AU2021104340A4 - Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier - Google Patents

Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier Download PDF

Info

Publication number
AU2021104340A4
AU2021104340A4 AU2021104340A AU2021104340A AU2021104340A4 AU 2021104340 A4 AU2021104340 A4 AU 2021104340A4 AU 2021104340 A AU2021104340 A AU 2021104340A AU 2021104340 A AU2021104340 A AU 2021104340A AU 2021104340 A4 AU2021104340 A4 AU 2021104340A4
Authority
AU
Australia
Prior art keywords
nanocarrier
reversible
ompe
charge
dioleoyl
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
AU2021104340A
Inventor
Luyuan Li
Qiangzhe Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nankai University
Original Assignee
Nankai University
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 Nankai University filed Critical Nankai University
Priority to AU2021104340A priority Critical patent/AU2021104340A4/en
Application granted granted Critical
Publication of AU2021104340A4 publication Critical patent/AU2021104340A4/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • 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
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2123/00Preparations for testing in vivo

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Dispersion Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention discloses a surface charge-reversible nanocarrier for drug delivery based on a novel charge reversible amphiphilic molecule, which can be applied to specifically detect and high- effectively treat of MMP9-expressing tumors. The functional amphiphilic molecule consists of three parts: oleic acid is the hydrophobic region of the molecule, the middle connecting part is a MMP9 responsive cleavage polypeptide (-GPLGLPG-), and the other part is a segment of poly-glutamic acid residues (-GEEEEEG), which constitutes a negatively charged amphiphilic molecule (named as OMPE). The nanocarrier (0-NP) consists of cationic lipid core and OMPE. The cationic lipid core is composed of 1,2-dioleoyl-3 trimethylammonium-propane (DOTAP), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), Soy L-a-phosphatidylcholine (SPC), Cholesterol (CHO), and 1,2-distearoyl-sn-glycero-3 phosphoethanolamine-N-[poly(ethylene glycol)2000] (DSPEPEG2000). After modified by OMPE, the surface charge is reversed to negative charge. Cell and animal experiments show that the nanocarrier can effectively deliver detection probes and anti-tumor drugs, which improve the sensitivity of diagnosis agents and the antitumor effects of drugs in vivo by overcoming their low solubility and/or nonspecific enrichment. So this nanocarrier can be a promising delivery platform for cancer diagnosis and therapy..

