain is added to compete, the adsorption of cells can be reduced. 4A shows that RGD-VEGF and RGD4C-VEGF are applied to the cell plate to increase the number of U87MG cells attached to the culture plate, and FIG. 4B shows the amount of cells attached to the culture plate after the addition of the RGDfV polypeptide. Inhibition, Figure 4C-4D shows that RGD-VEGF and RGD4C-VEGF bind to HUVEC cells, and then compete with RGDfV polypeptide or VEGFR2 recombinant protein, respectively, in terms of absorbance and percentage.
图5A-5B显示RGD-EGF及RGD4C-EGF或RGD-VEGF及RGD4C-VEGF分别可活化EGFR或VEGFR下游的讯息传递途径。本发明双重标靶药物载体的EGF或VEGF仍保有其原本的生物特性。其中5A图的M为Marker、C为控制组(无加入蛋白)、E为EGF(25nM)、R-E为RGD-EGF(25nM)以及R4C-E为RGD4C-EGF(25nM),而图5B的C为控制组(无加入蛋白)、R-V为RGD-VEGF、R4-V RGD4C-VEGF。Figures 5A-5B show that RGD-EGF and RGD4C-EGF or RGD-VEGF and RGD4C-VEGF activate a signaling pathway downstream of EGFR or VEGFR, respectively. The EGF or VEGF of the dual target drug carrier of the invention retains its original biological properties. Among them, M of Figure 5A is Marker, C is the control group (no protein added), E is EGF (25 nM), RE is RGD-EGF (25 nM), and R4C-E is RGD4C-EGF (25 nM), while C of Figure 5B For the control group (no protein added), RV is RGD-VEGF, R4-V RGD4C-VEGF.
图6为放射性标志融合蛋白于缓冲溶液(HEPES,4℃)及血清(37℃)的稳定度试验。将放射性标志融合蛋白保存于Hepes缓冲液(4℃)及胎牛血清(37℃)的环境下,经24小时后放射化学纯度皆仍高于90%,显示经放射性In-111标志的融合蛋白的血清稳定度极佳。 Figure 6 is a graph showing the stability of a radiolabeled fusion protein in a buffer solution (HEPES, 4 ° C) and serum (37 ° C). The radiolabeled fusion protein was stored in Hepes buffer (4 ° C) and fetal bovine serum (37 ° C), and the radiochemical purity was still higher than 90% after 24 hours, indicating the radioactive In-111-labeled fusion protein. The serum stability is excellent.
图7为荧光摄影图。RGD-VEGF与RGD4C-VEGF标志重组蛋白可结合至细胞。Fig. 7 is a fluorescent photograph. The RGD-VEGF and RGD4C-VEGF marker recombinant proteins can bind to cells.
图8为放射性标志重组蛋白的单光子及计算机断层扫瞄(SPECT/CT)造影。荷U87MG皮下肿瘤小鼠经尾静脉注射111In–DTPA(二乙基三胺五乙酸,Diethylene triamine pentacetate aci)-EGF、111In-DTPA-RGD-EGF及111In-DTPA-RGD4C-EGF后1、4、8及24小时,进行小动物单光子及计算机断层扫瞄(SPECT/CT)造影。影像显示施打In-111标志重组蛋白后,三者于肿瘤的专一性积聚(以肿瘤/肌肉积聚比值表示)皆随时间而提高,皆于药物注射后8小时达高峰,此时111In-DTPA-RGD4C-EGF的肿瘤/肌肉积聚比为4.4,略高于111In-DTPA-RGD-EGF的3.6,而远高于111In-DTPA-EGF的1.7。Figure 8 is a single photon and computed tomography (SPECT/CT) angiogram of radiolabeled recombinant protein. U87MG subcutaneous tumor mice were injected with 111 In-DTPA (Diethylene triamine pentacetate aci)-EGF, 111 In-DTPA-RGD-EGF and 111 In-DTPA-RGD4C-EGF. Small animal single photon and computed tomography (SPECT/CT) angiography was performed at 4, 8 and 24 hours. The image shows that after the In-111 marker recombinant protein is applied, the specific accumulation of the tumor in the tumor (expressed as the tumor/muscle accumulation ratio) increases with time, reaching a peak at 8 hours after the drug injection, at this time 111 In The tumor/muscle accumulation ratio of -DTPA-RGD4C-EGF was 4.4, slightly higher than 3.6 of 111 In-DTPA-RGD-EGF, and much higher than 1.7 of 111 In-DTPA-EGF.
具体实施方式detailed description
本发明提供一种双重标靶药物载体,包括第一标靶分子以及第二标靶分子。The invention provides a dual target drug carrier comprising a first target molecule and a second target molecule.
本发明所述的标靶分子为胜肽、抗体或蛋白质。本发明的标靶分子可经化学性修饰以增加稳定性。化学修饰胜肽或胜肽类似物包括任何可in vivo(体内)或in vitro(体外)增加稳定性及/或效力的化学功能性等同物。胜肽类似物是指任何胜肽的氨基酸衍生物。胜肽类似物包括,但不限于,在合成时修饰支链、合并未饱和氨基酸及/或其衍生物、交联与其它可增加胜肽象约束的方法。例如,支链修饰包括胺基的修饰。 The target molecule of the present invention is a peptide, antibody or protein. The target molecules of the invention can be chemically modified to increase stability. Chemically modified peptides or peptide analogs include any chemically functional equivalent that increases stability and/or potency either in vivo or in vitro. A peptide analog refers to an amino acid derivative of any peptide. Peptide analogs include, but are not limited to, modified branches upon synthesis, combined with unsaturated amino acids and/or derivatives thereof, cross-linking and other methods that increase the peptide identity. For example, a branched modification includes a modification of an amine group.
标靶分子可in vivo及in vitro结合至病灶组织,特别为肿瘤、癌症组织/细胞及肿瘤微环境的血管内皮细胞。因此,当标靶胜肽与报导分子(生物成像的荧光分子或放射线)共轭时,肿瘤标靶胜肽可直接显示肿瘤位置,有助于癌症的诊断。本发明所述的“共轭”是指2个有助于治疗/诊断肿瘤的分子(如肿瘤标靶胜肽与报导分子)通过足够的亲合力结合。共轭可以共价、非共价键或其它形式的结合方式完成,例如,包覆。The target molecule can bind to the lesion tissue in vivo and in vitro, especially for vascular endothelial cells of tumor, cancer tissue/cell and tumor microenvironment. Therefore, when the target peptide is conjugated to a reporter molecule (bioimaging fluorescent molecule or radiation), the tumor target peptide can directly display the tumor location and contribute to the diagnosis of cancer. "Conjugation" as used in the present invention means that two molecules that contribute to the treatment/diagnosis of a tumor, such as a tumor target peptide and a reporter molecule, are bound by sufficient affinity. Conjugation can be accomplished by covalent, non-covalent bonds or other forms of bonding, for example, coating.
