CN113967260A - Application of HEXB inhibitor in preparation of medicine for treating tumor - Google Patents

Application of HEXB inhibitor in preparation of medicine for treating tumor Download PDF

Info

Publication number
CN113967260A
CN113967260A CN202111463377.7A CN202111463377A CN113967260A CN 113967260 A CN113967260 A CN 113967260A CN 202111463377 A CN202111463377 A CN 202111463377A CN 113967260 A CN113967260 A CN 113967260A
Authority
CN
China
Prior art keywords
hexb
tumor
biomarker
kit
inhibitor
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
CN202111463377.7A
Other languages
Chinese (zh)
Inventor
吴安华
程文
朱晨
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to CN202111463377.7A priority Critical patent/CN113967260A/en
Publication of CN113967260A publication Critical patent/CN113967260A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7008Compounds having an amino group directly attached to a carbon atom of the saccharide radical, e.g. D-galactosamine, ranimustine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Pathology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The disclosure relates to application of a HEXB inhibitor in preparing a medicine for treating tumors, application of a reagent capable of quantitatively detecting HEXB in preparing a kit for detecting tumors and a kit for detecting tumors. Through the technical scheme, the embodiment of the disclosure takes the HEXB as a therapeutic target and a diagnostic marker of the tumor with the aerobic glycolytic phenotype, and provides a new therapeutic means and a new diagnostic means for the tumor with the aerobic glycolytic phenotype.

