CN114010635A - New application of nilapanib and derivatives thereof - Google Patents

New application of nilapanib and derivatives thereof Download PDF

Info

Publication number
CN114010635A
CN114010635A CN202111381376.8A CN202111381376A CN114010635A CN 114010635 A CN114010635 A CN 114010635A CN 202111381376 A CN202111381376 A CN 202111381376A CN 114010635 A CN114010635 A CN 114010635A
Authority
CN
China
Prior art keywords
nilapanib
derivative
restenosis
expression
pdgfr
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.)
Pending
Application number
CN202111381376.8A
Other languages
Chinese (zh)
Inventor
王成
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.)
Tongji Medical College of Huazhong University of Science and Technology
Original Assignee
Tongji Medical College of Huazhong University of Science and Technology
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 Tongji Medical College of Huazhong University of Science and Technology filed Critical Tongji Medical College of Huazhong University of Science and Technology
Priority to CN202111381376.8A priority Critical patent/CN114010635A/en
Publication of CN114010635A publication Critical patent/CN114010635A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Abstract

The invention belongs to the technical field of medicines, and particularly relates to a novel application of nilapanib and a derivative thereof. Application of nilapanib or a derivative thereof in preparation of a preparation for inhibiting PDGFR beta expression and/or PDGFR beta downstream signaling pathway. Application of nilapanib or a derivative thereof in preparing a preparation for inhibiting smooth muscle phenotype switching. Application of nilapanib in preparing medicine for preventing and treating vascular diseases. The concentration of the nilapanib in the medicine for preventing and treating vascular diseases is 0.5 mu M or 20 mg/kg/d. An agent for inhibiting PDGFR β expression and/or PDGFR β downstream signaling pathway, comprising nilapanib or a derivative thereof as an active ingredient. Application of nilapanib or a derivative thereof in preparing a preparation for promoting alpha SMA gene expression. The nilapanib provided by the invention can be used as a PAPR1/2 inhibitor and has an inhibitory effect on PDGFR beta, so that the nilapanib can inhibit smooth muscle phenotype transition, and can prevent and/or treat restenosis after vascular injury.