本发明的标靶分子可为小分子胜肽及/或大分子胜肽。小分子胜肽包括,但不限于,氨酸-甘氨酸-天冬氨酸(RGD)、天冬酰胺-甘氨酸-精氨酸(NGR)、环状NGR、内化RGD(iRGD)、胱氨酸-甘氨酸-天门冬酰氨酸-赖氨酸-精氨酸-苏氨酸-精氨酸-甘氨酸-丙氨酸(CGNKRTRGA)、胃泌素(gastrin)、蛙皮素(bombesin)、奥曲肽(octreotide)或其衍生物。大分子胜肽包括,但不限于,表皮生长因子(EGF)、抗-EGFR抗体、血管内皮生长因子(VEGF)、抗-VEGFR抗体、抗-HER2抗体、肝细胞生长因子受体(HGFR)、抗-HGFR抗体、肿瘤坏死因子(TNF)或抗-TNF抗体。The target molecule of the present invention may be a small molecule peptide and/or a macropeptide. Small molecule peptides include, but are not limited to, lysine-glycine-aspartate (RGD), asparagine-glycine-arginine (NGR), cyclic NGR, internalization RGD (iRGD), cystine - glycine-aspartate-lysine-arginine-threonine-arginine-glycine-alanine (CGNKRTRGA), gastrin, bombesin, octreotide Octreotide) or a derivative thereof. Macromolecular peptides include, but are not limited to, epidermal growth factor (EGF), anti-EGFR antibodies, vascular endothelial growth factor (VEGF), anti-VEGFR antibodies, anti-HER2 antibodies, hepatocyte growth factor receptor (HGFR), Anti-HGFR antibody, tumor necrosis factor (TNF) or anti-TNF antibody.
在实施例中,标靶分子可与成像物质或放疗物质(放射性药物或同位素),形成共轭物。In an embodiment, the target molecule can form a conjugate with an imaging substance or a radiotherapeutic substance (radiopharmaceutical or isotope).
放射性成像物质包括,但不限于,122I、123I、124I、125I、131I、18F、75Br、76Br、76Br、77Br、211At、225Ac、177Lu、153Sm、186Re、188Re、67Cu、213Bi、212Bi、212Pb与67Ga。Radioactive imaging materials include, but are not limited to, 122 I, 123 I, 124 I, 125 I, 131 I, 18 F, 75 Br, 76 Br, 76 Br, 77 Br, 211 At, 225 Ac, 177 Lu, 153 Sm , 186 Re, 188 Re, 67 Cu, 213 Bi, 212 Bi, 212 Pb and 67 Ga.
放射性药物包括,但不限于,111In羟基喹啉、131I碘化钠、99mTc苯溴氨乙酸、99mTc红血球、123I碘化钠、99mTc Exametazime、99mTc 大颗粒聚合白蛋白、99mTc亚甲膦酸盐、99mTc Mertiatide、99mTc羟亚甲基二膦酸盐、99mTc三胺五乙酸、99mTc Pertechnetate、99mTcSestamibi、99mTc硫胶体、99mTc Tetrofosmin、201Tl与133Xe。Radiopharmaceuticals include, but are not limited to, 111 In hydroxyquinoline, 131 I sodium iodide, 99m Tc phenyl bromide, 99m Tc red blood cells, 123 I sodium iodide, 99m Tc Exametazime, 99m Tc large particle polymeric albumin, 99m Tc-methylene phosphonate, 99m Tc Mertiatide, 99m Tc hydroxymethylene diphosphonate, 99m Tc triamine pentaacetic acid, 99m Tc Pertechnetate, 99m TcSestamibi, 99m Tc sulphur colloid, 99m Tc Tetrofosmin, 201 Tl and 133 Xe .
放射线同位素包括,但不限于,52Fe、52mMn、55Co、64Cu、67Ga、68Ga、99mTc、111In、123I、125I、131I、32P、47Sc、67Cu、90Y、109Pd、111Ag、149Pm、186Re、188Re、211At、212Bi、213Bi、105Rh、153Sm、177Lu与198Au。Radiation isotopes include, but are not limited to, 52 Fe, 52 mMn, 55 Co, 64 Cu, 67 Ga, 68 Ga, 99 mTc, 111 In, 123 I, 125 I, 131 I, 32 P, 47 Sc, 67 Cu, 90 Y, 109 Pd, 111 Ag, 149 Pm, 186 Re, 188 Re, 211 At, 212 Bi, 213 Bi, 105 Rh, 153 Sm, 177 Lu and 198 Au.
本发明中所述的肿瘤标靶胜肽或其共轭物可经非肠胃、局部、直肠、鼻、阴道、植入、或喷雾吸入等方式给予。本发明中所述对的“非肠胃”包括皮下、皮内、静脉、肌内、关节、动脉、滑膜、病灶或颅内注射。The tumor target peptide or its conjugate described in the present invention can be administered by parenteral, topical, rectal, nasal, vaginal, implantation, or spray inhalation. "Non-gastrointestinal" as used in the present invention includes subcutaneous, intradermal, intravenous, intramuscular, articular, arterial, synovial, focal or intracranial injections.
本发明所述的“连接符”是指2-50个合成氨基酸的序列,较佳5-20个合成氨基酸的序列,其连接二个胜肽序列,例如,连接2个胜肽结构。本发明的连接符可透过胜肽键连接2个氨基酸序列。本发明的连接符胜肽为直链序列,连接第一部分与第二部分。在实施例中,可置换连接符可为(GGGGS)n、(G)n、(EAAAK)n、(XP)n或(PAPAP)n,例如,GG、GGSG、GGGS、GGGGS、GGSGG、GGGSGG、GGGSGGG、GGGSGGGS、GGGSGGGGS、ASGG、GGGSASGG、SGCGS、GGGGSGGGG、GGSHG、SGGCGGS及AACAA,其中所述G表示甘氨酸;所述S表示絲氨酸;所述E表示谷氨酸;所述A表示丙氨酸;所述K表示赖氨酸;所述P表示脯胺酸;所述H表示组氨酸;所述C表示胱氨酸。The "linker" as used in the present invention refers to a sequence of 2 to 50 synthetic amino acids, preferably 5 to 20 synthetic amino acids, which are linked to two peptide sequences, for example, to link two peptide structures. The connector of the present invention can link two amino acid sequences through a peptide bond. The linker peptide of the present invention is a linear sequence linking the first portion to the second portion. In an embodiment, the replaceable connector may be (GGGGS)n, (G)n, (EAAAK)n, (XP)n or (PAPAP)n, eg, GG, GGSG, GGGS, GGGGS, GGSGG, GGGSGG, GGGSGGG, GGGSGGGS, GGGSGGGGS, ASGG, GGGSASGG, SGCGS, GGGGSGGGG, GGSHG, SGCGGGS, and AACAA, wherein said G represents glycine; said S represents serine; said E represents glutamic acid; said A represents alanine; K represents lysine; P represents valine; said H represents histidine; and C represents cystine.
综上所述,本发明双重标靶药物载体可携带肿瘤治疗药物或其 它物质(如治疗性放射核种),以成为肿瘤标靶药物载体。此外,也可携带可进行分子造影的药物,作为肿瘤诊断地分子影像探针。本发明双重标靶显影剂载体并具肿瘤诊断的效果。In summary, the dual target drug carrier of the present invention can carry a tumor therapeutic drug or It is a substance (such as a therapeutic radionuclide) that becomes a tumor target drug carrier. In addition, a drug capable of molecular imaging can also be carried as a molecular imaging probe for tumor diagnosis. The double target developer carrier of the invention has the effect of tumor diagnosis.
实施例1Example 1
1.融合蛋白质体的制备1. Preparation of fusion protein bodies