Description

Application of HEXB inhibitor in preparation of medicine for treating tumor
Technical Field
The disclosure relates to the technical field of tumor detection and treatment, in particular to application of an HEXB inhibitor in preparing a medicine for treating tumors, application of a reagent capable of quantitatively detecting HEXB in preparing a kit for tumor detection and a kit for tumor detection.
Background
Glioblastoma multiforme (GBM) is a primary tumor with a high incidence and high malignancy in the brain. Because of its aggressive growth, GBM often recurs after microsurgical resection, and therefore, postoperative adjuvant therapy is a standard therapeutic approach to GBM, primarily targeting proliferating tumor cells, abnormally produced blood vessels, or activated oncogenic pathways to slow tumor progression. In the past 30 years, although some progress has been made in the molecular typing and comprehensive treatment strategies of GBM, the life expectancy of GBM patients has not been significantly extended, only about 15 months. In the face of this serious challenge, there is a need to develop effective treatment strategies from a more novel and comprehensive perspective.
Disclosure of Invention
The purpose of the present disclosure is to provide a drug that can be used for the treatment of glioblastoma multiforme. The application of the HEXB inhibitor in preparing the medicine for treating the tumor, the application of the reagent capable of quantitatively detecting the HEXB in preparing the kit for detecting the tumor and the kit for detecting the tumor.
To achieve the above objects, the present disclosure provides the use of a HEXB inhibitor for the preparation of a medicament for the treatment of a tumor having an aerobic glycolytic phenotype;
preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
Optionally, the HEXB inhibitor comprises at least one of an agent capable of inhibiting HEXB expression, an agent capable of inhibiting HEXB activity, an agent capable of eliminating HEXB, and an agent capable of promoting in vivo degradation of HEXB; preferably, the HEXB inhibitor comprises Gal-PUGNAC, or at least one of a functional fragment, derivative, and salt or solvate thereof of Gal-PUGNAC; more preferably, the HEXB inhibitor is Gal-PUGNAC encapsulated by a nano-drug carrier.
The present disclosure also provides the use of a biomarker detection reagent in the preparation of a kit for the detection of a tumor, wherein the biomarker is HEXB, the biomarker detection reagent is a reagent capable of quantitatively detecting the biomarker, the tumor has an aerobic glycolytic phenotype;
preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
Optionally, the kit for detecting tumor is a kit for detecting the content of the biomarker by a colloidal gold strip method, an immunoblotting method, an immunohistochemistry method, an immunofluorescence method or an ELISA method.
Optionally, said reagent capable of quantitatively detecting said biomarker comprises an anti-HEXB monoclonal antibody; more preferably, the anti-HEXB monoclonal antibody is a commercial sc-376781 monoclonal antibody available from Santa Cruz Biotechnology.
The present disclosure also provides a kit for tumor detection, wherein the kit comprises a reagent for detecting a biomarker, the biomarker is HEXB, the reagent for detecting a biomarker is a reagent capable of quantitatively detecting the biomarker, and the tumor has an aerobic glycolytic phenotype;
preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
Optionally, the kit is a kit for detecting the content of the biomarker by a colloidal gold test paper method, an immunoblotting method, an immunohistochemistry method, an immunofluorescence method or an ELISA method.
Optionally, said reagent capable of quantitatively detecting said biomarker comprises an anti-HEXB monoclonal antibody; more preferably, the anti-HEXB monoclonal antibody is a commercial sc-376781 monoclonal antibody available from Santa Cruz Biotechnology.
The present disclosure also provides for the use of a HEXB inhibitor for the preparation of a medicament for preventing or inhibiting the conversion of macrophages to M2-type macrophages.
Optionally, the HEXB inhibitor comprises at least one of an agent capable of inhibiting HEXB expression, an agent capable of inhibiting HEXB activity, an agent capable of eliminating HEXB, and an agent capable of promoting in vivo degradation of HEXB; preferably, the HEXB inhibitor includes Gal-PUGNAC or at least one of a functional fragment, derivative, and salt or solvate thereof of Gal-PUGNAC.
Through the technical scheme, the embodiment of the disclosure takes the HEXB as a therapeutic target and a diagnostic marker of the tumor with the aerobic glycolytic phenotype, and provides a new therapeutic means and a new diagnostic means for the tumor with the aerobic glycolytic phenotype.
Additional features and advantages of the disclosure will be set forth in the detailed description which follows.
Drawings
The accompanying drawings, which are included to provide a further understanding of the disclosure and are incorporated in and constitute a part of this specification, illustrate embodiments of the disclosure and together with the description serve to explain the disclosure without limiting the disclosure. In the drawings:
FIG. 1 is a schematic representation of the correlation of TAM marker IBA1 with the glycolytic marker PKM2 in GBM patients;
FIG. 2 is a graph showing the expression levels of HEXB in normal brain, low-grade glioma and GBM;