Description

New application of nilapanib and derivatives thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a novel application of nilapanib and a derivative thereof.
Background
Nilapanib, also known as zejua, MK-4827, Niraparib, jele, nilapali; CAS: 1038915-60-4; the chemical name is dis 2- {4- [ (3S) -piperidine-3-yl ] phenyl } -2Hindazole 7-carboxamide 4-methyllbenzenesulfonate hydrate (1:1: 1). Is an organic matter with a chemical formula of C26H30N4O5S, white to off-white, and does not absorb water. The molecular weight was 510.61 amu. The structural formula is shown in fig. 9.
Nilaparib has been studied mainly as a PARP inhibitor, i.e., a selective PARP1/2 inhibitor, in the current state. The Niraparib is combined with nicotinamide adenine dinucleotide + (nicotinamide adenine dinucleotide +, NAD +) of PARP1 and/or PARP2 in a highly selective manner, so that the separation of PARP and DNA is prevented, cancer cells cannot be subjected to subsequent DNA repair, a large amount of genome damage (genomic damage) is accumulated in the tumor cells, the tumor cells are self-destroyed (self-destruct), and finally the purpose of treating tumors is achieved. In vitro studies show that niraparib-induced cytotoxicity may be involved in the inhibition of PARP enzyme activity and in the increased formation of PARP-DNA complexes leading to DNA damage, apoptosis and cell death. Increased niraparib-induced cytotoxicity was observed in BRCA1/2 tumor cell lines with or without defects. Xenograft models of mouse tumor cells deficient in BRCA1/2 and Niraparib, a model of derived transplantable tumors in patients with homologous recombination or mutations or wild-type BRCA1/2 deficiency in humans, reduced tumor growth. Therefore, the above drugs are widely used in recurrent epidermal ovarian, fallopian tube, or primary peritoneal cancer patients as a maintenance therapy for platinum-based chemotherapy in patients with complete or partial remission in adult patients.
In recent years, the incidence of coronary heart disease in China is on the rise. Coronary heart disease is a vascular disease with vascular stenosis caused by atherosclerosis as the main pathological process. Interventional therapy and coronary artery bypass grafting effectively improve many clinical symptoms of complex and critical coronary heart disease, however, restenosis at the site of vascular disease remains one of the major clinical problems that plague today. In the pathological process of restenosis after atherosclerosis and angioplasty, endothelial cells are damaged under the action of factors such as pressure and inflammation, so that abnormal proliferation of vascular smooth muscle cells is caused, and the vascular smooth muscle cells are transformed from contractile smooth muscle cells to secretory smooth muscle cells, thereby thickening the vascular wall and narrowing the lumen. Under pathological conditions such as hypertension, vascular smooth muscle is subjected to different shearing forces to generate phenotypic transformation, so that vascular remodeling is caused.
Vascular smooth muscle cell phenotypic transformation is an important pathophysiological process mediating membranous calcification in blood vessels. In normal vascular tissue, Vascular Smooth Muscle Cells (VSMCs) exhibit a contractile phenotype, are highly differentiated cells, and have the primary functions of maintaining the morphology of blood vessels and regulating vasoconstriction and tone, with characteristics of low proliferation, low migration, and low secretion. Major marker proteins such as smooth muscle alpha actin (SM α actin), smooth muscle 22 α, smooth muscle myosin heavy chain, etc. Various cardiovascular risk factors become inducers for driving Vascular Smooth Muscle Cell (VSMC) phenotype transdifferentiation, and can induce smooth muscle cell transdifferentiation from a contractile type (differentiation type) to a synthetic type (dedifferentiation type), and the cells have the characteristics of high proliferation, high migration and high protein secretion. Research shows that VSMCs phenotype transformation is a key step for the occurrence and development of a plurality of vascular diseases such as atherosclerosis, hypertension, restenosis after angioplasty and the like.
The vascular restenosis is a complex pathological process, effectively solves the clinical problems of the vascular restenosis, and can greatly expand the application of angioplasty, thereby better applying the blood flow re-transport operation to patients with vascular proliferative diseases and the like and maintaining the health of human beings. At present, there are several drugs used in scientific research to inhibit restenosis after vascular injury, among which the most common are: anti-inflammatory drugs (such as dexamethasone and estrogen), anti-cell proliferation agents (such as rapamycin, sirolimus and paclitaxel), Angiotensin Converting Enzyme (ACE) inhibitors cilazapril and spirapril, and coating drugs [ such as antithrombotic agents (such as hirudin and heparin) used in drug-coated stents (DES) ]. Previous studies have shown that Niraparib can exert therapeutic effects in tumor diseases by promoting apoptosis, and its role in restenosis after vascular injury has not been studied.
Disclosure of Invention
Aiming at the problems, the invention provides new application of the nilapanib and the derivatives thereof, mainly provides a new treatment scheme aiming at smooth muscle degeneration, develops new application of the nilapanib and expands application prospect of the nilapanib.
In order to solve the problems, the invention adopts the following technical scheme:
application of nilapanib or a derivative thereof in preparation of a preparation for inhibiting PDGFR beta expression and/or PDGFR beta downstream signaling pathway.
Application of nilapanib or a derivative thereof in preparing a preparation for inhibiting smooth muscle phenotype switching.
In some forms, the smooth muscle phenotype transitions to a contractile phenotype to a synthetic phenotype.
Application of nilapanib or a derivative thereof in preparing a medicament for preventing and treating vascular diseases.
In some forms, vascular disease includes tunica media calcification, atherosclerosis, hypertension, endothelial cell hyperproliferation due to vascular injury, and vascular restenosis.
In some embodiments, the restenosis includes restenosis following vascular injury.
In some embodiments, restenosis after vascular injury comprises PCI restenosis, in-stent restenosis, or restenosis after bypass grafting.
In some modes, the concentration of the nilapanib effect in the vascular disease prevention and treatment drug is 0.5-5 μ M or 5-20 mg/kg/d; preferably, the concentration used is 0.5. mu.M or 20 mg/kg/d.
An agent for inhibiting PDGFR β expression and/or PDGFR β downstream signaling pathway comprising nilapanib or a derivative thereof as an active ingredient.
Application of nilapanib or a derivative thereof in preparing a preparation for promoting alpha SMA gene expression.
The invention has the beneficial effects that:
nilaparib has an inhibitory effect on PDGFR beta, so that Niraparib can inhibit smooth muscle phenotype switching, thereby preventing and/or treating restenosis after vascular injury. Furthermore, nilapanib has no cytotoxic effect typical of cardiovascular drugs or other inhibitors of smooth muscle phenotype switch, and therefore its safety can be expected. It can be widely used as a medicine for treating related diseases involving smooth muscle, such as vascular restenosis, tumor and the like.
Drawings
In fig. 1, a is a group of a pseudo operation and a carrier treatment, b is a group of a ligation and a carrier treatment, c is a group of a ligation and a nilapani treatment, and the morphology of a blood vessel of a rat after carotid artery ligation is detected by an oil red-Hematoxylin (HE) staining experiment;
FIG. 2 shows VSMC proliferation assay using EdU assay with varying concentrations of Niraparib and vehicle (DMSO);
FIG. 3 shows stimulation of VSMC with Niraparib (0.5,1, 5. mu.M) and vehicle (DMSO) following PDGF-BB administration. In the figure, the expression of cyclin and oncostatin is detected by using a PCR (polymerase chain reaction) experiment after the Niraparib and the carrier with different concentrations, and the cyclin expression is increased and the oncostatin expression is reduced after the VSMC is treated by PDGF-BB; after the Niraparib is given for stimulation, the cyclin expression is reduced, and the cancer suppressor protein expression is increased;
FIG. 4 shows stimulation of VSMC with Niraparib (0.5,1, 5. mu.M) and vehicle (DMSO) following PDGF-BB administration. In the figure, the expression and the activity of Caspase 3 are detected by the Niraparib and the carrier with different concentrations, and no obvious change is seen;
FIG. 5 shows stimulation of VSMC with Niraparib (0.5,1, 5. mu.M) and vehicle (DMSO) following PDGF-BB administration. In the figure, Niraparib with different concentrations is used for detecting the migration condition of cells by using a transwell experiment;
FIG. 6 shows stimulation of VSMC with Niraparib (0.5,1, 5. mu.M) and vehicle (DMSO) following PDGF-BB administration. In the figure, Niraparib with different concentrations is used for detecting the proliferation and migration of cells by using a scratch experiment;
in FIG. 7, after the carotid artery of the rat is ligated, the rat is treated with a vector and 20mg/kg dNairaparib, and the Niraparib with different concentrations is used for detecting the expression of the contractile gene alpha SMA by an immunofluorescence experiment;
FIG. 8 shows stimulation of VSMC with Niraparib (0.5,1, 5. mu.M) and vehicle (DMSO) following PDGF-BB administration. In the figure, the expression level of PDGFR beta is detected by using western blot experiments for Niraparib with different concentrations;
FIG. 9 is a Nilaparib structural formula.
Detailed Description
The term is defined as:
the term "nilapani derivative"The nilapanib derivative is a compound which is different from nilapanib in partial position structure and functional group change, but the structure and functional group change of the positions keeps the nilapanib derivative and the nilapanib derivative still have the same similar control and regulation effects on the diseases related to the invention. For example, derivatives of nilapanib moiety-H or functional group-NH 2, altered, are intended to be within the scope of the present invention as long as they are capable of modulating the conditions to which the present invention relates. In some aspects described below, the nilapanib derivative is intended to be equivalent in scope to the nilapanib derivative in substantially the same effect as nilapanib for the treatment of the same condition, although the mechanisms involved are substantially the same and are intended to fall within the scope of the corresponding claims.