Description

Preparation and Application of Matrix Metalloproteinase-9-Responsive
Surface Charge-Reversible Nanocarrier
TECHNICAL FIELD
The invention belongs to the field of nano drugs, and particularly relates to a
surface charge-reversible nanocarrier for drug delivery based on a novel charge
reversible amphiphilic molecule
BACKGROUND
Nanotechnology has attracted increasing attention in the diagnosis and
therapeutics of cancer, largely due to its unique properties, such as improving drug
delivery efficiency, achieving long-term circulation in the body and reducing toxicity.
A large number of nano-drug carriers, such as liposomes, polymer micelles,
dendrimers and other nanotechnology models, have been used in clinical research and
even approved for cancer therapeutics. Due to the leakage characteristics of tumor
blood vessels, nanoparticles of appropriate size can be preferentially enriched in
tumor lesions to effectively provide chemotherapeutic drugs or imaging probes.
However, there are multiple complex biological steps in the effective delivery of the
payload to the tumor site, such as nanocarrier/protein interaction, blood circulation,
extravasation, tumor penetration, and cell uptake. In addition, the properties of
nanocarriers (e.g., surface charge, size, geometry, porosity, and targeting ligands)
have been shown to influence or interact with these biological processes. Compared
with anions, PEGylated cationic nanoparticles showed better inhibition of tumor
growth, but poorer effects on blood circulation time and tumor enrichment.
Nonetheless, smart nanomaterials with sensitive biological responses remain
attractive for cancer diagnosis and therapeutics.
Bio-specific stimulation-responsive nanomaterials are typically triggered by that
microenvironment of the lesion. Bioresponsive nanomaterials generally contain
functional molecules that have specific sensitivity to tumor microenvironment
features, such as factors that respond to specific functions such as enzyme, pH,
hypoxia, redox state, and glucose. Since enzymes are of great significance in many
biological processes, tumor-related enzymes have become new targets for drug
therapy.
Matrix metalloproteinases (MMPs), as a kind of proteolytic enzyme, play a vital
role in all stages of tumor development by degrading the extracellular matrix. MMPs
can regulate apoptosis, angiogenesis, tumor growth, metastasis and many other
aspects, making them become specific biological candidate molecules for malignant
tumors. MMP9 in MMPs can be released and activated as secreted type IV
collagenase/gelatinase in locally inflamed tissues. The expression of MMP9 in tumor
tissues is related to the malignant degree of various cancers including breast cancer,
colon cancer and gastric cancer. At present, the drug delivery systems developed
based on MMPs response are mainly of the following types, such as binding ligands,
enzyme-responsive cleavage prodrugs, MMPs-related effect targets and matrix
degradation. Over-expression of MMPs in various solid tumors makes MMPs
sensitive antibodies or polypeptides become popular target components. Polypeptides,
as targeted functional ligands, are more favoured by people because of their simple preparation method, low cost, low immunogenicity and low opsonin function. In addition, peptides modified by long circulating motifs can overcome the obvious defect of rapid metallization.
At present, it is still difficult to detect tumors with non-invasive, repeatability
and high accuracy during or after clinical therapeutics of tumors, so specific image
detection technology is urgently needed to guide tumor therapeutics. Molecular
imaging plays an increasingly important role in immunotherapy and personalized
medicine. The preparation of molecular probes is the key to molecular imaging. Only
after the molecular probes with high sensitivity and specificity are introduced into the
body, can they specifically bind to specific target molecules in cells and generate
certain signals, which are then collected in vitro by specific imaging devices, such as
positron emission computed tomography (PET-CT), single-photon emission
computed tomography (SPECT), magnetic resonance imaging (MRI), and
chemiluminescent devices, so as to achieve the purpose of specific diagnosis.
Therefore, it is an effective way to solve the above-mentioned problems by adopting
non-invasive methods, rationally designing molecules responding to cancer cells
according to tumor markers, and then developing them into diagnostic reagents and
therapeutic drugs for tumors. In a word, the research and development of nano-drug
delivery carrier with specific targeting/responsiveness for tumors with high
expression of MMP9 will have great breakthrough significance in the diagnosis and
therapeutics of various cancers.
SUMMARY
The technical problem to be solved by the invention is how to provide a surface
charge-reversible nanocarrier for drug delivery based on a novel charge reversible
amphiphilic molecule, which is particularly suitable for microenvironment of MMP9
expressing tumors, such as colorectal cancer, gastrointestinal cancer, bladder cancer,
brain cancer and the like, which can promote the charge reversion of the invention.
According to the technical scheme of the invention, a functional amphiphilic
molecule OMPE is formed by connecting oleic acid and a polypeptide segment,
wherein the amino acid sequence of the polypeptide segment is GPLGLPGGEEEEEG
(SEQ ID No.1), and the structure is as follows: OH 0 H
HO HN
0 0 00 HN 0 HN NH C N NH O 0 OH o o 0 N ~ HHN O H