将EGF、RGD-EGF、RGD4C-EGF、VEGF、RGD-VEGF及RGD4C-VEGF基因以Nco I及Xho I限制酶切位建构于pET28a(+)的表达载体。RGD、RGD4C的C端(即碳端)与EGF或VEGF的N端(即氮端)相接,且EGF或VEGF的C端有His6标志。His6标志有利于后续的纯化及分析,其中RGD-EGF、RGD4C-EGF、RGD-VEGF及RGD4C-VEGF的基因序列包含连接符GG。将建构完成的pET28a(+)EGF、RGD-EGF、RGD4C-EGF、VEGF、RGD-VEGF及RGD4C-VEGF表现载体转染至E.coli.DH5α,以生产及保存载体DNA。经DNA定序确认序列正确。EGF, RGD-EGF, RGD4C-EGF, VEGF, RGD-VEGF and RGD4C-VEGF genes were constructed in the expression vector of pET28a(+) with Nco I and Xho I restriction enzyme cleavage sites. The C-terminus (ie, the carbon terminus) of RGD and RGD4C is ligated to the N-terminus (ie, the nitrogen terminus) of EGF or VEGF, and the C-terminus of EGF or VEGF has a His 6 flag. The His 6 marker facilitates subsequent purification and analysis, wherein the gene sequences of RGD-EGF, RGD4C-EGF, RGD-VEGF, and RGD4C-VEGF comprise the linker GG. The constructed pET28a(+) EGF, RGD-EGF, RGD4C-EGF, VEGF, RGD-VEGF and RGD4C-VEGF expression vectors were transfected into E. coli.DH5α to produce and preserve vector DNA. The sequence was confirmed by DNA sequencing.
2.标靶融合蛋白的表现及纯化2. Expression and purification of target fusion protein
将实施例1的载体以热休克的方式送入E.coli BL21(DE3),以卡纳霉素(50μg/mL)筛选及确认。将带有蛋白表现载体的E.coliBL21(DE3)菌株,以1mM IPTG在37℃诱导16小时。将菌液经由法国压破菌仪(French press)以30PSI压力破菌,13000g离心30分钟,分别收取上清液及沉淀物(pellet)的样本进行SDS-PAGE电泳分析,由SDS-PAGE可观察到标靶融合蛋白的表现。若是以不可溶的包含体(inclusion body)形式存在菌体中, 后续蛋白生产收取破菌后的沉淀物进行蛋白纯化。溶解后的标靶融合蛋白以0.45μm滤膜过滤后,接着将标靶融合蛋白以1:50比例的复性(refolding)缓冲液(20mM Tris,0.1Mm GSSG,1mM GSH,1mM EDTA pH 8.0)进行复性稀释(dilution refolding)。将标靶蛋白样本缓慢地加入复性缓冲液中,且标靶融合蛋白稀释后的浓度不超过0.2mg/ml,于4℃进行复性12小时,再以0.45μm滤膜过滤,并去除沉淀的标靶蛋白。The vector of Example 1 was sent to E. coli BL21 (DE3) by heat shock, and screened and confirmed by kanamycin (50 μg/mL). The E. coli BL21 (DE3) strain carrying the protein expression vector was induced with 1 mM IPTG for 16 hours at 37 °C. The bacterial solution was sterilized by a French press at a pressure of 30 PSI, centrifuged at 13,000 g for 30 minutes, and samples of the supernatant and the pellet were collected for SDS-PAGE electrophoresis analysis, and observed by SDS-PAGE. Performance to the target fusion protein. If it is in the form of an insoluble body, the bacteria are present in the form of an insoluble body. Subsequent protein production receives a pellet after the disruption for protein purification. The lysed target fusion protein was filtered through a 0.45 μm filter, followed by a 1:50 ratio of refolding buffer (20 mM Tris, 0.1 Mm GSSG, 1 mM GSH, 1 mM EDTA pH 8.0). Dilution refolding. The target protein sample was slowly added to the refolding buffer, and the concentration of the target fusion protein after dilution was not more than 0.2 mg/ml, and the renaturation was carried out at 4 ° C for 12 hours, and then filtered through a 0.45 μm filter to remove the precipitate. Target protein.
将蛋白复性液以5mL/min流速通过镍管柱(nickel column)。先以缓冲液A(20mM Tris(pH8.0),500mM NaCl,20mM咪唑,0.5mM PMSF)清洗管柱,接着进行洗堤,利用缓冲液A与缓冲液B(20mM Tris(pH8.0),500mM NaCl,500mM咪唑,0.5mM PMSF)的比例调整溶液的咪唑浓度,以洗涤出标靶融合蛋白。由于收集的标靶融合蛋白仍有其它杂质蛋白,因此利用凝胶管柱(HiPrap 26/60S-100)分离出融合蛋白。以
Figure PCTCN2016075329-appb-000001
浓缩离心管(Millipore)浓缩蛋白样本,将蛋白浓度调整至约0.5~3mg/mL,并分装至微量离心管,贮存于-80℃。以SDS-PAGE分析确认蛋白纯度,如图1所示。
The protein refolding solution was passed through a nickel column at a flow rate of 5 mL/min. The column was first washed with buffer A (20 mM Tris (pH 8.0), 500 mM NaCl, 20 mM imidazole, 0.5 mM PMSF), followed by washing, using buffer A and buffer B (20 mM Tris (pH 8.0), The ratio of the imidazole of the solution was adjusted in proportion to 500 mM NaCl, 500 mM imidazole, 0.5 mM PMSF) to wash out the target fusion protein. Since the collected target fusion protein still has other impurity proteins, the fusion protein was isolated using a gel column (HiPrap 26/60S-100). Take
Figure PCTCN2016075329-appb-000001
A concentrated centrifuge tube (Millipore) was used to concentrate the protein sample, the protein concentration was adjusted to about 0.5 to 3 mg/mL, and dispensed into a microcentrifuge tube and stored at -80 °C. Protein purity was confirmed by SDS-PAGE analysis as shown in Figure 1.
3.受体结合试验3. Receptor binding assay
3.1标靶融合蛋白与Integrin的结合3.1 Target fusion protein binding to Integrin
以ELISA cell binding(ELISA细胞结合)试验分析EGF、RGD-EGF、NGR-EGF、RGD4C-EGF、VEGF、RGD-VEGF、NGR-VEGF、及RGD4C-VEGF标靶融合蛋白与细胞的结合。将25μg/well溶于PBS(含钙、镁离子)的integrin(αvβ3)受体置 于96well ELISA微量盘,于4℃培养12小时后,以3%BSA于25℃反应1小时。加入系列稀释的标靶融合蛋白于25℃培养2小时,经PBS缓冲液(含钙、镁离子)清洗后,以小鼠抗-His-HRP抗体侦测蛋白结合到受体的情形,测量波长450nm的吸光值以量化结合于受体上的蛋白量。所得数据利用prism软件作非线性曲线拟合(curve fitting)可得蛋白结合的解离常数Kd值,进而可作为蛋白对不同受体结合能力的比较。The binding of EGF, RGD-EGF, NGR-EGF, RGD4C-EGF, VEGF, RGD-VEGF, NGR-VEGF, and RGD4C-VEGF target fusion proteins to cells was analyzed by ELISA cell binding assay. 25 μg/well of integrin (αvβ3) receptor dissolved in PBS (calcium and magnesium ions) was placed in a 96well ELISA microplate, and cultured at 4 ° C for 12 hours, and then reacted with 3% BSA at 25 ° C for 1 hour. The serially diluted target fusion protein was added to culture at 25 ° C for 2 hours, and after washing with PBS buffer (containing calcium and magnesium ions), the mouse anti-His-HRP antibody was used to detect the binding of the protein to the receptor, and the wavelength was measured. The absorbance at 450 nm is used to quantify the amount of protein bound to the receptor. The obtained data using the prism software for nonlinear curve fitting can obtain the dissociation constant K d value of protein binding, which can be used as a comparison of the binding ability of the protein to different receptors.