The term "control"Refers to the medical management of a patient with the intent to cure, ameliorate, stabilize or prevent a disease, pathological condition or disorder. The term includes active treatment, i.e., treatment specifically directed to the amelioration of a disease, pathological condition, or disorder, and also includes causal treatment, i.e., treatment directed to the removal of the cause of the associated disease, pathological condition, or disorder. In addition, the term also includes palliative treatments, i.e., treatments designed to alleviate symptoms rather than cure a disease, pathological condition, or disorder; prophylactic treatment, i.e. treatment aimed at minimising or partially or completely inhibiting the development of the relevant disease, pathological condition or disorder; and supportive treatment, i.e. treatment for supplementing another specific therapy directed to an improvement of the relevant disease, pathological condition or disorder. Meanwhile, in commercial activities, if a product containing the same components as the present invention is produced, the product specification does not describe the same similar use as the present invention, but it should be regarded as an application when it gives a hint that the corresponding product is used for achieving the same purpose as the present invention.
The definitions of the terms used herein are to be construed as being given their ordinary meaning in the art unless otherwise indicated. In the absence of counterexamples, no diminutive explanation is made, i.e., except for the case where evidence of counterexamples exists, the rest should fall within the scope.
The invention is further illustrated below:
a first aspect of this section relates to:
application of nilapanib or a derivative thereof in preparation of a preparation for inhibiting PDGFR beta expression and/or PDGFR beta downstream signaling pathway.
The purpose of inhibiting the expression of PDGFR β is mainly to keep it at a normal level and to prevent it from being abnormally changed to cause lesions. The purpose of inhibiting the expression of the PDGFR β downstream pathway is also to keep it at a normal level and prevent its abnormal changes from causing lesions. Wherein the normal level in this paragraph is medically indicated as a medical reference value, which is subject to a decrease in inhibition when the expression is ultrahigh. However, it is within the scope of the invention if it is used for some extreme purposes. For example, in some environments, when PDGFR β expression needs to be controlled to be greatly reduced and PDGFR β downstream pathway expression needs to be controlled to the maximum extent; for another example, PDGFR β expression is prevented from increasing to maintain a certain level, and PDGFR β downstream pathway expression is maintained at a certain level.
When the polypeptide is used in other ways, the polypeptide can also be directly added to a substance to be regulated, for example, in commercial research experiments, when the polypeptide is used as a PDGFR beta expression and/or PDGFR beta downstream pathway expression regulating reagent, the polypeptide can be directly added to relevant cell sap to achieve the same basic purpose of the invention, and the polypeptide and the cell sap are also within the scope of the invention.
The PDGFR beta downstream pathway expression inhibitor related to the invention is not limited to the specific application mode, and also belongs to the relevant content of the aspect when the inhibitor is used for preparing medicines for treating diseases related to, for example, PDGFR beta downstream pathway overexpression triggering.
In some embodiments, the agent that inhibits PDGFR β expression and/or PDGFR β downstream pathway expression is present at a concentration of 0.5 to 5 μ M or 5 to 20 mg/kg/d; preferably, the concentration used is 0.5. mu.M or 20 mg/kg/d. Wherein the action concentration is used for treatment, the specific concentration parameter (content index) refers to the concentration of effective components in the conventional medicine, and the effective component can be made into preparation or medicine granule, and the application method is oral administration or injection.
A second aspect of this section relates to:
application of nilapanib or a derivative thereof in preparing a preparation for inhibiting smooth muscle phenotype switching.
In some forms, the smooth muscle phenotype transitions to a contractile phenotype to a synthetic phenotype.
Whether vascular smooth muscle cells can maintain the contractile phenotype is a key link in determining vascular homeostasis and remodeling. Inhibition of smooth muscle to other phenotypes can cause some pathologies.
In some embodiments, the smooth muscle phenotype switch comprises a switch from contractile to secretory. The transition from systolic to secretory is inhibited by nilapanib, particularly nilapanib.
A third aspect of this section relates to:
application of nilapanib or a derivative thereof in preparing a medicament for preventing and treating vascular diseases.
In some embodiments, vascular diseases including tunica media calcification, atherosclerosis, hypertension, endothelial cell hyperproliferation caused by vascular injury, and vascular restenosis, and cardiovascular disorders caused by other proximate pathogenic mechanisms are also within the scope of the present invention.
In some embodiments, the restenosis includes restenosis following vascular injury.
In some embodiments, restenosis following vascular injury includes PCI restenosis, in-stent restenosis, restenosis following bypass grafting, and other conditions with similar pathogenesis.
Can be used for preventing and treating restenosis after vascular injury. If aiming at some patients who have coronary stent operation and the like, the nilapanib can also play a role in subsequent adjuvant therapy, and further play a role in preventing restenosis of blood vessels; adjunctive therapeutic agents similar to these should also be within the scope of the invention.