_NH OH 0 0
The functional amphiphilic molecule consists of three parts: oleic acid is the
hydrophobic region of the molecule, and the intermediate connecting part is a MMP9
responsive cleavage polypeptide, as well as a glutamic acid-rich polypeptide segment,
which constitute negatively charged amphiphilic molecules.
A tumor microenvironment responsive charge-reversible nanocarrier is prepared
from 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 1,2-dioleoyl-sn-glycero
3-phosphocholine (DOPC), Soy L-a-phosphatidylcholine (SPC), Cholesterol (CHO),
distearyl phosphatidyl ethanolamine-polyethylene glycol 2000 with the mass ratio of
m : 4: m : 1: 1 (m =2-4) , the foundation frame with positive charge was formed, and the nano-carrier with negative charge on the surface was formed by OMPE modification.
The preparation method of the tumor microenvironment responsive charge
reversible nanocarrier comprises the following steps:
1) Weighing 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 1,2
dioleoyl-sn-glycero-3-phosphocholine (DOPC), Soy L-a-phosphatidylcholine (SPC),
Cholesterol (CHO), distearyl phosphatidyl ethanolamine-polyethylene glycol 2000
and the OMPE in a mass ratio of m: 4: m: 1: 1/2 (m = 2-4), and dissolving in
trichloromethane/methanol solution;
2) After completely dissolving, rotating and evaporating the solvent at 39 deg°C
to form a uniform film, and removing the residual organic solvent by inert gas;
3) Adding PBS, heating and hydrating in water bath at 60°C for 15 min, and
ultrasonicating in water bath for 15 min to obtain clear suspension; Filtering the
suspension to obtain the tumor microenvironment responsive charge-reversible nano
carrier.
The delivery system is composed of the tumor microenvironment responsive
charge-reversible nanocarrier and the loaded active ingredient described above.
Further, the active ingredient is any one of a chemical drug, a biological drug, a
nano drug, a radiopharmaceutical, a photothermal therapy, a photodynamic therapy
drug or a carrier wrapping these drugs capable of killing cancer cells.
Further, the active ingredient is any one of an alkylating agent, an antimetabolite,
an anti-tumor natural drug, an anti-tumor antibiotic, a hormone, a metal complex or a
tumor radiotargeted marker.
The functional amphiphilic macromolecule derived from the functional
amphiphilic molecule OMPE of the invention is responsively cut off by other matrix
metalloenzymes and the like to generate charge reversion.
The tumor microenvironment responsive charge-reversible nanocarrier of the
present invention derivatizes a functional nanodrug/probe delivery carrier.
Compared with that prior art, the invention has the following beneficial effects:
(1) The invention can be widely applied to diagnosis and therapeutics of tumors
with high expression of MMP9. It can be used for targeted therapy or diagnosis of
various tumors by being conjugated or mixed with a preparation or a detection probe
capable of killing cancer cells.
(2) The amphiphilic functional molecules in the invention can be cut off by the
specific response to reverse the surface charge of the nanocarrier and realize the
efficient delivery of drugs.
(3) The nanocarrier loads the diagnostic reagent or the killer drug to realize high
efficiency tumor diagnosis and therapeutic effect.
(4) The invention has simple preparation method, low economic cost and wide
application prospect. It can be used to monitor the curative effect of drugs and the
corresponding patient's condition in real time. The invention provides a simple, rapid,
economical and accurate detection means for monitoring the recurrence and metastasis of tumor after therapeutics in real time, so as to adjust the treatment scheme in time, carry out clinical intervention as soon as possible, prevent the disease from progressing and improve the prognosis of patients.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is a structure diagram of OMPE;
Figure 2 is an O-NP composition diagram and a characterization diagram;
Figure 3 is a cell uptake diagram of O-NP;
Figure 4 is a drug delivery diagram of O-NP in vivo. Nanocarrier (0-NP)
enhanced specific enrichment of H2S probe in LS180 xenografts. A) Distribution of
Probe 1 encapsulated in the indicated nanocarriers in whole body post 48 h injection.
B) NIR images of retrieved tumors post 48 h treatment with Probe 1 encapsulated in
the indicated nanocarriers. C) Quantification of relative fluorescence intensities of the
tumors (data are mean SD, n = 3; **p <0.01; Student's t-test with two tails)
Figure 5 : Therapeutic outcomes of mice with LS180 xenografts. A)
Photographs of tumors retrieved from experimental animals treated with PBS,
Nanocarrier (0-NP), free DOX, or Nanocarrier (0-NP)-DOX. The animals were
treated seven times, intravenously with the indicated nanoparticles every other day,
and the tumors were retrieved on day 15. B) Growth curves of the LS180 tumors with
the indicated treatments (data are mean SD, n = 5 biologically independent samples;
**p <0.01, ***p <0.001; two-way ANOVA with Dunnett's multiple comparisons
test). C) Typical images of tumor infiltration of DOX (green) with various treatments
via frozen sections. Nuclei were indicated by DAPI (blue). Scale bar, 25 im. D)
Typical images of H&E and Ki67 immunohistochemical staining of the tumors with
various treatments. Scale bar, 100 im.
DESCRIPTION OF THE INVENTION
The technical scheme of the present invention will be further explained by
specific embodiments below. It should be clear to those skilled in the art that the
described embodiments are only to support the present invention, and should not be
regarded as a specific limitation of the present invention.
The methods used in the following examples are all conventional methods unless
otherwise specified.
Materials and reagents used in the following examples are commercially
available unless otherwise specified.
Example 1: Synthesis of amphiphilic molecule OMPE according to the present
invention
(1) Materials
N-methylmorpholine (NMM), piperidine, trifluoroacetic acid (TFA),
dichloromethane (DCM), ninhydrin, vitamin C, phenol, benzotriazole -N,N,N',N'
tetramethylurea hexafluorophosphate (HBTU), diisopropylethylamine (DIPEA),
triisopropylsilane (TIS), N,N- dimethylformamide (DMF), anhydrous ether, Wang
resin, methanol, various Fmoc-protected amino acids, oleic acid, polypeptide
synthesis tube, shaker, vacuum water pump, rotary evaporator. The above reagents
and materials are commercially available.
(2) Preparation of solvent
The reaction solution (act)-n-methylmorpholine: n, n dimethylformamide (1: 24)
Deprotection solvent-piperidine: N, N dimethylformamide (1: 4)
Lysates-trifluoroacetic acid (95%), triisopropylsilane (2.5%) and ultrapure water
(2.5%).
Ninhydrin detection solution-5% ninhydrin: 5% vitamin C: 4g/mL phenol (1: 1:
1).
(3) Synthesis of OMPE
The Fmoc solid-phase peptide synthesis method is adopted to synthesize OMPE,
and the specific method is as follows:
(A) Fmoc-Gly-Wang resin was weighed, placed into a polypeptide synthesis
tube, and 8 mL DMF DMF was added for swelling for 30 min.
(B) Deprotection: adding a deprotection solvent, and performing deprotection at
room temperature for 10 min; ; Washing: DMF and DCM were washed alternately
for 3 times.
(C) Coupling: sequentially weighing the corresponding amino acids with 5 times
of redundancy amount from theC end to the N end according to the sequence,
mixing with HBTU with an equal molar ratio, adding 6 mL of activation reaction
reagent (ACT) for dissolution, adding into the polypeptide synthesis tube, placing on
a shaking table, and reacting at room temperature for 40 min-I h; The N end is
connected with oleic acid for reaction, namely, 10 times of redundant amount of
corresponding amino acid is mixed with HBTU with equal molar ratio, 6 mL of
activation reaction reagent containing 1% DIPEA is added for dissolution, and then the mixture is added into the polypeptide synthesis tube and placed on a shaking bed for reaction overnight at room temperature;
(D) Washing: DMF and DCM are washed alternately for 3 times (the same as the
washing process mentioned above). Deprotection: The deprotection reagent is added
and the mixture is deprotected at room temperature for 10 min; . Washing:DMFand
DCM are washed alternately 3 times each.
At each step of coupling and deprotection, the free amino group is detected by
ninhydrin detection reagent to determine whether the deprotection and coupling are
complete. Deprotection may only be carried out after the test coupling is complete;
Similarly, the coupling reaction of the next amino acid residue cannot proceed until
the deprotection is complete.
Cyclic reaction: according to the above deprotection-washing-coupling-washing
deprotection cycle until all the amino acids are coupled. In each step, the free amino
group can be detected by ninhydrin detection reagent to determine whether the
deprotection and coupling are complete.
Resin shrinkage: the resin is washed after synthesis of the target sequence.
Adding methanol into that polypeptide synthesis tube for washing, contracting and
draining.
Transfer the prepared OMPE- resin to a 10 mL brown bottle, add 5 mL of lysis
buffer, place on ice, and lyse under mild stirring in the dark for 3 h. Subsequently, that
polypeptide lysate is evaporate under the condition of vacuum rotation at 42C until
the lysate is basically volatilize; Precipitate the crude product in a flask with 10 mL of pre-cooled ether, which was centrifuged several times. Vacuum dry the crude product of OMPE, and store the dried crude product at -20 °C for standby.
Figure 1 is a structure diagram of OMPE. The structural sequences are as
follows: Oleic acid-gplglp-ggeeeeeeg; The results of high performance liquid
chromatography and time-of-flight mass spectrometry confirme that the synthesized
OMPE could be used for subsequent experiments.
Example 2 construction of functional nanocarrier O-NP
(1) Materials
1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 1,2-dioleoyl-sn-glycero
3-phosphocholine (DOPC), Soy L-a-phosphatidylcholine (SPC), Cholesterol (CHO),
1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[poly(ethylene glycol)2000]
(DSPEPEG2000),Methanol, Trichloromethane, Phosphate Buffer Salt Solution (PBS).
(2) Preparation of O-NP
The nanocarrier O-NP is prepared by a film dispersion method, and that specific
steps are as follow:
Accurately weigh 1mg of DOTAP, 2 mg of DOPC, 1 mg of SPC, 0.5 mg of
DSPE-PEG 2000, 0.5 mg of CHO, and 0.5 mg of OMPE in 5 mL of solvent
(trichloromethane/methanol, 2: 1 by volume) and place in a round-bottomed flask
after complete dissolution. At 39C. The rotary evaporator is slowly and uniformly
rotated to volatilize the organic solvent, and a uniform film is formed and apply to the
flask wall. Remove the residual organic solvent from the flask with inert gas. Then 5
mL PBS is added, heated and hydrated in a 60°Cwater bath for 15 min, following by water-bath ultrasound for 15 min to obtain a clear suspension. The prepared samples are filtered (0.224m) three times to obtain sample O-NP (1 mg/mL) and stored at 4C for subsequent use.