3.2标靶融合蛋白与EGFR、VEGFR的结合3.2 Binding fusion protein binding to EGFR and VEGFR
将25μg/well溶于PBS缓冲液(含钙、镁离子)的抗-IgG抗体置于96well ELISA微量盘,于4℃培养12小时后,以3%BSA于25℃反应1小时。加入25ng/well EGFR或VEGFR,于25℃反应2小时。经PBS缓冲液(含钙、镁离子)清洗后,加入系列稀释的标靶融合蛋白于25℃培养2小时。经由PBS缓冲液(含钙、镁离子)清洗后,以小鼠抗-His-HRP抗体侦测蛋白结合到受体的情形,测量波长450nm的吸光值以量化结合于受体上的蛋白量。所得数据利用prism软件作非线性曲线拟合(curve fitting)可得蛋白结合的解离常数Kd值,进而可作为蛋白对不同受体结合能力的比较。25 μg/well of anti-IgG antibody dissolved in PBS buffer (calcium, magnesium ion) was placed in a 96well ELISA microplate, and cultured at 4 ° C for 12 hours, and then reacted with 3% BSA at 25 ° C for 1 hour. 25 ng/well EGFR or VEGFR was added and reacted at 25 ° C for 2 hours. After washing with PBS buffer (containing calcium and magnesium ions), serially diluted target fusion proteins were added and incubated at 25 ° C for 2 hours. After washing with PBS buffer (containing calcium and magnesium ions), the mouse anti-His-HRP antibody was used to detect binding of the protein to the receptor, and the absorbance at a wavelength of 450 nm was measured to quantify the amount of protein bound to the receptor. The obtained data using the prism software for nonlinear curve fitting can obtain the dissociation constant Kd value of protein binding, which can be used as a comparison of the binding ability of the protein to different receptors.
EGF、RGD-EGF、RGD4C-EGF与EGFR的解离常数Kd值分别为2.38、20.35及14.33nM。VEGF、RGD-VEGF、RGD4C-VEGF与Integrin的解离常数Kd值分别为0、5.0及1.0。VEGF、RGD-VEGF、RGD4C-VEGF与VEGFR1的解离常数Kd值分别为15.3、11.3及11.4。VEGF、RGD-VEGF、RGD4C-VEGF与VEGFR2 的解离常数Kd值分别为22.2、12.1及13.9。The dissociation constant K d values of EGF, RGD-EGF, RGD4C-EGF and EGFR were 2.38, 20.35 and 14.33 nM, respectively. The dissociation constant K d values of VEGF, RGD-VEGF, RGD4C-VEGF and Integrin were 0, 5.0 and 1.0, respectively. VEGF, RGD-VEGF, VEGFR1 solution RGD4C-VEGF with a dissociation constant K d values of 15.3,11.3 and 11.4. The dissociation constant K d values of VEGF, RGD-VEGF, RGD4C-VEGF and VEGFR2 were 22.2, 12.1 and 13.9, respectively.
参照图2A-2F,各类双重标靶融合蛋白可与两种以上的受体进行结合,具有双重标靶的能力。RGD-EGF及RGD4C-EGF可分别与Integrin(αvβ3)及EGFR结合,RGD-VEGF及RGD4C-VEGF可分别与Integrin(αvβ3)、VEGFR1及VEGFR2结合。Referring to Figures 2A-2F, various dual-target fusion proteins can bind to more than two receptors, with the ability to dual target. RGD-EGF and RGD4C-EGF can bind to Integrin (αvβ3) and EGFR, respectively, and RGD-VEGF and RGD4C-VEGF can bind to Integrin (αvβ3), VEGFR1 and VEGFR2, respectively.
4.细胞结合试验4. Cell binding assay
本实施例使用MDA-MB468(高度表现EGFR)、MDA-MB231(中度表现EGFR)、MCF-7(低度表现)、HT1080(表现APN)、U87MG(表现αvβ3)与HUVEC(表现VEGFR与αvβ3)细胞株,以分析双重标靶融合蛋白与不同受体的专一性结合。This example uses MDA-MB468 (highly expressed EGFR), MDA-MB231 (moderately expressed EGFR), MCF-7 (low performance), HT1080 (expressed APN), U87MG (expressed αvβ3) and HUVEC (expressed VEGFR and αvβ3) Cell lines to analyze the specific binding of the dual-target fusion protein to different receptors.
将MDA-MB468、MDA-MB231、MCF-7、HT1080、U87MG与HUVEC细胞,分别以20,000cells/well的量接种于96孔微量盘,于37℃培养12小时。待细胞贴附后,以4℃预冷的4%三聚甲醛(para-formadehyde)固定后,于室温培养15分钟。固定后的细胞与3%FBS于25℃培养1小时后,加入连续稀释的标靶融合蛋白,于25℃培养1小时。经PBS缓冲液(含1mmol/L CaCl2及0.5mmol/L MgCl2)清洗后,以老鼠抗-His-HRP抗体侦测蛋白结合到细胞的情形,再利用HRP的受质TMB呈色,测量波长450nm的吸光值,以量化结合于细胞上的蛋白量。所得数据利用prism软件作非线性曲线拟合(curve fitting)可得蛋白结合的解离常数Kd值。MDA-MB468, MDA-MB231, MCF-7, HT1080, U87MG and HUVEC cells were seeded in a 96-well microplate at a dose of 20,000 cells/well, and cultured at 37 ° C for 12 hours. After the cells were attached, they were fixed with 4% paraformaldehyde hyde precooled at 4 ° C, and then incubated at room temperature for 15 minutes. After the fixed cells were incubated with 3% FBS at 25 ° C for 1 hour, serially diluted target fusion proteins were added and incubated at 25 ° C for 1 hour. After washing with PBS buffer (containing 1 mmol/L CaCl 2 and 0.5 mmol/L MgCl 2 ), the mouse anti-His-HRP antibody was used to detect the binding of the protein to the cells, and then the TMP of the HRP was used for color measurement. The absorbance at a wavelength of 450 nm to quantify the amount of protein bound to the cells. The data obtained using prism software for non-linear curve fitting (curve fitting) available binding protein dissociation constant K d value.
EGF、RGD-EGF、RGD4C-EGF与MDA MB 468细胞上的EGFR的解离常数Kd值分别为16.38、26.97及22.22nM。VEGF、 RGD-VEGF、RGD4C-VEGF、NGR-VEGF与U87MG细胞上的intergrin的解离常数Kd值分别为1.74、1.83、1.9及1.9μM。VEGF、RGD-VEGF、RGD4C-VEGF、NGR-VEGF与HUVEC细胞上的VEGFR及intergrin的解离常数Kd值分别为12.7、11.9、6.4及9.4nM。EGF, RGD-EGF, EGFR solution on RGD4C-EGF with the MDA MB 468 cell dissociation constant K d values of 16.38,26.97 and 22.22nM. VEGF, RGD-VEGF, RGD4C- VEGF, NGR-VEGF and decompression intergrin U87MG cells on the dissociation constant K d values of 1.74,1.83,1.9 and 1.9μM. VEGF, RGD-VEGF, RGD4C- VEGF, NGR-VEGF and VEGFR intergrin solution on HUVEC cells with a dissociation constant K d values of 12.7,11.9,6.4 and 9.4nM.
参照图3A-3D,RGD-EGF、RGD4C-EGF、RGD-VEGF及RGD4C-VEGF可与表达对应受体的细胞结合。Referring to Figures 3A-3D, RGD-EGF, RGD4C-EGF, RGD-VEGF, and RGD4C-VEGF can bind to cells expressing the corresponding receptor.