In some modes, the concentration of the nilapanib effect in the vascular disease prevention and treatment drug is 0.5-5 μ M or 5-20 mg/kg/d; preferably, the concentration used is 0.5. mu.M or 20 mg/kg/d.
A fourth aspect of this section relates toAnd:
an agent for inhibiting PDGFR β expression and/or PDGFR β downstream signaling pathway, comprising nilapanib as an active ingredient.
The nilapanib derivative is a compound that has a partial functional group and a carbon chain length that varies from that of nilapanib.
The inhibitor is a mixture containing Nilaparib and other drugs; wherein the effective action concentration of the nilapanib in the inhibitor on the corresponding mechanism is 0.5 mu M or 20 mg/kg/d.
Taking nilapanib or a derivative thereof as an example, the inhibitor comprises at least one of nilapanib or a derivative thereof:
one of the inhibitors contains the effective component of Nilaparib and other auxiliary drugs, thereby forming a novel pharmaceutical composition with inhibitory effect on PDGFR beta expression and/or PDGFR beta downstream pathway expression.
In the second aspect, the inhibitor contains an active ingredient nilapanib derivative and other auxiliary drugs, thereby forming a novel pharmaceutical composition having an inhibitory effect on PDGFR β expression and/or PDGFR β downstream pathway expression.
Therefore, the nilapanib or the derivative thereof is mixed with other components to form a novel preparation and a novel medicine for preventing and treating related diseases.
A fifth aspect of this section relates to:
application of nilapanib or a derivative thereof in preparing a preparation for promoting alpha SMA gene expression. The promoting effect is mainly determined according to the treatment purpose, and can be expressed as exceeding the normal level or maintaining the normal level.
The sixth aspect of this section is described in conjunction with specific items:
the Niraparib used in the tests of this example was purchased from seleck, catalog No.: s2741 alias: MK-4827. The phenotypic switching of VSMC in the following experiments was examined using the methods in the following steps, respectively.
Experiment one
After carotid artery ligation injury or sham-operated injury in C57BL/6 rats, Niraparib (20 mg/kg. d) and vehicle (DMSO) were intraperitoneally injected, respectively. After 14 days, the mice were euthanized and the injured blood vessels were subjected to a vascular resection procedure. After paraffin is fixed and embedded by formaldehyde with the mass concentration of 4%, the blood vessel is cut into sections. FIG. 1 shows the detection of the vascular morphology of a rat carotid artery after ligation by using an oil red-Hematoxylin (HE) staining experiment after treatment of a sham operation + carrier treatment group, a ligation + carrier treatment group and a ligation + Nilaparib treatment group. The results are shown in FIG. 1. Wherein, the figure 1 is a statistical chart of the average thickness of the carotid artery of a rat in a sham operation + carrier treatment group, a ligation + carrier treatment group and a ligation + nilapani treatment group from left to right. As can be seen from fig. 1, when rats were subjected to carotid artery ligation, the carotid artery was increased in thickness, i.e., vascular injury resulted in excessive proliferation of vascular endothelial cells, compared to the sham surgery + vehicle treatment group; and the nilapanib can inhibit the excessive proliferation of the endothelial cells in the blood vessels caused by the blood vessel injury.
Experiment two
VSMC cells were treated with PDGF-BB (30ng/ml) and stimulated with different concentrations of Niraparib (0.5. mu.M, 1. mu.M, 5. mu.M) and vehicle DMSO, respectively. FIG. 2 shows VSMC proliferation assay using EdU assay for varying concentrations of Niraparib. In FIG. 2, the first column is the DAPI stained nuclei, the second column is the EdU stained nuclei, and the third column is the fusion map of the first two columns; while the first row was a vector treated group, the second row was a PDGF-BB and vector treated group, the third row was a PDGF-BB and 0.5. mu.M Niraparib treated group, the fourth row was a PDGF-BB and 1. mu.M Niraparib treated group, and the fifth row was a PDGF-BB and 5. mu.M Niraparib treated group. As can be seen from fig. 2, after VSMC cells were treated with PDGF-BB, EdU-positive cells increased, and after further treatment with Niraparib, EdU-positive cells decreased, and the amount of EdU-positive cells decreased as the concentration of Niraparib increased. In conclusion, after the VSMC cells are treated by the PDGF-BB, the proliferation of the VSMC cells can be promoted, and the Niraparib can inhibit the PDGF-BB-induced cell proliferation, and the inhibition effect is enhanced along with the increase of the concentration of the Niraparib.
EdU cell proliferation assay: rat-derived primary cells (VSMC) were seeded in 96-well plates and treated with different concentrations (0.5. mu.M, 1. mu.M, 5. mu.M) of Niraparib and vehicle DMSO, respectively, for 4 h. After 48h, the four treatments were stimulated with PDGF-BB (30ng/ml) for 48h (control with an equal volume of DSMO), and EdU incorporation was analyzed according to the manufacturer's instructions and photographed with an Olympus cellSens Entry.
Experiment three