Placed 1 mL of nanoparticle suspension O-NP (1 mg/mL) in a Dynamic Light
Scattering (DLS) sample cell, and the size of O-NP and its surface potential (Q) can be
measured by a DLS granulometer. The test is repeated three times at the test
temperature of 25C, laser wavelength of 633 nm, probe angle of 173, and sample
equilibration time of 2 min. Experimental data are processed and analysed by
software Zetasizer Software.
The morphology of nanoparticles is observed by electron microscope (TEM).
Take 8 L of O-NP (1 mg/mL) and drop it on the copper mesh supported by common
carbon membrane. After 2 min, absorb the excess sample with filter paper. Then drop
6 L of 1% phosphotungstic acid for negative dyeing for 5 min. Absorb the excess
liquid with filter paper. Finally, wash it with water once and put it in a drying oven for
later use. Under the voltage of 200 kV, it is observed by biological transmission
electronmicroscope TalosTF200C (TEM, FEI, USA)
Example 3 Uptake of O-NP by cells
(1) Preparation of imaging agent (0-NP-Cy2): According to the preparation
method of O-NP, 1 mg DOTAP, 2 mg DOPC, 1 mg SPC, 0.5 mg DSPE-PEG2000, 0.5
mg CHO, 0.5 mg OMPE and 0.5 mg Cy2 are accurately weighed and dissolved in 5
mL solvent (chloroform/methanol, volume ratio 2:1). At the temperature of 39C, the
rotary evaporator rotates slowly and evenly. After the organic solvent volatilizes, a uniform film is formed and attached to the flask wall. The residual organic solvent in the flask Is removed with inert gas. Adding 5 mL of PBS, heating and hydrating in 60
°C water bath for 15 min, and ultrasonic treatment in water bath for 15 min, a clear
suspension is obtained. The prepared sample is filtered (0.22 m) for three times to
obtain sample O-NP (1 mg/mL), which is stored at 4C for later use.
(2) Cell culture: Human colorectal cancer cell LS180 and human breast cancer
cell Hela are cultured in a 5% carbon dioxide incubator at 37Cwith high glucose
DMEM containing 10% fetal bovine serum and 1% double antibodies (100 U/mL
penicillin and 100 g/mL streptomycin).
(3) O-NP-Cy2 interacts with cells and cell uptake is observed.
Hela cells are incubated at a cell concentration of 1x105/mL, with 1 mL of cell
suspension in a circular petri dish (35 mm) at 37C in a 5% C02 cell incubator. After
12 h, discarding the culture solution, then the petri dishes are incubated with different
concentrations of exogenous MP9 (0, 1, or 10 ng/mL) for 1 h, following by 0-NP
Cy2(30 [g/mL, 1 mL) for 2 h. Then washed twice with pre-cooled 1xPBS. The cells
are incubated with Hoechst 33342 containing 10 g/mL for 10 min at 37 C, and
washed twice with pre-cooled 1xPBS. The fluorescence distribution in the cells is
detected by a laser scanning confocal microscope (ZEISS TCS SP8).
As shown in Part A of Figure 3, the addition of exogenous MP9 promoted the
entry of O-NP into cells, and in particular, 10 ng/mL MMP9 significantly promoted
the entry of O-NP into Hela cells.
Hela and LS180 are incubated 1 mL of the cell suspension at a cell concentration
of 1x105/mL in a round petri dish (35 mm) at 37C in a 5% C02 cell incubator for 12
h, discarding the culture medium and incubating with the MMP9 inhibitor 3B-3CT (0
or 20 g/mL) for 30 min following by 0-NP-Cy2 (30 [g/mL, 1 mL) for 2 h. Then
washed twice with pre-cooled 1xPBS. The cells are incubated with Hoechst 33342
containing 10 g/mL for 10 min at 37C, and washed twice with pre-cooled 1xPBS.
The fluorescence distribution in the cells is detected by a laser scanning confocal
microscope (ZEISS TCS SP8).
As shown in part B of Figure 3, the high expression of MMP9 in tumor cell line
LS180 results in higher uptake of O-NP than in Hela cells. The addition of inhibitor
3B-3CT could significantly inhibit the uptake of O-NP by cells.
Example 4 Enrichment of O-NP in solid tumor
(1) The experimental animals are female BALB/c nude mice with the age of 6-8
weeks, which were purchased from Unilever. The animal experiments are conducted
in an environment free of specific pathogenic microorganisms. All animal
experiments are conducted in accordance with the Guidefor LaboratoryAnimal Care
and Use issued by Tianjin Science and Technology Commission.
(2) Preparing O-NP-Cy7. Referring to the preparation method of O-NP-Cy2, 0.5
mg of Cy2 was replaced by an equal mass of Cy7 to obtain sample O-NP-Cy7 (1
mg/mL) which was stored at 4C for later use.
(3) Establishing animal model of tumor in vivo.
1x108/mL (100 L) of Hela cells are subcutaneously inoculated in the left thigh
of mice, and 5x107/mL(100 L) of LS180 cells are subcutaneously inoculated in the
right thigh. O-NP-Cy7(1 mg/mL, 200 L) is given by tail vein when the tumor size
was about 150 mm3, corresponding to 200 ng Cy7. Six hours after administration,
IVIS Lumina II (Xenogen/Perkin Elmer); The 710 nm excitation is observed by IVIS
Lumina II (Xenogen/Perkin Elmer), and ICG transmitted and filtered to receive
signals.
Figure 4 shows that the LS180 tumor had a higher residual uptake of O-NP than
the Hela tumor tissue.
In summary, from experimental examples 1to 4, it can be concluded that the
nanocarrier of the present invention has the characteristic of specific charge reversion
to increase the drug uptake efficiency by cell. In practical application, the nanocarrier
of the present invention can be conjugated or mixed with a preparation capable of
killing or indicating cancer cells as a functional delivery system for targeted therapy
or imaging of tumors.