以RGD胜肽或VEGFR抗体与RGD-VEGF及RGD4C-VEGF于U87MG及HUVEC细胞中进行竞争性结合试验。竞争物的添加可有效降低RGD-VEGF及RGD4C-VEGF与细胞的结合。结果证实,本发明的双重标靶分子可以专一性的结合至肿瘤的生物标记上。Competitive binding assays were performed in U87MG and HUVEC cells with RGD peptide or VEGFR antibody and RGD-VEGF and RGD4C-VEGF. The addition of the competitor can effectively reduce the binding of RGD-VEGF and RGD4C-VEGF to cells. As a result, it was confirmed that the dual target molecule of the present invention can specifically bind to the biomarker of the tumor.
5.细胞黏附试验5. Cell adhesion test
本实施例使用ECM细胞黏附分析系统来分析细胞的黏附特性。于96孔微量盘每孔涂覆不同浓度的标靶融合蛋白,于16℃培养12小时。待蛋白贴附后,加入3%BSA,于25℃培养1小时。将已饥饿(starvation)一小时的肿瘤细胞(U87MG,5x 105cells/well)加至96孔微量盘,于37℃培养2小时后,以D-PBS清洗,去除未结合到蛋白的细胞。最后加入MTT,于37℃培养1小时,存活的细胞会将MTT药剂代谢成甲腊(formazan)蓝紫色结晶。以DMSO溶解甲腊,侦测波长570nm的吸光值。以下列公式计算细胞贴附特性。This example uses an ECM cell adhesion analysis system to analyze the adhesion characteristics of cells. Different concentrations of the target fusion protein were coated per well in a 96-well microplate and incubated at 16 ° C for 12 hours. After the protein was attached, 3% BSA was added and incubated at 25 ° C for 1 hour. Tumor cells (U87MG, 5 ×10 5 cells/well) that had been starved for one hour were added to a 96-well microplate, and after incubation at 37 ° C for 2 hours, they were washed with D-PBS to remove cells not bound to the protein. Finally, MTT was added and incubated at 37 ° C for 1 hour. The surviving cells metabolized the MTT agent to formazan blue-violet crystals. The wax was dissolved in DMSO to detect the absorbance at a wavelength of 570 nm. The cell attachment characteristics were calculated by the following formula.
细胞贴附(%)=(试验组OD值/对照组OD值)×100%Cell attachment (%) = (test group OD value / control group OD value) × 100%
参照图4A-D,将RGD-VEGF及RGD4C-VEGF涂覆于细胞盘 后,可增加U87MG细胞及HUVEC细胞贴附于培养盘上的数量,若加入大量RGD胜肽链竞争,可减少细胞的吸附。图4A为RGD-VEGF及RGD4C-VEGF涂覆于细胞盘后,可增加U87MG细胞贴附于培养盘上的数量,而图4B为添加RGDfV多肽后,细胞贴附于培养盘上的数量即受到抑制,图4C-4D为RGD-VEGF及RGD4C-VEGF与HUVEC细胞结合后,再分别以RGDfV多肽或VEGFR2重组蛋白竞争,分别以吸光值及百分比表示。Referring to Figures 4A-D, RGD-VEGF and RGD4C-VEGF were applied to the cell disk After that, the number of U87MG cells and HUVEC cells attached to the culture plate can be increased, and if a large amount of RGD peptide chain is added to compete, the adsorption of cells can be reduced. Figure 4A shows that RGD-VEGF and RGD4C-VEGF are coated on the cell plate to increase the amount of U87MG cells attached to the culture plate, while Figure 4B shows the amount of cells attached to the plate after the addition of the RGDfV polypeptide. Inhibition, Figure 4C-4D shows that RGD-VEGF and RGD4C-VEGF bind to HUVEC cells, and then compete with RGDfV polypeptide or VEGFR2 recombinant protein, respectively, in terms of absorbance and percentage.
6.细胞活化试验6. Cell activation test
以细胞实验测试,融合蛋白活化细胞受体及其下游讯息传递分子磷酸化反应。将各类人类肿瘤细胞与25nM标靶融合蛋白于37℃培养60分钟,收细胞内蛋白萃取物,以西方墨点法分析细胞受体及其下游讯号分子的磷酸化反应情形。In a cellular assay, the fusion protein activates the cellular receptor and its downstream signaling molecule phosphorylation. Various human tumor cells were cultured with 25nM target fusion protein at 37 ° C for 60 minutes to collect intracellular protein extracts, and the phosphorylation reaction of cell receptors and their downstream signal molecules was analyzed by Western blotting method.
参照图5,将RGD-EGF及RGD4C-EGF与表达EGFR的MDA-MB468细胞共培养;或将RGD-VEGF及RGD4C-VEGF与表达VEGFR1和VEGFR2的HUVEC血管内皮细胞共培养。分析EGFR(图5A)或VEGFR(图5B)下游的讯息传递分子。RGD-EGF及RGD4C-EGF或RGD-VEGF及RGD4C-VEGF可分别活化EGFR或VEGFR下游的讯息传递途径。结果显示本发明双重标靶分子,不仅具有与相对生物标记分子结合的能力外,大胜肽分子,如EGF或VEGF,仍保有其原本的生物特性。Referring to Figure 5, RGD-EGF and RGD4C-EGF were co-cultured with MDA-MB468 cells expressing EGFR; or RGD-VEGF and RGD4C-VEGF were co-cultured with HUVEC vascular endothelial cells expressing VEGFR1 and VEGFR2. The signaling molecules downstream of EGFR (Fig. 5A) or VEGFR (Fig. 5B) were analyzed. RGD-EGF and RGD4C-EGF or RGD-VEGF and RGD4C-VEGF can activate the signaling pathway downstream of EGFR or VEGFR, respectively. The results show that the double target molecule of the present invention not only has the ability to bind to a relatively biomarker molecule, but a large peptide molecule such as EGF or VEGF retains its original biological properties.
7.放射性标志标靶融合蛋白的制备7. Preparation of radioactive marker target fusion protein
将2-(4-异硫氰酸苯)二乙三胺五乙酸(2-(4-Iso thiocyanatobenzyl)-diethylenetriaminepentaacetic acid,p- SCN-Bn-DTPA)溶于HEPES缓冲溶液中,加入RGD-EGF与RGD4C-EGF等标靶融合蛋白(融合蛋白:p-SCN-Bn-DTPA=1:5~20(摩尔比))于室温下反应1小时。利用以胶体层析法(Sephadax G25)去除未反应的小分子反应物,获得DTPA-RGD-EGF及DTPA-RGD4C-EGF等放射性标志前驱蛋白。加入适量的HEPES缓冲溶液及放射性(111InCl367GaCl390YCl3177LuCl3等)至上述键结DTPA的融合蛋白溶液,于室温下反应1小时。加入过量的EDTA,螯合未结合于蛋白的放射性金属离子,并以薄膜过滤离心法及胶体层析法进行纯化,测量活性并以放射薄层分析法分析产物的放射化学纯度,并分析放射性标志融合蛋白于缓冲溶液(HEPES,4℃)及血清(37℃)的稳定度。参照图6,111In-DTPA-RGD4C-EGF于Hepes缓冲液(4℃)及胎牛血清(37℃)的经时稳定度。将三者(原放射化学纯度皆大于95%)保存于Hepes缓冲液(4℃)及胎牛血清(37℃)中,经24小时后放射化学纯度仍高于90%。