After VSMC cells were treated with PDGF-BB (30ng/ml), they were stimulated with different concentrations of Niraparib (0.5. mu.M, 1. mu.M, 5. mu.M) and DMSO, cells were collected, and after protein extraction, cyclin and oncostatin expression was detected by Polymerase Chain Reaction (PCR) assay, as shown in FIG. 3. VSMC cells expressed cyclin (PCNA, CyclinD1) and oncostatin (P27, P21) under different treatments. As shown in FIG. 3, after treatment with PDGF-BB, the expression of cyclin was increased and the expression of oncostatin was decreased in VSMC cells; after the Niraparib is given for stimulation, the cyclin expression is reduced, and the cancer suppressor protein expression is increased; the expression level of the protein related to the apoptosis has no obvious change.
Experiment four
After VSMC cells are treated by PDGF-BB (30ng/ml), Niraparib (0.5 mu M,1 mu M and 5 mu M) and carrier DMSO with different concentrations are respectively given for stimulation, the cells are collected, after protein is extracted, Caspase 3 expression and activity kit (BIOMOL) experiments are utilized to detect the apoptosis level, and the result is shown in figure 4. As can be seen from FIG. 4, the different concentrations of Niraparib had no significant effect on apoptosis.
Experiment five
VSMC cells were treated with PDGF-BB (30ng/ml) and stimulated with different concentrations of Niraparib (0.5. mu.M, 1. mu.M, 5. mu.M) and vehicle DMSO, respectively, to detect cell migration using the transwell assay. The results are shown in FIG. 5. As shown in FIG. 5, PDGF-BB promotes cell migration, while Niraparib inhibits cell migration, and the inhibition increases with increasing concentration.
Cell migration was measured by the Transwell method: VSMC was pretreated with Niraparib for 4h, seeded in the upper chamber, 500L DMEM and fetal bovine serum at 10% concentration by volume and PDGF-BB (30ng/ml) placed in the lower chamber. After 24h, the cells were fixed in the lower air chamber with 4% by mass of formaldehyde for 20 minutes and stained with 0.1% by mass of crystal violet for 20 minutes. Migrated cells were photographed using the Olympus cellSens channel.
Experiment six
VSMC were stimulated with different concentrations of Niraparib (0.5. mu.M, 1. mu.M, 5. mu.M) and vehicle DMSO after PDGF-BB treatment (30ng/ml) and examined for cell proliferation and migration by the scratch assay, as shown in FIG. 6. From FIG. 6, it is seen that PDGF-BB promotes the rate of closure of cell scratch, while Niraparib inhibits the promotion, and that the inhibition of cell scratch closure is increased with the increase of the concentration of Niraparib.
Cell scratch test: VSMC were seeded into 6-well plates and cultured to 80% density. The cell monolayer was scratched with a 1ml pipette tip. After pre-incubation of cells with different concentrations of Niraparib for 4h, the cells were stimulated with PDGF-BB (30ng/ml) for 48h (control group with equal volume of DSMO) and then cultured in DMEM containing 10% by volume fetal bovine serum. Cells were visualized using an Olympus cellSens entry and wound closure rate was measured using the Image J program.
Experiment seven
After the carotid artery of a rat is respectively ligated, the rat is treated by a carrier DMSO and 20mg/kg dNairanib, the expression condition of the contractile gene alpha SMA is detected by an immunofluorescence experiment, a tissue section is incubated overnight at 4 ℃ by using an SM alpha-actin primary antibody (the volume ratio is 1: 100), and then incubated for 2 hours at 37 ℃ by using a FITC-conjugated fluorescent secondary antibody. Nucleic acids were stained with DAPI at 37 ℃ for 15 min. The sections were finally visualized using an Olympus cellSens entry. Carotid morphology was observed and the results are shown in FIG. 7. In fig. 7, the first column is the sham-operated and empty-load treatment group, the second column is the operated and empty-load treatment group, the third column is the operated and Niraparib treatment group, and the first row is the immunofluorescence staining of carotid artery α SMA, the second row is the staining of carotid artery nuclei by DAPI, and the third row is the fusion map of the first two rows. As shown in FIG. 7, after the rat was ligated to the carotid artery, the carotid artery wall was thickened, and the expression level of the contractile gene α SMA in the carotid artery was decreased; after further treatment with Niraparib, the carotid wall thickness decreased and the expression level of the contractile gene alpha SMA increased. After the blood vessel is damaged, the blood vessel wall is thickened, and the expression quantity of the contraction gene alpha SMA is reduced.
Experiment eight
After the VSMC cells were treated with PDGF-BB, they were stimulated with different concentrations of Niraparib (0.5. mu.M, 1. mu.M, 5. mu.M) and vehicle DMSO, respectively, and the level of PDGFR β protein was detected by Western blotting, as shown in FIG. 8. From fig. 8, it can be seen that PDGFR β protein levels decreased with increasing concentrations of Niraparib after VSMC cells were treated with PDGF-BB and further treated with Niraparib. Therefore, Niraparib inhibited protein expression of PDGFR β, and this inhibition was increased with increasing Niraparib concentration.
From the experimental results, it is clear that nilapanib can directly inhibit the phenotypic transformation of VSMC.
It will be apparent to those skilled in the art that various modifications may be made to the above embodiments without departing from the general spirit and concept of the invention. All falling within the scope of the present invention. The protection scheme of the invention is subject to the appended claims.