FIG. 3 is a graph showing the expression amount of HEXB in the mesenchymal and IDH1 wild-type groups of GBM;
FIG. 4 is a graphical representation of the results of glucose and lactate levels measurements in HEXB silenced U87 cells and PGC1 cells;
FIG. 5 is a graphical representation of the effect of RNA interference and Gal-PUGNAC inhibition of HEXB on the glycolytic rate of U87 cells;
FIG. 6 is a graphical representation of the effect of RNA interference and Gal-PUGNAC inhibition of HEXB on the glycolytic rate of PGC1 cells;
FIG. 7 is a schematic representation of the effect of siRNA silencing HEXB on GBM cell proliferation rate;
FIG. 8 is a graphical representation of the effect of HEXB inhibition on the growth status of GBM subcutaneous xenograft tumors;
FIG. 9 is a graphical representation of the effect of HEXB silencing on the survival of GBM orthotopic xenograft tumor growth status;
FIG. 10 is a schematic comparison of methylation levels of the HEXB promoter in IDH1 wild-type GBM and IDH1 mutant GBM;
FIG. 11 is a graph showing comparison of the expression amounts of HEXB in IDH1 wild-type tissue and IDH1RG32H mutant tissue;
FIG. 12 is the effect of IDH1 mutation on HEXB expression in GBM cells and the associated reversion;
fig. 13 is that targeting HEXB was more significant in IDH1 wild-type GBM model (SB sleeping beauty spontaneous tumor model).
Detailed Description
The following detailed description of specific embodiments of the present disclosure is provided in connection with the accompanying drawings. It should be understood that the detailed description and specific examples, while indicating the present disclosure, are given by way of illustration and explanation only, not limitation.
A first aspect of the disclosure provides the use of a HEXB inhibitor for the preparation of a medicament for the treatment of a tumor having an aerobic glycolytic phenotype; preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
Aerobic glycolysis, also known as the Warburg effect, is a typical metabolic pattern in malignant tumor cells where tumor cells convert most of the glucose to lactate through glycolysis reactions, even in the presence of sufficient oxygen to maintain the oxidative phosphorylation metabolic pattern. Aerobic glycolysis is not only a byproduct of malignant transformation of tumors, but also a key driving force of tumorigenesis, so that a molecular mechanism for activating and maintaining aerobic glycolysis is disclosed, and a new means can be provided for detection and treatment of tumors.
GBM has a complex tumor microenvironment in which macrophages of type M2 (TAMs) account for up to 30% of all cells, and enrichment of TAMs promotes malignant development of tumor cells. The inventors of the present disclosure found that HEXB can be a target for aerobic glycolysis and TAM infiltration, which can enhance aerobic glycolysis of tumor cells through YAP1/HIF1A pathway and form an external positive feedback regulation loop with TAM in microenvironment.
Specifically, to assess the interrelationship of aerobic glycolysis and TAM in GBM, the inventors performed ssGSEA quantitative scoring based on the classical gene set of glycolytic metabolism. The relative proportion of TAM in GBM tissues is analyzed through CIBERSORT, and the glycolytic score and the macrophage proportion are all obviously and positively correlated in a plurality of GBM queues; by the grouping of GBM patients, it was confirmed that there was also a strong positive correlation between TAM marker IBA1 and glycolytic marker PKM2, as shown in fig. 1; HEXB is screened out by carrying out cross analysis on glucose metabolism related genes and characteristic genes (containing macrophages and brain microglia) of TAM; the expression of HEXB was analyzed based on the single cell RNA sequencing cohort (GSE131928), and HEXB was found to be expressed predominantly in GBM cells and macrophages; immunofluorescence confirms that the HEXB is co-expressed and adjacently positioned with IBA1 and GFAP, which indicates that the HEXB is simultaneously expressed in TAM and GBM cells; immunohistochemistry and enzyme-linked immunosorbent assay found that HEXB was significantly overexpressed and secreted in GBM compared to normal brain and low-grade glioma, as shown in figure 2; other bioinformatic analyses found that HEXB was significantly highly expressed in subtypes whose metabolic patterns such as the mesenchymal and IDH1 wild-type groups of GBM and immune microenvironment constituted complex, as shown in fig. 3, indirectly confirmed the strong association of HEXB with aerobic glycolysis and immune microenvironment remodeling that may exist.
To further illustrate the biological role of HEXB in GBM cells, the inventors evaluated the expression of HEXB in two HEXB high expression level cell lines (U87 cell line and PGC1 cell line). In the HEXB silencing group, the glucose consumption and lactate production of tumor cells were severely reduced, as shown in fig. 4; using a hippocampal analyzer to explore the role of HEXB in modulating glycolytic trends, RNA interference and inhibition of HEXB by Gal-punac (a HEXB inhibitor, inhibiting its enzymatic activity) were found to significantly reduce the glycolytic rate of GBM cells, as shown in figures 5 and 6.
Furthermore, silencing HEXB by siRNA can significantly reduce the proliferation rate of GBM cells, as shown in fig. 7; in addition, knockout of HEXB does not affect the apoptotic state, pyruvate was used to mimic glycolysis and was found to reverse the growth inhibitory effect of silencing HEXB, indicating that HEXB's promotion of tumor cell proliferation depends on its regulation of carbohydrate metabolism rather than by other means (e.g., apoptosis); five GBM neurospheres with high levels of HEXB expression were established under serum-free culture conditions, and ball formation and self-renewal capacity of GBM neurospheres was inhibited following endogenous knockout of HEXB or administration of Gal-punac; establishing a GBM subcutaneous xenograft model, and finding that inhibition of HEXB can obviously inhibit the growth rate of subcutaneous tumors, as shown in FIG. 8; meanwhile, xenotransplantation in situ of HEXB silenced GBM cells delayed tumor formation and prolonged survival, as shown in fig. 9. Thus, these findings suggest that HEXB promotes GBM cell proliferation by enhancing the glycolytic process.
In addition, the inventors of the present disclosure also found that in glioblastoma multiforme, mutation of IDH1 gene could eliminate the glycolytic effect of HEXB to some extent by epigenetic means, resulting in low expression of HEXB. Specifically, the expression of HEXB in IDH1 wild-type GBM was significantly higher than IDH1 mutant GBM, as shown in fig. 3; and the methylation level of the HEXB promoter in IDH1 wild-type GBM was significantly lower than that of IDH1 mutant GBM, as shown in FIG. 10; IHC in GBM tissue showed that IDH1 wild-type tissue had more HEXB than IDH1RG 132H mutant as shown in fig. 11; construction of U87 and PGC1 cell lines with different IDH1 mutations, found that the expression level of HEXB was significantly reduced in IDH1RG32H mutant cells, as shown in FIG. 12; administration of IDH1 mutant metabolite 2HG inhibitor in mutant cells was found to restore HEXB expression, as shown in figure 12. At the same time, the therapeutic effect of targeted HEXB therapy was more pronounced in IDH1 wild-type GBM, as shown in fig. 13.
In accordance with the present disclosure, the HEXB inhibitor may be selected within a range, for example, the HEXB inhibitor may include at least one of an agent capable of inhibiting HEXB expression, an agent capable of inhibiting HEXB activity, an agent capable of eliminating HEXB, and an agent capable of promoting in vivo degradation of HEXB; preferably, the HEXB inhibitor can include Gal-PUGNAC, or at least one of a functional fragment, derivative, and salt or solvate thereof; more preferably, the HEXB inhibitor is Gal-PUGNAC encapsulated by a nano-drug carrier. Wherein, the nano-drug carrier is a solid paper nano-particle. The weight ratio of the nano-drug carrier to the Gal-PUGNAC is 0.5: 1. Gal-PUGNAC wrapped by the nano-drug carrier can have higher stability and blood brain barrier transmittance, thereby being more beneficial to realizing sustained release and site-specific release of a tumor microenvironment.
A second aspect of the present disclosure provides the use of a biomarker detection reagent in the preparation of a kit for the detection of a tumor, wherein the biomarker is HEXB, the biomarker detection reagent is a reagent capable of quantitatively detecting the biomarker, the tumor has an aerobic glycolytic phenotype; preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
In the embodiments of the present disclosure, the principle of HEXB as a biomarker for tumor detection with aerobic glycolytic phenotype is similar or identical to the aforementioned principle of HEXB as a target for tumor therapy with aerobic glycolytic phenotype, and is not repeated here.
According to the disclosure, the method for detecting HEXB by using the kit can be selected within a certain range, for example, the kit for detecting tumor is a kit for detecting the content of the biomarker by a colloidal gold test paper method, an immunoblotting method, an immunohistochemistry method, an immunofluorescence method or an ELISA method.
According to the present disclosure, the reagent capable of quantitatively detecting the biomarker may be selected within a certain range, for example, the reagent capable of quantitatively detecting the biomarker may include a monoclonal antibody against HEXB.
A third aspect of the present disclosure provides a kit for tumor detection, wherein the kit comprises a reagent for detecting a biomarker, the biomarker is HEXB, the reagent for detecting a biomarker is a reagent capable of quantitatively detecting the biomarker, and the tumor has an aerobic glycolysis phenotype; preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
Optionally, the kit is a kit for detecting the content of the biomarker by a colloidal gold test paper method, an immunoblotting method, an immunohistochemistry method, an immunofluorescence method or an ELISA method.
Optionally, the reagent capable of quantitatively detecting the biomarker comprises an anti-HEXB monoclonal antibody.
A fourth aspect of the disclosure provides the use of a HEXB inhibitor for the manufacture of a medicament for preventing or inhibiting the conversion of macrophages to M2-type macrophages.
The inventors of the present disclosure found that HEXB is able to form an external positive feedback regulatory loop with macrophages of type M2 in the tumor microenvironment. Specifically, the content of immune cell components in the GBM database was shown by CIBERSORT, and the high-expression HEXB group was found to be accompanied by more M0 macrophages and M2 macrophages in multiple cohorts. In addition, exogenous HEXB can significantly promote chemotaxis of THP 1-derived macrophages. The chemotactic capacity of macrophages can be significantly reduced by adding Gal-PUGNAC under the stimulation of exogenous HEXB. The positive correlation and co-location between HEXB and M2-type macrophages was verified by immunofluorescence and IHC in GBM samples. To explore the role of HEXB in macrophage polarization, flow cytometry was performed in patient-derived PBMC. The results show that exogenous HEXB significantly reversed the induced M1 polarization process, whereas treatment with Gal-PUGNAC strongly suppressed M2 polarization, and in the GL261 in situ model, Gal-PUGNAC also promoted M2 transitions. The above results indicate that HEXB can be a key target for macrophage chemotaxis and M2 alternate polarization.
By analysis of the GSE5099 cohort, it was found that endogenous expression of HEXB increased during macrophage differentiation to M2 type. Experiments were performed in THP1 and PBMC and found that expression and secretion of HEXB in macrophages of the M2 subtype were significantly upregulated. Thus, in the glioma microenvironment, a positive feedback loop exists between HEXB and M2 macrophages. The U87 intracranial tumor model was used to verify the external positive feedback loop, and it was found that the tumor growth in the U87 and macrophage co-injected group was accelerated, and that the growth in the U87 and HEXB pretreated macrophage co-injected group was fastest, and Gal-PUGNAC could significantly inhibit the tumor, compared to the control group.
Optionally, the HEXB inhibitor comprises at least one of an agent capable of inhibiting HEXB expression, an agent capable of inhibiting HEXB activity, an agent capable of eliminating HEXB, and an agent capable of promoting in vivo degradation of HEXB; preferably, the HEXB inhibitor includes Gal-PUGNAC or at least one of a functional fragment, derivative, and salt or solvate thereof of Gal-PUGNAC.
The present disclosure is further illustrated by the following examples, but is not to be construed as being limited thereby.
The starting materials, reagents, instruments and equipment referred to in the examples of the present disclosure may be obtained by purchase, unless otherwise specified.
Example 1
This example serves to illustrate that HEXB is highly expressed in GBM cells.
Tumor tissues and clinical pathology treatments were collected with the patient and their therapists informed and consented to procedures in compliance with medical ethics committee standards, in which two independent neuropathologists diagnosed the tumor type corresponding to the tumor tissue and confirmed that these tumor tissues met the quality requirements for molecular detection.
After the tissue samples were collected, immunohistochemistry and ELISA were performed on human normal astrocytes (NHA), low-grade glioma cells and glioblastoma multiforme cells.
Immunohistochemical assays were performed as follows:
(1) preparing a slice: washing the sample by PBS to remove blood on the surface, burned tissue and attached paracancer brain tissue; fixing for more than 72 hours at 4 ℃ by using 4% paraformaldehyde; performing gradient dehydration on ethanol according to the concentration sequence of 50% ethanol, 60% ethanol, 70% ethanol, 80% ethanol, 90% ethanol, 95% ethanol I, 95% ethanol II, anhydrous ethanol I and anhydrous ethanol II, wherein the dehydration time of each concentration is 30 min; carrying out transdermal treatment for 10min by using a mixed solution of xylene and absolute ethyl alcohol in a ratio of 1:1, and then carrying out transdermal treatment for 10min by using xylene I and xylene II in sequence; wax dipping is carried out twice, each time for 30 min; finally, embedding a mold, and slicing after fully cooling;
(2) histochemical staining: baking the slices at 60 ℃ for 1 hour, and treating the slices for 10 minutes by using dimethylbenzene I and dimethylbenzene II respectively; then sequentially dewaxing by using absolute ethyl alcohol I, absolute ethyl alcohol II, 95% ethyl alcohol, 90% ethyl alcohol, 80% ethyl alcohol and 70% ethyl alcohol for 5 minutes respectively; distilled water treatment for 5 minutes, PBS washing 3 times, each time hydration for 5 minutes; adding peroxidase blocking solution to remove endogenous peroxidase, incubating at room temperature for 10min, and soaking in PBS for 5min for 3 times after incubation; selecting sodium citrate or pH8.0 EDTA as antigen according to the specification of primary antibody, sealing in sealing solution at 37 deg.C for 30min, diluting primary antibody according to the recommended concentration of the specification, and culturing in a humidified box at 4 deg.C overnight; performing secondary antibody subsequent incubation, DAB color development (30s-2mim), hematoxylin counterstaining (2min), hydrochloric acid acidification (10s), dehydration (in the same embedding and dehydration step) and mounting on the plate on the next day; the staining results were taken under a microscope, and at least 5 standard fields were randomly selected for each tissue section and quantified according to the internationally recognized GIS histochemical scoring formula for subsequent statistical analysis.
Enzyme-linked immunosorbent assay was performed according to the instructions of the HEXB-ELISA kit (CloudClone).
The measurement results are shown in fig. 2, and it can be seen from fig. 2 that the expression level of HEXB in glioblastoma multiforme cells is significantly higher than that of human normal astrocytes (NHA) and low-grade glioma cells.
Example 2
This example serves to illustrate the process of HEXB mediated aerobic glycolysis of GBM tumor cells.
The U87 cell line and PGC1 cell line used in this example were high HEXB expressing glioma cell lines determined after HEXB expression profiling. Wherein, the culture conditions of the U87 cell line are as follows: dulbecco's modified Eagle Medium (DMEM; Gibco, Grand Island, NY, USA) at 37 ℃ and 5% CO2Supplemented with 10% FBS (Gibco) and 1% penicillin/streptomycin (Gibco) under conditions; the culture conditions of the PGC1 cell line were: cultured in RPMI-1640 medium (Gibco) containing 10% FBS and 1% penicillin/streptomycin (Gibco), at 37 ℃ and 5% CO2
(1) The U87 cell line and PGC1 cell line were transfected with lentiviral-coated siRNA (sequence information shown in table 1) in 6-well plates via nvitrogen Lipofectamine 3000 (sequomieheishi science, shanghai, china) to silence the HEXB gene in the U87 and PGC1 cell lines. After successful transfection, the cells were cultured, and then glucose and lactate levels were measured using a high-sensitivity glucose assay kit (Sigma, shanghai, china) and a lactate assay kit II (Sigma, shanghai, china), with the results shown in fig. 4.
TABLE 1
Figure BDA0003390153300000101
As can be seen from fig. 4, in the HEXB silencing group, the glucose consumption and lactate production of tumor cells were severely reduced.
(2) RNA interference and Gal-PUGNAC inhibition were performed on the U87 cell line, and then the reduction effect on the glycolytic rate of the U87 cell line after HEXB inhibition was evaluated by measuring the rate of extracellular acidification with a hippocampal xfe96 extracellular flux analyzer (Seahorse Bioscience), the measurement results being shown in FIG. 5;
RNA interference and Gal-PUGNAC inhibition were performed on PGC1 cell line, and then the reduction effect on the glycolysis rate of PGC1 cell line after HEXB inhibition was evaluated by measuring the rate of extracellular acidification with hippocampal xfe96 extracellular flux analyzer (Seahorse Bioscience), the results of which are shown in FIG. 6.
As can be seen from fig. 5 and 6, RNA interference and inhibition of HEXB by Gal-punac (a HEXB inhibitor, inhibiting its enzymatic activity) significantly attenuated the glycolytic rate of GBM cells.
(3) The U87 cell line was treated with recombinant human HEXB (rhhexb), and then extracellular acidification rate was measured with hippocampal xfe96 extracellular flux analyzer (Seahorse Bioscience) to evaluate the enhancement effect on glycolysis rate of the U87 cell line after HEXB activation, the measurement results are shown in fig. 7;
PGC1 cell line was treated with recombinant human HEXB (rhhexb), and then extracellular acidification rate was measured with hippocampal xfe96 extracellular flux analyzer (Seahorse Bioscience) to evaluate the effect of increasing glycolysis rate of PGC1 cell line after HEXB activation, the results of which are shown in fig. 8.
As can be seen in fig. 7 and 8, treatment with recombinant human hexb (rhhexb) enhances the glycolytic process of GBM cells.
In summary, it can be seen from operations (1) to (3) of this example that HEXB mediates the aerobic glycolysis process of GBM tumor cells.
Example 3
This example serves to illustrate that HEXB promotes GBM cell proliferation by enhancing the glycolytic process.
And respectively establishing a GBM subcutaneous xenograft model and an orthotopic xenograft model. Inhibition of HEXB in a subcutaneous xenograft model, the growth rate of subcutaneous tumors was observed, as shown in figure 10; HEXB was filmed in an orthotopic xenograft model and observed for tumor formation in situ as shown in FIG. 11.
The material sources for establishing the xenograft model are as follows: female BALB/C nude mice and male C57BL/6N mice (6-8 weeks old) were purchased from Charles river, Beijing, China.
For the subcutaneous xenograft model, U87 cells were plated at 2X 10 per 200. mu.l6The density of cells was injected into the right posterior side of nude mice. Tumor growth was measured every 3 days in mice bearing tumors implanted subcutaneously. Subcutaneous tumor volume was calculated as follows: tumor volume (mm)3) Long (longest diameter) x (shortest diameter)2/2。
For intracranial tumor xenografts, cell concentrations of 5X 10 were used5Cells, 3 μ L in volume, were injected intracerebrally, U87 cells were transplanted 3.0mm deep into the right cerebral cortex, and the whole brain was taken at the time of death of tumor-bearing mice or 3 weeks after transplantation for further examination. The section with the largest tumor cross-sectional area in the intracranial glioma model was selected for tumor size measurement.
As can be seen from fig. 8, inhibition of HEXB significantly inhibited the growth rate of subcutaneous tumors; as can be seen in fig. 9, xenotransplantation of HEXB-silenced GBM cells in situ delayed tumor formation and prolonged survival.
The preferred embodiments of the present disclosure are described in detail with reference to the accompanying drawings, however, the present disclosure is not limited to the specific details of the above embodiments, and various simple modifications may be made to the technical solution of the present disclosure within the technical idea of the present disclosure, and these simple modifications all belong to the protection scope of the present disclosure.
It should be noted that, in the foregoing embodiments, various features described in the above embodiments may be combined in any suitable manner, and in order to avoid unnecessary repetition, various combinations that are possible in the present disclosure are not described again.
In addition, any combination of various embodiments of the present disclosure may be made, and the same should be considered as the disclosure of the present disclosure, as long as it does not depart from the spirit of the present disclosure.

Claims (10)

  1. Use of a HEXB inhibitor for the preparation of a medicament for the treatment of a tumor having an aerobic glycolytic phenotype;
    preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
  2. 2. The use of claim 1, wherein the HEXB inhibitor comprises at least one of an agent capable of inhibiting HEXB expression, an agent capable of inhibiting HEXB activity, an agent capable of eliminating HEXB, and an agent capable of promoting in vivo degradation of HEXB; preferably, the HEXB inhibitor comprises Gal-PUGNAC, or at least one of a functional fragment, derivative, and salt or solvate thereof of Gal-PUGNAC; more preferably, the HEXB inhibitor is Gal-PUGNAC encapsulated by a nano-drug carrier.
  3. 3. Use of a biomarker detection reagent in the preparation of a kit for the detection of a tumor, wherein the biomarker is HEXB, the biomarker detection reagent is a reagent capable of quantitatively detecting the biomarker, the tumor has an aerobic glycolytic phenotype;
    preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
  4. 4. The use according to claim 3, wherein the kit for tumor detection is a kit for detecting the content of the biomarker by a colloidal gold strip method, an immunoblotting method, an immunohistochemistry method, an immunofluorescence method or an ELISA method.
  5. 5. Use according to claim 3 or 4, wherein said reagent capable of quantitatively detecting said biomarker comprises a monoclonal antibody against HEXB; more preferably, the anti-HEXB monoclonal antibody is a commercial sc-376781 monoclonal antibody available from Santa Cruz Biotechnology.
  6. 6. A kit for detecting a tumor, wherein the kit comprises a reagent for detecting a biomarker, the biomarker is HEXB, the reagent for detecting a biomarker is a reagent capable of quantitatively detecting the biomarker, and the tumor has an aerobic glycolytic phenotype;
    preferably, the tumor is glioblastoma multiforme, more preferably, the tumor is IDH1 wild-type glioblastoma multiforme.
  7. 7. The kit according to claim 6, wherein the kit is a kit for detecting the content of the biomarker by a colloidal gold test paper method, an immunoblotting method, an immunohistochemistry method, an immunofluorescence method or an ELISA method.
  8. 8. The kit of claim 6 or 7, wherein said reagent capable of quantitatively detecting said biomarker comprises an anti-HEXB monoclonal antibody; more preferably, the anti-HEXB monoclonal antibody is a commercial sc-376781 monoclonal antibody available from Santa Cruz Biotechnology.
  9. Use of a HEXB inhibitor for the manufacture of a medicament for preventing or inhibiting the conversion of macrophages to M2-type macrophages.
  10. 10. The use of claim 9, wherein the HEXB inhibitor comprises at least one of an agent capable of inhibiting HEXB expression, an agent capable of inhibiting HEXB activity, an agent capable of eliminating HEXB, and an agent capable of promoting in vivo degradation of HEXB; preferably, the HEXB inhibitor includes Gal-PUGNAC or at least one of a functional fragment, derivative, and salt or solvate thereof of Gal-PUGNAC.
CN202111463377.7A 2021-12-02 2021-12-02 Application of HEXB inhibitor in preparation of medicine for treating tumor Pending CN113967260A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111463377.7A CN113967260A (en) 2021-12-02 2021-12-02 Application of HEXB inhibitor in preparation of medicine for treating tumor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111463377.7A CN113967260A (en) 2021-12-02 2021-12-02 Application of HEXB inhibitor in preparation of medicine for treating tumor

Publications (1)

Publication Number Publication Date
CN113967260A true CN113967260A (en) 2022-01-25

Family

ID=79590572

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111463377.7A Pending CN113967260A (en) 2021-12-02 2021-12-02 Application of HEXB inhibitor in preparation of medicine for treating tumor

Country Status (1)

Country Link
CN (1) CN113967260A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040204381A1 (en) * 1997-11-12 2004-10-14 Moskal Joseph R Detection and treatment of glyco-enzyme-related disease
CN107034305A (en) * 2017-06-19 2017-08-11 上海市第十人民医院 A kind of diagnosis marker of glioblastoma
US20210311076A1 (en) * 2018-07-16 2021-10-07 University Of Virginia Patent Foundation Compositions and methods of diagnosis and treatment for neurological diseases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040204381A1 (en) * 1997-11-12 2004-10-14 Moskal Joseph R Detection and treatment of glyco-enzyme-related disease
CN107034305A (en) * 2017-06-19 2017-08-11 上海市第十人民医院 A kind of diagnosis marker of glioblastoma
US20210311076A1 (en) * 2018-07-16 2021-10-07 University Of Virginia Patent Foundation Compositions and methods of diagnosis and treatment for neurological diseases

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
COLIN R.KENNEDY,ET AL.: "Differential sensitivities of glioblastoma cell lines towards metabolic and signaling pathway inhibitions", 《CANCER LETTERS》 *
JIA MENGXIAN ET AL.: "Microglia-Specific Expression of HEXA and HEXB Leads to Poor Prognosis in Glioblastoma Patients", 《FRONTIERS IN ONCOLOGY》 *
STUBBS KEITH A.ET AL.: "A Selective Inhibitor Gal-PUGNAc of Human Lysosomal beta-Hexosaminidases Modulates Levels of the Ganglioside GM2 in Neuroblastoma Cells", 《 ANGEW. CHEM. INT. ED.》 *

Similar Documents

Publication Publication Date Title
Xu et al. FTO expression is associated with the occurrence of gastric cancer and prognosis
Wang et al. RETRACTED: Knockdown of USF1 Inhibits the Vasculogenic Mimicry of Glioma Cells via Stimulating SNHG16/miR-212-3p and linc00667/miR-429 Axis
Liu et al. ADAR1 promotes the epithelial-to-mesenchymal transition and stem-like cell phenotype of oral cancer by facilitating oncogenic microRNA maturation
Zhao et al. An extensive invasive intracranial human glioblastoma xenograft model: role of high level matrix metalloproteinase 9
Kang et al. EGFL7 is an intercellular EGFR signal messenger that plays an oncogenic role in glioma
Yu et al. MicroRNA-146b-3p promotes cell metastasis by directly targeting NF2 in human papillary thyroid cancer
Huang et al. Fatty acid binding protein 4 enhances prostate cancer progression by upregulating matrix metalloproteinases and stromal cell cytokine production
Mao et al. RETRACTED ARTICLE: Molecular basis underlying inhibition of metastasis of gastric cancer by anti-VEGFa treatment
Yao et al. Role of HGF/c‐Met in the treatment of colorectal cancer with liver metastasis
Feng et al. Low levels of AMPK promote epithelial‐mesenchymal transition in lung cancer primarily through HDAC4‐and HDAC5‐mediated metabolic reprogramming
CN108949984B (en) Application of gene DESI2 in diagnosis, prognosis evaluation and treatment of triple negative breast cancer
Zhu et al. RETRACTED ARTICLE: miR-142-3p targeting NUCKS1 inhibits proliferation and invasion of pancreatic cancer cells
Yu et al. Linc00702 inhibits cell growth and metastasis through regulating PTEN in colorectal cancer.
Jia et al. Decorin suppresses invasion and EMT phenotype of glioma by Inducing autophagy via c-Met/Akt/mTOR axis
Fang et al. Cancer-associated fibroblast-like fibroblasts in vocal fold leukoplakia suppress CD8+ T cell functions by inducing IL-6 autocrine loop and interacting with Th17 cells
KR101539138B1 (en) Combination therapy and method for assessing resistance to treatment
Jiang et al. miR-155-5p promotes progression of acute respiratory distress syndrome by inhibiting differentiation of bone marrow mesenchymal stem cells to alveolar type II epithelial cells
Xue et al. Chlorogenic acid inhibits epithelial-mesenchymal transition and invasion of breast cancer by down-regulating LRP6
Ma et al. CCNE1 promotes progression and is associated with poor prognosis in lung adenocarcinoma
CN113967260A (en) Application of HEXB inhibitor in preparation of medicine for treating tumor
Kamoshida et al. Expression of cancer cachexia-related factors in human cancer xenografts: an immunohistochemical analysis
Wang et al. miR-125b enhances metastasis and progression of cancer via the TXNIP and HIF1α pathway in pancreatic cancer
Li et al. Relationship between clinicopathological features and HIF-2α in gastric adenocarcinoma
CN109385475B (en) Product for evaluating effect of propranolol on treating infantile hemangioma
CN112852960A (en) Papillary thyroid carcinoma biomarker and application thereof

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
RJ01 Rejection of invention patent application after publication

Application publication date: 20220125

RJ01 Rejection of invention patent application after publication