2-(4-Iso thiocyanatobenzyl-diethylenetriaminepentaacetic acid, p-SCN-Bn-DTPA) is dissolved in HEPES buffer solution, and RGD-EGF is added. The target fusion protein (fusion protein: p-SCN-Bn-DTPA = 1:5 to 20 (molar ratio)) such as RGD4C-EGF was reacted at room temperature for 1 hour. The unreacted small molecule reactant was removed by colloidal chromatography (Sephadax G25) to obtain a radioactive marker precursor protein such as DTPA-RGD-EGF and DTPA-RGD4C-EGF. An appropriate amount of HEPES buffer solution and radioactive ( 111 InCl 3 , 67 GaCl 3 , 90 YCl 3 and 177 LuCl 3 , etc.) were added to the above-mentioned fusion protein solution bound to DTPA, and reacted at room temperature for 1 hour. Excess EDTA was added to chelate the radioactive metal ions not bound to the protein, and purified by membrane filtration centrifugation and colloidal chromatography. The activity was measured and the radiochemical purity of the product was analyzed by radioactive thin layer analysis. The stability of the fusion protein in buffer solution (HEPES, 4 ° C) and serum (37 ° C). Referring to Figure 6, the stability of 111 In-DTPA-RGD4C-EGF over time in Hepes buffer (4 ° C) and fetal bovine serum (37 ° C). The three (the original radiochemical purity were greater than 95%) were stored in Hepes buffer (4 ° C) and fetal bovine serum (37 ° C), and the radiochemical purity was still higher than 90% after 24 hours.
8.细胞荧光摄影实验8. Cell fluorescence photography experiment
肿瘤细胞或血管细胞(2.5x105cells/well)于12孔细胞培养盘培养中,在37℃下,CO2培养箱中培养一天。去除培养盘中的培养基,以1mL PBS清洗细胞后,分别加入VEGF、RGD-VEGF与RGD4C-VEGF,于37℃下,CO2培养箱中静置2小时。去除培养基后,并以0.5mL PBS清洗3次,于添加anti-His6荧光标志抗体后,直接以荧光显微镜观察并拍摄影像。参照图7,荧光摄影显示RGD-VEGF与RGD4C-VEGF标志重组蛋白可结合至细胞。 Tumor cells or vascular cells (2.5× 10 5 cells/well) were cultured in a 12-well cell culture dish at 37 ° C for one day in a CO 2 incubator. The medium in the culture plate was removed, and the cells were washed with 1 mL of PBS, and then VEGF, RGD-VEGF and RGD4C-VEGF were separately added, and the cells were allowed to stand at 37 ° C for 2 hours in a CO 2 incubator. After the medium was removed, it was washed three times with 0.5 mL of PBS, and after adding an anti-His 6 fluorescent-labeled antibody, the image was directly observed with a fluorescence microscope. Referring to Figure 7, fluorescent photography revealed that RGD-VEGF and RGD4C-VEGF marker recombinant protein can bind to cells.
9.放射性标志重组蛋白的单光子及计算机断层扫瞄(SPECT/CT)造影9. Single photon and computed tomography (SPECT/CT) angiography of radioactive marker recombinant protein
肿瘤小鼠(肿瘤大小约50-100mm3)分别于尾静脉注射500μCi的放射性标志重组蛋白。于注射后1、2、4、8、24、48及72小时进行小动物SPECT/CT造影。在造影结束后以软件处理影像,由影像圈选ROI(region of interest)分析计算肿瘤与肌肉积聚比(tumor to muscle ratio),评估药物于动物活体内的动态积聚变化。参照图8。荷U87MG皮下肿瘤小鼠经尾静脉注射111In-DTPA-EGF、111In-DTPA-RGD-EGF及111In-DTPA-RGD4C-EGF后1、4、8及24小时,进行小动物单光子及计算机断层扫瞄(SPECT/CT)造影。影像显示施打In-111标志重组蛋白后,三者于肿瘤的专一性积聚(以肿瘤/肌肉积聚比值表示)皆随时间而提高,皆于药物注射后8小时达高峰,此时111In-DTPA-RGD4C-EGF的肿瘤/肌肉积聚比为4.4,略高于111In-DTPA-RGD-EGF的3.6,而远高于111In-DTPA-EGF的1.7。Tumor mice (tumor size about 50-100 mm 3 ) were injected with 500 μCi of radiolabeled recombinant protein in the tail vein, respectively. Small animal SPECT/CT angiography was performed at 1, 2, 4, 8, 24, 48 and 72 hours after the injection. After the end of the angiography, the image was processed by software, and the tumor to muscle ratio was calculated by ROI (region of interest) analysis to evaluate the dynamic accumulation of the drug in the living body of the animal. Refer to Figure 8. U87MG subcutaneous tumor mice were injected with 111 In-DTPA-EGF, 111 In-DTPA-RGD-EGF and 111 In-DTPA-RGD4C-EGF in the tail vein for 1, 4, 8 and 24 hours. Computed tomography (SPECT/CT) angiography. The image shows that after the In-111 marker recombinant protein is applied, the specific accumulation of the tumor in the tumor (expressed as the tumor/muscle accumulation ratio) increases with time, reaching a peak at 8 hours after the drug injection, at this time 111 In The tumor/muscle accumulation ratio of -DTPA-RGD4C-EGF was 4.4, slightly higher than 3.6 of 111 In-DTPA-RGD-EGF, and much higher than 1.7 of 111 In-DTPA-EGF.
肿瘤专一性积聚量化分析结果显示,RGD4C-EGF及RGD-EGF融合蛋白皆具双重标靶能力,于肿瘤的积聚明显高于仅具单一标靶能力的EGF重组蛋白;此外,具环状RGD序列的RGD4C-EGF融合蛋白因与integrinαvβ3有较佳的结合能力,使其在肿瘤的积聚略优于具线性RGD序列的RGD-EGF融合蛋白。Quantitative analysis of tumor specific accumulation showed that RGD4C-EGF and RGD-EGF fusion proteins have dual targeting ability, and the accumulation of tumors is significantly higher than that of EGF recombinant protein with only single target ability; The sequence of RGD4C-EGF fusion protein has better binding ability to integrinαvβ3, which makes it accumulate in tumors slightly better than RGD-EGF fusion protein with linear RGD sequence.
所有说明书中所揭示的发明技术特点可以任意方式组合。说明书中揭示的每一技术特点可以提供相同、等同或相似目的的其他方式替换。因此,除非另有特别说明,文中所有揭示的特点均是 等同或相似特点的一般系列的实例。The technical features of the invention disclosed in all of the specification can be combined in any manner. Each of the technical features disclosed in the specification may be replaced by other means that provide the same, equivalent or similar purpose. Therefore, unless otherwise stated, all the features disclosed herein are An example of a general series of equivalent or similar features.
由上述可知,熟习此技艺者能轻易地了解本发明的必要特征,在不脱离其精神与范围之下能就本发明做许多改变与调整以应用于不同用途与条件。 It will be apparent to those skilled in the art that <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt;