Claims (10)

1. Application of nilapanib or a derivative thereof in preparation of a preparation for inhibiting PDGFR beta expression and/or PDGFR beta downstream signaling pathway.
2. Application of nilapanib or a derivative thereof in preparing a preparation for inhibiting smooth muscle phenotype switching.
3. Use according to claim 2, wherein the smooth muscle phenotype is switched to a contractile to a synthetic phenotype.
4. Application of nilapanib or a derivative thereof in preparing a medicament for preventing and treating vascular diseases.
5. The use of claim 4, wherein the vascular disease comprises tunica media calcification, atherosclerosis, hypertension, endothelial cell hyperproliferation due to vascular injury, and vascular restenosis.
6. The use of claim 5, wherein the restenosis comprises restenosis following vascular injury.
7. The use of claim 6, wherein the post-vascular injury restenosis comprises PCI restenosis, in-stent restenosis, post-bypass graft restenosis.
8. The use of claim 4, wherein the concentration of nilapanib present in the prophylactic or therapeutic agent is 0.5-5 μ M or 5-20 mg/kg/d; preferably, the concentration used is 0.5. mu.M or 20 mg/kg/d.
9. An agent for inhibiting PDGFR β expression and/or PDGFR β downstream signaling pathway, which comprises nilapanib or a derivative thereof as an active ingredient.
10. Application of nilapanib or a derivative thereof in preparing a preparation for promoting alpha SMA gene expression.
CN202111381376.8A 2021-11-21 2021-11-21 New application of nilapanib and derivatives thereof Pending CN114010635A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111381376.8A CN114010635A (en) 2021-11-21 2021-11-21 New application of nilapanib and derivatives thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111381376.8A CN114010635A (en) 2021-11-21 2021-11-21 New application of nilapanib and derivatives thereof