Claims (6)

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. The functional amphiphilic molecule OMPE is formed by connecting oleic
acid and a polypeptide segment, the amino acid sequence of the polypeptide segment
is GPLGLPGGEEEEEG, and the structure is as follow: OH
HOO
HO I NH
0 0 0 0 HN
N NH OH H
NHN O 0 N NH 0 OH
00
2. The tumor microenvironment responsive charge-reversible nanocarrier
consists of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 1,2-dioleoyl-sn
glycero-3-phosphocholine (DOPC), Soy L-a-phosphatidylcholine (SPC), Cholesterol
(CHO), and distearyl phosphatidyl ethanolamine-polyethylene glycol 2000 in a mass
ratio of m: 4: m: 1: 1, m = 2-4, and forms a basic frame with positive charge, and the
nanocarrier with negative surface charge is formed after modification by OMPE as
claimed in claim 1.
3. The preparation method of the tumor microenvironment responsive charge
reversible nanocarrier according to claim 2, which is characterized in that the steps are
as follows:
1) Weighing 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 1,2
dioleoyl-sn-glycero-3-phosphocholine (DOPC), Soy L-a-phosphatidylcholine (SPC),
Cholesterol (CHO), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-
[poly(ethylene glycol)2000] (DSPEPEG2ooo) and OMPE of claim 1in a mass ratio of
m: 4: m: 1: 1/2, m= 2-4, and dissolving in trichloromethane/methanol solution;
2) After complete dissolution, rotary evaporation is carried out at 39C to form a
uniform film, and residual organic solvent is removed by inert gas;
3) PBS is added, and the mixture is hydrated by heating in a 60°C water bath for
min, following by water-bath ultrasound for 15 min to obtain a clear suspension;
Filter that suspension agent by a filter membrane to obtain the tumor
microenvironment responsive charge-reversible nanocarrier.
4. A delivery system comprising the tumor microenvironment responsive
charge-reversible nanocarrier and a loaded active ingredient according to claim 2.
5. The delivery system according to claim 4, which is characterized in that the
active ingredient is any one of a chemical drug, a biological drug, a nano drug, a
radiopharmaceutical, a photothermal therapy, a photodynamic therapy drug, or a
carrier encapsulating these drugs that can kill cancer cells.
6. The delivery system according to claim 4, which is characterized in that the
active ingredient is any one of an alkylating agent, an antimetabolite, an antineoplastic
natural drug, an antineoplastic antibiotic, a hormone, a metal complex or a tumor
radiotargeted marker.
AU2021104340A 2021-07-20 2021-07-20 Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier Active AU2021104340A4 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2021104340A AU2021104340A4 (en) 2021-07-20 2021-07-20 Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
AU2021104340A AU2021104340A4 (en) 2021-07-20 2021-07-20 Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier

Publications (1)

Publication Number Publication Date
AU2021104340A4 true AU2021104340A4 (en) 2021-09-16

Family

ID=77666612

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021104340A Active AU2021104340A4 (en) 2021-07-20 2021-07-20 Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier

Country Status (1)

Country Link
AU (1) AU2021104340A4 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115282273A (en) * 2022-07-19 2022-11-04 山西大学 Preparation method and application of multifunctional organic anti-tumor nano self-assembly

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115282273A (en) * 2022-07-19 2022-11-04 山西大学 Preparation method and application of multifunctional organic anti-tumor nano self-assembly

Similar Documents

Publication Publication Date Title
Hu et al. Regulating cancer associated fibroblasts with losartan-loaded injectable peptide hydrogel to potentiate chemotherapy in inhibiting growth and lung metastasis of triple negative breast cancer
Seo et al. Self-assembled 20-nm 64Cu-micelles enhance accumulation in rat glioblastoma
WO2021147585A1 (en) Use of chitosan modified by fluorine-containing compound as drug carrier and preparation method therefor
Zhu et al. Targeted delivery of methotrexate to skeletal muscular tissue by thermosensitive magnetoliposomes
Tesarova et al. Taking advantage of cellular uptake of ferritin nanocages for targeted drug delivery
CA2982853C (en) Texaphyrin-phospholipid conjugates and methods of preparing same
Ren et al. MRI-visible liposome nanovehicles for potential tumor-targeted delivery of multimodal therapies
An et al. A sulfur dioxide polymer prodrug showing combined effect with doxorubicin in combating subcutaneous and metastatic melanoma
CN108653733B (en) Polymer vesicle of double-loaded anthracycline drug and photosensitizer with bubble generation function and preparation method thereof
CN107184987B (en) Lipoic acid modified targeted integrin alpha v beta 3 nano-polypeptide carrier and preparation method and application thereof
Liu et al. Theranostic polymeric micelles for the diagnosis and treatment of hepatocellular carcinoma
KR20180120220A (en) Biodegradable amphiphilic polymers specifically targeting ovarian cancer, polymeric vesicle made therefrom and uses thereof
WO2017063542A1 (en) Stabilized a7r polypeptides, and use thereof in constructing tumor targeted therapeutic drug delivery system
CN112773766A (en) Liposome delivery system for tumor treatment and preparation method and application thereof
CN105434347A (en) Polypeptide nano-micelle, and preparation method and application thereof
CN105534896A (en) Polypeptide and chemotherapy drug combined drug-loaded micelle and preparation method and application thereof
AU2021104340A4 (en) Preparation and Application of Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier
CN107550864B (en) EPPT polypeptide-polyethylene glycol-phospholipid composite membrane material, preparation method thereof, active targeting liposome drug delivery system and application
CN112999359A (en) Tumor-targeted redox-response prodrug nano preparation and preparation method and application thereof
CN102429868A (en) Liposome medicinal composition with tumor targeting, in-vivo tracing and treating functions and preparation method thereof
CN113144172B (en) Preparation method of liposome containing vancomycin, IR780 and oxygen-carrying perfluorohexane
CN105770912B (en) Load medicine ATP sensitive liposome with tumour near-infrared fluorescent image displaying function and preparation method thereof
CN104045823A (en) Glycyrrhetinic acid derivative, and preparation method and application thereof
CN111494641B (en) Tumor microenvironment responsive surface charge reversible nano-drug delivery vehicle
CN112661812B (en) KK-LC-1 antigen targeted binding peptide, derivative, probe and application thereof

Legal Events

Date Code Title Description
FGI Letters patent sealed or granted (innovation patent)