Claims (11)

  1. 一种双重标靶药物载体,其特征是,包括第一标靶分子以及第二标靶分子。A dual target drug carrier characterized by comprising a first target molecule and a second target molecule.
  2. 如权利要求1所述的双重标靶药物载体,其特征是,所述第一、第二标靶分子为胜肽或蛋白质。The dual target pharmaceutical carrier according to claim 1, wherein the first and second target molecules are peptides or proteins.
  3. 如权利要求1所述的双重标靶药物载体,其特征是,所述第一或第二标靶分子专一性地结合至肿瘤细胞或/和肿瘤微环境中的血管内皮细胞。The dual target pharmaceutical carrier according to claim 1, wherein the first or second target molecule specifically binds to vascular endothelial cells in tumor cells or/and tumor microenvironments.
  4. 如权利要求1所述的双重标靶药物载体,其特征是,所述第一及第二标靶分子包括,但不限于,精氨酸-甘氨酸-天冬氨酸(RGD)、天冬酰胺-甘氨酸-精氨酸(NGR)、环状NGR、内化RGD(iRGD)、胱氨酸-甘氨酸-天门冬酰氨酸-赖氨酸-精氨酸-苏氨酸-精氨酸-甘氨酸-丙氨酸(CGNKRTRGA)、胃泌素(gastrin)、蛙皮素(bombesin)、奥曲肽(octreotide)或其衍生物。The dual target drug carrier according to claim 1, wherein the first and second target molecules include, but are not limited to, arginine-glycine-aspartate (RGD), asparagine - glycine-arginine (NGR), cyclic NGR, internalization RGD (iRGD), cystine-glycine-aspartate-lysine-arginine-threonine-arginine-glycine - Alanine (CGNKRTRGA), gastrin, bombesin, octreotide or a derivative thereof.
  5. 如权利要求1所述的双重标靶药物载体,其特征是,所述第一及第二标靶分子包括,但不限于,表皮生长因子(EGF)、抗-EGFR抗体、血管内皮生长因子(VEGF)、抗-VEGFR抗体、抗-HER2抗体、肝细胞生长因子受体(HGFR)、抗-HGFR抗体、肿瘤坏死因子(TNF)或抗-TNF抗体。The dual target drug carrier according to claim 1, wherein the first and second target molecules include, but are not limited to, epidermal growth factor (EGF), anti-EGFR antibody, vascular endothelial growth factor ( VEGF), anti-VEGFR antibody, anti-HER2 antibody, hepatocyte growth factor receptor (HGFR), anti-HGFR antibody, tumor necrosis factor (TNF) or anti-TNF antibody.
  6. 如权利要求1所述的双重标靶药物载体,其特征是,所述第一标靶分子以连接符与所述第二标靶分子连结。The dual target drug carrier of claim 1 wherein said first target molecule is linked to said second target molecule by a linker.
  7. 如权利要求5所述的双重标靶药物载体,其特征是,所述连接符为具有5至20个氨基酸的胜肽。 The dual target pharmaceutical carrier according to claim 5, wherein the connector is a peptide having 5 to 20 amino acids.
  8. 如权利要求5所述的双重标靶药物载体,其特征是,所述连接符包括,但不限于,GG、PGGGG或GGGGSGGGGS。The dual target pharmaceutical carrier of claim 5, wherein the connector comprises, but is not limited to, GG, PGGGG or GGGGSGGGGS.
  9. 一种放射性标靶标志蛋白,其特征是,所述放射性标靶标志蛋白由如权利要求1所述的双重标靶药物载体及放射性核种所组成。A radioactive target marker protein, characterized in that the radioactive target marker protein consists of the dual target drug carrier of claim 1 and a radioactive nucleus.
  10. 如权利要求8所述的放射性标靶标志蛋白,其特征是,所述双重标靶药物载体是通过金属螯合剂与所述放射性核种连接。The radiolabeled marker protein according to claim 8, wherein the dual target drug carrier is linked to the radioactive nucleus by a metal chelating agent.
  11. 如权利要求9所述的放射性标靶标志蛋白,其特征是,所述金属螯合剂由DTPA(二乙烯三胺五乙酸,diethylene triamine pentaacetic acid)、NOTA(1,4,7-三氮杂环壬烷,1,4,7-triazacyclononane-N,N',N”-triacetic acid)或DOTA(1,4,7,10-四氮杂环十二烷-1,4,7,10-四乙酸,1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid)各式金属螯合剂及其各类衍生物所组中的一种或多种。 The radioactive target marker protein according to claim 9, wherein the metal chelating agent is composed of DTPA (diethylene triamine pentaacetic acid) and NOTA (1,4,7-triazacyclocycle). Decane, 1,4,7-triazacyclononane-N, N', N"-triacetic acid) or DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-four Acetic acid, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) One or more of a group of various metal chelators and various derivatives thereof.
PCT/CN2016/075329 2016-03-02 2016-03-02 Dual-targeting drug carrier WO2017147820A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/CN2016/075329 WO2017147820A1 (en) 2016-03-02 2016-03-02 Dual-targeting drug carrier
CN201680083086.6A CN108699164B (en) 2016-03-02 2016-03-02 Dual target drug carrier

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2016/075329 WO2017147820A1 (en) 2016-03-02 2016-03-02 Dual-targeting drug carrier

Publications (1)

Publication Number Publication Date
WO2017147820A1 true WO2017147820A1 (en) 2017-09-08

Family

ID=59743389

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/075329 WO2017147820A1 (en) 2016-03-02 2016-03-02 Dual-targeting drug carrier

Country Status (2)

Country Link
CN (1) CN108699164B (en)
WO (1) WO2017147820A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108187064A (en) * 2017-11-28 2018-06-22 南京大学 The preparation method and purposes of double targent fused protein adriamycin couplets of one type elastin laminin-anti-EGFR nano antibodies-iRGD

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113307857B (en) * 2021-01-14 2022-11-01 艾时斌 Scaffold proteins derived from epidermal growth factor, lectin and Tat proteins

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1766115A (en) * 2005-09-06 2006-05-03 中国人民解放军第四军医大学 Preparation method of fusion protein of tumour blood-targeted peptide and human interferon alpha-2b
CN101124243A (en) * 2004-12-23 2008-02-13 莫尔梅德股份有限公司 Conjugation product

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6372172B1 (en) * 1997-12-19 2002-04-16 Kimberly-Clark Worldwide, Inc. Nonwoven webs having improved softness and barrier properties

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101124243A (en) * 2004-12-23 2008-02-13 莫尔梅德股份有限公司 Conjugation product
CN1766115A (en) * 2005-09-06 2006-05-03 中国人民解放军第四军医大学 Preparation method of fusion protein of tumour blood-targeted peptide and human interferon alpha-2b

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108187064A (en) * 2017-11-28 2018-06-22 南京大学 The preparation method and purposes of double targent fused protein adriamycin couplets of one type elastin laminin-anti-EGFR nano antibodies-iRGD
CN108187064B (en) * 2017-11-28 2021-05-28 南京大学 Preparation method and application of elastin-like protein-anti-EGFR nano antibody-iRGD double-targeting fusion protein adriamycin conjugate

Also Published As

Publication number Publication date
CN108699164B (en) 2022-06-07
CN108699164A (en) 2018-10-23

Similar Documents

Publication Publication Date Title
JP7127008B2 (en) Novel PD-L1 binding polypeptides for imaging
KR102397783B1 (en) PET Imaging with PD-L1 Binding Polypeptides
US8299030B2 (en) Peptide-based compounds
JPWO2017217347A1 (en) Site-specific RI-labeled antibody by IgG binding peptide
CA2891190C (en) Aprotinin-derived polypeptide-antibody conjugates
US20160106858A1 (en) Targeting of pharmaceutical agents to pathologic areas using bifunctional fusion polymers
KR20200143366A (en) Modified antibodies and radioactive metal labeled antibodies
WO2011071279A2 (en) Bpb-based cargo delivery system
KR101286721B1 (en) Recombinant albumins fused with poly-cysteine peptide and the methods for preparing the same
TWI602577B (en) Dual targeting drug carrier and application thereof
Chastel et al. Design, synthesis, and biological evaluation of a multifunctional neuropeptide-Y conjugate for selective nuclear delivery of radiolanthanides
WO2017147820A1 (en) Dual-targeting drug carrier
JP6612063B2 (en) Malignant glioma molecule targeting peptide
US11357863B2 (en) Peptide conjugates
US20230203129A1 (en) Anti-her2 polypeptides derivatives as new diagnostic molecular probes
US20130323171A1 (en) Radiolabeled bbn analogs for pet imaging of gastrin-releasing peptide receptors
WO2024046469A1 (en) Cyclic peptide and preparation method therefor, and complex comprising same and use thereof
US20230040008A1 (en) Anti-her2 polypeptides derivatives as new diagnostic molecular probes
US20230365636A1 (en) Bioorthogonal reporter gene system
JP2024506070A (en) Novel HER2 binding polypeptide
CN117327183A (en) Preparation method and application of nuclide-labeled Trop2 specific single-domain antibody probe
TW202014517A (en) Peptide and use thereof

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 16892011

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 16892011

Country of ref document: EP

Kind code of ref document: A1