Publications (1)

Publication Number Publication Date
CN114010635A true CN114010635A (en) 2022-02-08

Family

ID=80065667

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111381376.8A Pending CN114010635A (en) 2021-11-21 2021-11-21 New application of nilapanib and derivatives thereof

Country Status (1)

Country Link
CN (1) CN114010635A (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109475533A (en) * 2016-06-15 2019-03-15 剑桥企业有限公司 The treatment of angiosteosis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109475533A (en) * 2016-06-15 2019-03-15 剑桥企业有限公司 The treatment of angiosteosis

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CHAO ZHANG等: "Phenotypic switching of atherosclerotic smooth muscle cells is regulated by activated PARP1-dependent TET1 expression", 《J ATHEROSCLER THROMB》 *
ROBERT J. HENNING等: "Poly(ADP ribose) Polymerase (PARP) and PARP Inhibitors: Mechanisms of Action and Role in Cardiovascular Disorders", 《CARDIOVASCULAR TOXICOLOGY》 *
XIAO-FEI GAO等: "Plasma Small Extracellular Vesicle-Carried miRNA-501-5p Promotes Vascular Smooth Muscle Cell Phenotypic ModulationMediated In-Stent Restenosis", 《OXIDATIVE MEDICINE AND CELLULAR LONGEVITY》 *
XIAOTING LI等: "PARP inhibitors promote stromal fibroblast activation by enhancing CCL5 autocrine signaling in ovarian cancer", 《PRECISION ONCOLOGY》 *
姜宗来等: "《血管力学生物学》", 31 December 2017, 上海交通大学出版社 *

Similar Documents

Publication Publication Date Title
Shih et al. Physiology and pathophysiology of renal erythropoietin-producing cells
Hu et al. Cellular senescence in cardiovascular diseases: a systematic review
Wu et al. Genetic and pharmacological inhibition of Rheb1-mTORC1 signaling exerts cardioprotection against adverse cardiac remodeling in mice
Li et al. Quercetin attenuates trauma-induced heterotopic ossification by tuning immune cell infiltration and related inflammatory insult
Shin et al. Iron released from reactive microglia by noggin improves myelin repair in the ischemic brain
Pallet et al. Rapamycin inhibits human renal epithelial cell proliferation: effect on cyclin D3 mRNA expression and stability
Li et al. Oxygen homeostasis and cardiovascular disease: A role for HIF?
Li et al. Induction of inducible nitric oxide synthase by isoflurane post-conditioning via hypoxia inducible factor-1α during tolerance against ischemic neuronal injury
Cai et al. MicroRNA-24 attenuates vascular remodeling in diabetic rats through PI3K/Akt signaling pathway
Jin et al. L-type calcium channel modulates cystic kidney phenotype
Martínez-Salgado et al. Involvement of H-and N-Ras isoforms in transforming growth factor-β1-induced proliferation and in collagen and fibronectin synthesis
Gao et al. Ginsenoside Re inhibits vascular neointimal hyperplasia in balloon-injured carotid arteries through activating the eNOS/NO/cGMP pathway in rats
Mohamed et al. Role of nitric oxide and KATP channel in the protective effect mediated by nicorandil in bile duct ligation-induced liver fibrosis in rats
Belayev et al. Protection against blood–brain barrier disruption in focal cerebral ischemia by the type IV phosphodiesterase inhibitor BBB022: a quantitative study
Wang et al. Fastigial nucleus electrostimulation promotes axonal regeneration after experimental stroke via cAMP/PKA pathway
US20090118202A1 (en) Compositions and methods of treating hypertension with tannin complexes
Li et al. Osthole inhibits intimal hyperplasia by regulating the NF-κB and TGF-β1/Smad2 signalling pathways in the rat carotid artery after balloon injury
Du et al. Artemisinin attenuates the development of atherosclerotic lesions by the regulation of vascular smooth muscle cell phenotype switching
Astrike-Davis et al. Targeting cellular senescence as a novel treatment for osteoarthritis
WO2008001369A1 (en) Use of cb2 receptor agonists for promoting neurogenesis
Maita et al. Evaluation of the aging effect on peripheral nerve regeneration: a systematic review
Janardhanan et al. The role of repeat administration of adventitial delivery of lentivirus-shRNA-Vegf-A in arteriovenous fistula to prevent venous stenosis formation
Yoshimoto et al. Cilostazol may prevent cerebral vasospasm following subarachnoid hemorrhage
Zhan et al. Butylphthalide inhibits TLR4/NF-κB pathway by upregulation of miR-21 to have the neuroprotective effect
Li et al. Gomisin N attenuated cerebral ischemia-reperfusion injury through inhibition of autophagy by activating the PI3K/AKT/mTOR pathway

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination