WO2019218810A1 - Pharmaceutical composition for treating non-hodgkin's lymphoma - Google Patents

Pharmaceutical composition for treating non-hodgkin's lymphoma Download PDF

Info

Publication number
WO2019218810A1
WO2019218810A1 PCT/CN2019/082102 CN2019082102W WO2019218810A1 WO 2019218810 A1 WO2019218810 A1 WO 2019218810A1 CN 2019082102 W CN2019082102 W CN 2019082102W WO 2019218810 A1 WO2019218810 A1 WO 2019218810A1
Authority
WO
WIPO (PCT)
Prior art keywords
rituximab
cells
lymphoma
hodgkin
inhibitor
Prior art date
Application number
PCT/CN2019/082102
Other languages
French (fr)
Chinese (zh)
Inventor
胡维国
Original Assignee
复旦大学附属肿瘤医院
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 复旦大学附属肿瘤医院 filed Critical 复旦大学附属肿瘤医院
Publication of WO2019218810A1 publication Critical patent/WO2019218810A1/en

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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a pharmaceutical composition for treating non-Hodgkin's lymphoma, and more particularly to a pharmaceutical composition for treating B cell non-Hodgkin's lymphoma.
  • Burkitt's lymphoma is a highly invasive B-cell non-Hodgkin's lymphoma (NHL), accounting for 3-5% of lymphoma in all age groups, accounting for children. 40-50% of lymphomas are characterized by highly expressed c-MYC targets and germinal center-associated B cell genes, as well as low expressed MHC-I molecules and NF- ⁇ B target genes.
  • NHL non-Hodgkin's lymphoma
  • NHL non-Hodgkin's lymphoma
  • Rituximab combined with CHOP chemotherapy can increase overall survival in patients with diffuse large B-cell lymphoma (DLBCL) by at least 20%.
  • DLBCL diffuse large B-cell lymphoma
  • R-CHOP CHOP chemotherapy
  • many single-arm clinical trials have further demonstrated the efficacy of rituximab for BL-enhanced short-term chemotherapy regimens.
  • rituximab has achieved great success in treating a wide variety of B-cell lymphomas
  • resistance to rituximab is a major challenge for relapsed/refractory patients and remains Approximately 50% of patients do not respond to treatment with rituximab, and patients who initially respond will eventually develop resistance to further treatment with rituximab.
  • Rituximab kills lymphocytes primarily by antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) by binding to membrane CD20.
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Resistance to ADCC may be due to the intrinsic polymorphism of the Fc Fc receptor FcgammaRIIIa gene, and resistance to CDC is likely due to down-regulation of CD20 expression and upregulation of membrane complement regulatory proteins (mCRPs), particularly CD59.
  • CD20 In order to improve the therapeutic efficacy of rituximab, researchers in the field have made considerable efforts to improve the expression of CD20, such as by the histone deacetylase inhibitor trichostatin A or synthetic CpG oligodeoxynucleotides. , or the function of CD59 is inhibited by modified monoclonal antibody or bacterial toxin-derived ILYd4.
  • modified monoclonal antibody or bacterial toxin-derived ILYd4 modified monoclonal antibody or bacterial toxin-derived ILYd4.
  • CpG oligodeoxynucleotides were further evaluated for safety in B-cell NHL patients in clinical trials (stage I) and no further clinical trials have been reported. Therefore, there is a need to find alternative treatment strategies.
  • Pro-apoptotic agents are used in the treatment of B-cell lymphoma.
  • the present invention first provides a pharmaceutical composition for treating non-Hodgkin's lymphoma, including a protein kinase C (PKC) inhibitor and rituximab.
  • PLC protein kinase C
  • composition for treating non-Hodgkin's lymphoma of the present invention further comprises a pharmaceutically acceptable carrier or excipient.
  • the protein kinase C inhibitor of the present invention includes, but is not limited to, staurosporin and its analogs, carbamazepines, biguanide maleamides, biguanide maleamide macrocycles Compounds, 2-alkylindole maleimide compounds, Balanol compounds, N-phenyl-2-pyrimidinamine derivatives, Rottlerin, H-series inhibitors, sulfonamides and sulfonylbenzoyl derivatives , a monoterpene maleimide derivative, an adenosine-5'-terminal carboxylic acid peptide derivative.
  • the protein kinase C inhibitor is selected from the group consisting of militalin, a derivative thereof, a pharmaceutically acceptable salt thereof, a solvate thereof, and a prodrug thereof.
  • Midostaurin is a broad-spectrum PKC inhibitor (PKC412) derived from modified staurosporins to increase selectivity for PKC and further inhibit other kinases such as FLT3, PDGFR , KIT and VEGFR2 and their metabolites.
  • PKC412 broad-spectrum PKC inhibitor
  • other kinases such as FLT3, PDGFR , KIT and VEGFR2 and their metabolites.
  • chronic oral treatment with midazolam is safe and tolerable and confirmed by the FDA in clinical treatment; it has the chemical structure shown in Formula I below:
  • midazolam alone or in combination with rituximab can be used for the treatment of patients with BL, especially for patients with relapsed/refractory BL, and further for other highly active Treatment of patients with non-Hodgkin's lymphoma of PKC, especially for relapsed/refractory B-cell non-Hodgkin's lymphoma with highly activated PKC.
  • rituximab Although the effect of rituximab on the direct induction of cytotoxicity by lymphocytes is sometimes controversial, many studies have shown that rituximab lacks pro-apoptotic ability in various NHL cells. Our series of studies have revealed that rituximab fails to induce apoptosis to any detectable extent in non-Hodgkin's lymphoma, including BL cells, and therefore apoptosis in rituximab The role played by antitumor activity is very limited. Therefore, we attempted to increase the sensitivity of rituximab therapy by combining other drugs with pro-apoptotic effects.
  • Macromolecular reagents include humanized monoclonal mapramumab targeting TRAIL-R1, genetically engineered fusion protein scFvRit: sFasL, Apo2L/TRAIL (dulanermin), and anti-CD20-interleukin -21, and small molecule reagents including selective NEDD8 activating enzyme (NAE) inhibitor pevonedistat (MLN4924), mTOR inhibitor temsirolimus and proteasome inhibitor bortezomib, enhance the antitumor activity of rituximab on B cell NHL cells .
  • NAE NEDD8 activating enzyme
  • MN4924 selective NEDD8 activating enzyme
  • MN4924 selective NEDD8 activating enzyme
  • mTOR inhibitor temsirolimus and proteasome inhibitor bortezomib
  • enhance the antitumor activity of rituximab on B cell NHL cells enhance the antitumor activity of
  • the present invention also provides a pharmaceutical composition for treating non-Hodgkin's lymphoma comprising rituximab and protein kinase C inhibitors of different specification unit preparations and a pharmaceutically acceptable carrier Or excipients for simultaneous, separate or sequential administration.
  • the present invention provides a pharmaceutical composition for treating non-Hodgkin's lymphoma, comprising a first formulation of a protein kinase C inhibitor and a pharmaceutically acceptable carrier, and rituximab and a pharmaceutically acceptable A second formulation of the accepted carrier formed.
  • the dosage form of the above preparation is an injection administration preparation, a gastrointestinal administration preparation, a respiratory administration preparation, a dermal administration preparation, a mucosal administration preparation or a channel administration preparation.
  • the injection administration preparation of the present invention includes, but is not limited to, intravenous injection, intramuscular injection, subcutaneous injection, intradermal injection, intraluminal injection, and the like; and the gastrointestinal administration preparation of the present invention refers to after oral administration.
  • a pharmaceutical dosage form that enters the gastrointestinal tract and acts locally or absorbed to exert a systemic effect including but not limited to powders, tablets, granules, capsules, solutions, emulsions, suspensions, and the like;
  • Formulations include, but are not limited to, sprays, aerosols, powders, and the like;
  • the dermal administration preparations of the present invention include, but are not limited to, external solutions, lotions, tinctures, ointments, plasters, pastes, patches
  • the mucosal administration dosage form of the present invention includes, but is not limited to, eye drops, nasal drops, ophthalmic ointments, gargles, sublingual tablets, adhesive tablets and filming agents, etc.;
  • Pharmaceutical preparations include, but are not limited to, suppositories, aerosols, effervescent tablets, drops, and pills.
  • the protein kinase C inhibitor is selected from the group consisting of militalin, a derivative thereof, a pharmaceutically acceptable salt thereof, a solvate thereof, and a prodrug thereof.
  • the militalin is orally administered 1-3 times a day, each dose is 50-100 mg, and continuous administration is 10-18 days for one course of treatment; more preferably daily Oral administration twice, 50-100 mg each time, for 14 days for one course of treatment; further, the dosage of the mexetiline is 1-4 mg/Kg/day.
  • the rituximab is intravenous, 350-400 mg/m 2 body surface area, more preferably 375 mg/m 2 body surface area, once a week, one for each week Treatment.
  • the present invention also provides a protein kinase C inhibitor for use in the treatment of rituximab-resistant non-Hodgkin's lymphoma, and in the preparation of non-Hodgkin-resistant rituximab The application of the drug for the treatment of lymphoma.
  • the present invention also provides the use of rituximab and a protein kinase C inhibitor for the treatment of non-Hodgkin's lymphoma, and for the preparation of a combination therapy for non-Hodgkin's lymphoma.
  • non-Hodgkin's lymphoma is a B cell non-Hodgkin's lymphoma, or the non-Hodgkin's lymphoma is a non-Hodgkin's lymphoma resistant to rituximab, Alternatively, the non-Hodgkin's lymphoma is a non-Hodgkin's lymphoma with highly activated PKC.
  • PKC protein kinase C
  • pan-PKC inhibitors broad-spectrum PKC inhibitors
  • the combination of monoclonal antibodies can effectively trigger apoptosis in BL cells and significantly improve overall survival, especially in BL cells that are resistant to rituximab; this result can be further extended to resistance to rituximab Non-Hodgkin's lymphoma, or non-Hodgkin's lymphoma with highly activated PKC.
  • Figure 1 is a Western blot of mCRPs expression in Ramos cells and Ramos 640 cells;
  • Figure 2 is a comparison of FACS analysis of mCRPs expression in Ramos cells and Ramos640 cells: compared with the original cells, the expression of CD20 is decreased, the expression of CD59 is increased, the expression of CD55 is slightly decreased, and the expression of CD46 is not different.
  • Figure 3 is a Western blot of mCRPs expression in Raji cells and Raji32 cells
  • Figure 5 is a Western blot of Akt phosphorylation and mCRPs expression in four groups of cells before and after PI3K inhibitor IPI-145 (1 ⁇ M): phosphorylation of Akt (S473) in Ramos640 cells and Raji32 cells compared to their original cells An increase in level, which is inhibited by treatment with the PI3K inhibitor IPI-145, resulting in decreased expression of CD20 and CD55;
  • Figure 8 is a bar graph of up-regulated greater than 1.5-fold phosphorylated protein and its phosphorylation site in a phosphorylated antibody microarray
  • Figure 9 is an IPA (Ingenuity Pathway Analysis) analysis result of the up-regulated protein of Figure 8, the solid line indicates direct adjustment, and the broken line indicates indirect regulation;
  • Figure 10 is a GSEA enrichment profile of anti-apoptotic TNFs:NF- ⁇ B:Bcl-2 pathway obtained after comparison of Ramos640 cells with Ramos cell RNA-Seq data, FDR ⁇ 0.25 is considered significant;
  • Figure 11 is a GSEA enrichment characteristic map of the p53 pathway obtained by comparing Ramos640 cells with Ramos cell RNA-Seq data, and FDR ⁇ 0.25 is considered significant;
  • Figure 12 is a Western blot of PKC ⁇ / ⁇ 1/ ⁇ 2/ ⁇ / ⁇ expression in four BL cells and phosphorylation of PKC and its downstream anti-apoptotic proteins and the effect of PCK inhibitor methotrexate (1 ⁇ M) on them.
  • This figure shows that up-regulation of PKC isoforms in drug-resistant cells leads to an increase in PKC phosphorylation and activates downstream anti-apoptotic proteins, while midazolin inhibits PKC and its downstream signaling;
  • Figure 13 is a graph showing the results of apoptosis induced by the pan-PKC inhibitor militalin in Ramos cells and Ramos 640 cells, where Control: medium control, RTX: rituximab (640 ⁇ g/mL), Mido: rice bran Tolin (1 ⁇ M); this figure demonstrates that pan-PKC inhibitors effectively induce apoptosis in na[iota]ve and resistant Ramos cells, and the addition of rituximab to midazoline significantly enhances Ramos640 cells.
  • Figure 15 is a Western blot of the effect of the PKC inhibitor, mitutoline (1 ⁇ M), on the expression of mCRPs in four cells, which shows that midazolin significantly reduces CD20 expression in primary and drug-resistant cells as well as in resistant cells. CD59 expression, but did not alter the expression of CD55 and CD46 in protocell or resistant cells;
  • Figure 18 is the growth of tumors on day 50 after inoculation of Raji32-Luc cells, the tumor mass is represented by the intensity of firefly luciferase activity, RTX: rituximab, Mido: methotrexate;
  • Figure 20 is the growth of tumors on day 70 after inoculation of Raji32-Luc cells, the tumor mass is represented by the intensity of firefly luciferase activity, "X” represents mouse death, RTX: rituximab, Mido: rice bran forest;
  • Figure 22 shows tumor growth on day 90 after inoculation of Raji32-Luc cells. Tumor quality is expressed by the intensity of firefly luciferase activity, "X" represents death of mice, RTX: rituximab, Mido: rice bran forest;
  • Rituximab-mediated ADCC is carried out by immune cells from individual patients whose tolerance to ADCC is caused by the intrinsic characteristics of immune cells, such as the FcyRIIIa gene polymorphism. Apoptosis plays only a negligible role in the antitumor activity of rituximab. Therefore, we constructed two BL cell lines, Ramos640 and Raji32, which are resistant to rituximab-mediated complement-dependent cytotoxicity at rituximab concentrations of 640 and 32 ⁇ g/mL, respectively. The construction method is as follows:
  • Both BL cell lines Raji and Ramos were purchased from American type culture collection (ATCC) (Manassas, VA), and the cells were cultured in 10% (volume) fetal bovine serum (GIBCO BRL, Grand Island, NY) and 1% (by volume) penicillin/streptomycin (Ambion, Austin, TX) in RPMI 1640 medium.
  • normal human serum (NHS) was collected from 10 healthy humans and combined to serve as a source of complement. The above serum was aliquoted and stored at -80 ° C until use. In addition, heat-killed human serum (IHS) prepared by incubating the above serum for 30 minutes in a 65 ° C water bath was used as a negative control.
  • IHS heat-killed human serum
  • Raw Raji cells and Ramos cells were separately treated with increasing concentrations of rituximab (Roche, Basel, Switzerland).
  • concentrations were increased from 4 or 40 ⁇ g/mL, respectively, to a multiple of 2 or more to 32 or 640 ⁇ g/mL, and the resulting drug-resistant cells were called Raji32 and Ramos 640, respectively.
  • They were treated with 32 ⁇ g/mL and 640 ⁇ g/mL rituximab containing 20% (by volume) of NHS every 21 days to maintain resistance to rituximab.
  • the present invention was subjected to immunoblot analysis according to standard methods.
  • the CDC effect was determined by fluorescence-activated cell sorting (FACS) analysis to detect propidium iodide (PI) staining positive cells. Specifically, after washing with PBS, the cells were incubated with fluorescein-conjugated antibody for 30 minutes, then rinsed and resuspended in PBS. Flow cytometric analysis was performed on a Cytomics FC500 MPL flow cytometer (Beckman Coulter, Brea, CA) and analyzed using FlowJo software (Ashland, OR).
  • FACS fluorescence-activated cell sorting
  • the present invention further studies the mechanism of drug resistance by protein signaling pathway analysis.
  • This example uses a phosphorylated antibody microarray to identify functional proteins and signaling pathways that are activated in Ramos 640 cells.
  • the PI3K inhibitor IPI-145 was used to identify whether inhibition of PI3K can increase the sensitivity of resistant BL cells to rituximab treatment.
  • the PI3K inhibitor IPI-145 was purchased from Selleck Chemicals (Houston, TX).
  • the original cells and the drug-resistant cells were seeded into 96-well plates at a density of 1 ⁇ 10 4 cells/100 ⁇ L/well, and were divided into four groups, and the first group was cultured for 48 hours as a control, The two groups were treated with a medium containing IPI-145 at a concentration of 1 ⁇ M for 48 hours, and the third group was treated with a medium containing 20% (by volume) of NHS and rituximab at a concentration of 640 ⁇ g/mL or 32 ⁇ g/mL for 48 hours.
  • the fourth group was treated with a medium containing 20% (by volume) of NHS and having a rituximab concentration of 640 ⁇ g/mL or 32 ⁇ g/mL and an IPI-145 concentration of 1 ⁇ M for 48 hours.
  • CytoTox-Glo TM cytotoxicity assay kit Promega Corporation, Madison, WI
  • cytotoxicity assay performed according to its technical bulletin steps and cytotoxicity was calculated according to the following formula:
  • Cytotoxicity (%) dead cell luminescence / total luminescence ⁇ 100%
  • the data of the present invention are expressed as mean ⁇ standard deviation unless otherwise stated. Significant differences between the two groups were determined using a two-tailed Student's t-test of unpaired data. In all analyses, p ⁇ 0.05 was considered to be statistically significant.
  • Example 5 PKC-mediated apoptotic pathway is highly activated in drug-resistant cells
  • B-NHL B-cell non-Hodgkin's lymphoma
  • the present invention uses gene set enrichment analysis GSEA software (the Massachusetts Institute of Technology Broad Institute) to identify the function of differentially expressed genes found in RNA-seq. A predetermined version of the software was used to identify significantly enriched pathways, and an enrichment pathway with FDR ⁇ 0.25 was considered significant.
  • Anti-apoptotic TNFs used in the present invention: NF- ⁇ B: Bcl-2 pathway gene set is derived from the PathCards Pathway Uniform Database (version 4.0.6.37, Weizmann Academy of Sciences) "Apoptosis and Survival Anti-apoptotic TNFs: NF- ⁇ B : Bcl-2 pathway SuperPath" consists of 42 genes.
  • the p53 pathway gene set used in the present invention consists of 132 genes from the "p53 pathway (RnD) SuperPath" in the unified database of the PathCards pathway.
  • Mitalin is a multi-kinase inhibitor originally designed to inhibit PKC and is currently approved for acute myeloid leukemia (AML) and advanced systemic mast cells with FLT3 (Fms-like tyrosine kinase 3) mutations. Hyperplasia (SM).
  • the PKC inhibitor mitutolin used in the present invention was purchased from Selleck Chemicals (Houston, TX).
  • midazolin significantly increased or decreased the expression of Bad or p65 in all four primordial or drug-resistant cells, and reduced Bcl-2 expression only in naive and resistant Raji cells ( Figure 12).
  • militalin inhibits PKC and its downstream signaling, which may lead to pro-apoptotic effects in Ramos and Raji cells to varying degrees.
  • Raji cells appear to be more resistant to midazolin-mediated PKC phosphorylation than Ramos cells because midazolin failed to inhibit PKC phosphorylation.
  • pan-PKC inhibitor militalin (1 ⁇ M) alone or in combination with rituximab (32 ⁇ g/mL for Raji cells and 640 ⁇ g/mL for Ramos cells). Pro-apoptotic effects on naive and resistant BL cells.
  • the original cells and the drug-resistant cells were seeded into 96-well plates at a density of 1 ⁇ 10 4 cells/100 ⁇ L/well, and were divided into four groups, and the first group was cultured for 48 hours as a control, The two groups were treated with rituximab at a concentration of 640 ⁇ g/mL or 32 ⁇ g/mL for 48 hours, and the third group was treated with a medium containing 1 ⁇ M of miltazone for 48 hours. The medium was treated with a concentration of 640 ⁇ g/mL or 32 ⁇ g/mL and a midazorine concentration of 1 ⁇ M for 48 hours.
  • CytoTox-Glo TM cytotoxicity assay kit Promega Corporation, Madison, WI
  • cytotoxicity assay performed according to its technical bulletin steps and cytotoxicity was calculated according to the following formula:
  • Cytotoxicity (%) dead cell luminescence / total luminescence ⁇ 100%
  • the data of the present invention are expressed as mean ⁇ standard deviation unless otherwise stated. Significant differences between the two groups were determined using a two-tailed Student's t-test of unpaired data. In all analyses, p ⁇ 0.05 was considered to be statistically significant.
  • Example 7 Mitalantin enhances susceptibility of resistant BL cells to rituximab-mediated CDC
  • pan-PKC inhibitor midazolin
  • the original cells and the drug-resistant cells were seeded into 96-well plates at a density of 1 ⁇ 10 4 cells/100 ⁇ L/well, and were divided into four groups, and the first group was cultured for 48 hours as a control, The two groups were treated with a medium containing 20% (by volume) of NHS and a concentration of rituximab of 640 ⁇ g/mL or 32 ⁇ g/mL for 48 hours, and the third group was treated with a medium containing 1 ⁇ M of metoprolin.
  • the fourth group was treated with a medium containing 20% (by volume) of NHS and having a rituximab concentration of 640 ⁇ g/mL or 32 ⁇ g/mL and a myristicin concentration of 1 ⁇ M for 48 hours.
  • CytoTox-Glo TM cytotoxicity assay kit Promega Corporation, Madison, WI
  • cytotoxicity assay performed according to its technical bulletin steps and cytotoxicity was calculated according to the following formula:
  • Cytotoxicity (%) dead cell luminescence / total luminescence ⁇ 100%
  • the data of the present invention are expressed as mean ⁇ standard deviation unless otherwise stated. Significant differences between the two groups were determined using a two-tailed Student's t-test of unpaired data. In all analyses, p ⁇ 0.05 was considered to be statistically significant.
  • rituximab-mediated CDC can be regulated by expression of mCRP, such as CD46, CD55, and especially CD59.
  • mCRP such as CD46, CD55, and especially CD59.
  • midazolam (1 ⁇ M) significantly reduced the expression of CD20 in all four BL cells, whereas the decrease in CD59 expression was predominant in drug-resistant cells; it had no effect on the expression of CD55 and CD46 ( Figure 15).
  • midazolin may block rituximab-mediated CDC due to decreased expression of CD20 in naive BL cells, whereas expression of both CD20 and CD59 is reduced in resistant BL cells. Therefore, its effect on resistant BL cells requires further testing.
  • these results show the difference between the effects of IPI-145 and midazolam on the regulation of CD20 and CD59 expression, in which IPI-145 only reduces CD20 expression in resistant BL cells but does not reduce CD59 expression (Fig. 5).
  • Rituximab (a concentration of 32 ⁇ g/mL for Raji cells and 640 ⁇ g/mL for Ramos cells) plus NHS (20%, v/v) effectively induced CDC in naive Ramos cells and Raji cells.
  • militalin (1 ⁇ M) failed to enhance the susceptibility of rituximab-mediated CDC, although it promoted apoptosis ( Figures 16 and 17). This may be due to a decrease in CD20 expression and a already high cell death rate due to rituximab alone.
  • rituximab significantly enhanced the cytotoxic effect of militalin in the original BL cells ( Figure 16 and Figure 17), indicating that the effect of rituximab-mediated CDC is greater in primary BL cells. Mitotalin induces apoptosis.
  • rituximab and midazoline induced 17.1% and 54.3% cell death, respectively, and their combination induced 72.8% cell death.
  • rituximab and midazoline induced 28.0% and 25.8% cell death, respectively, and their combination induced 62.9% cell death. This result may be due to the unique anti-tumor mechanism of rituximab and midazolam.
  • the CDS (coding sequence) of the firefly luciferase gene was obtained from the pGL3-Basic plasmid by PCR amplification, and inserted into the pCDH cDNA clone and expression vector by EcoRI and BamHI endonuclease sites.
  • the primers for firefly luciferase CDS amplification are as follows:
  • the pCDH plasmid was co-transfected with the pMD.2G and psPAX2 plasmids in 293FT cells to generate firefly luciferase overexpressing lentivirus.
  • the lentivirus was then added to Raji32 cell culture medium for 48 hours. All cells transfected with lentivirus in the present invention were classified using GFP and MoFlo XDP instruments (Beckman Coulter, Brea, CA) and referred to as Raji32-Luc cells.
  • mice Eight-week-old female SCID mice were purchased from SLAC (Shanghai Laboratory Animal Center, Shanghai Experimental Animal Center). Raji32-Luc cells were resuspended in PBS, and then each mouse was intraperitoneally injected with 1.5 x 10 7 cells. Mice were divided into 4 groups (7 mice per group) based on the difference in drug administration, namely saline, rituximab, midazoline and rituximab plus militalin. On the 8th, 12th and 16th day after tumor inoculation, 118.4 mg/kg rituximab was intraperitoneally injected, and 8, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18 after tumor inoculation.
  • mice On days 19, 20 and 21, the rats were intragastrically administered with miltazone 20 mg/kg. The amount of saline administered was the same as that of rituximab. Tumor growth was monitored by bioluminescence on days 50, 70, and 90 after tumor inoculation, and D-luciferin (Promega, Madison, WI) was intraperitoneally injected into mice (150 mg/kg), and ten minutes later, intraperitoneal injection was performed. Mice were anesthetized with pentobarbital (50 mg/kg) and bioluminescence detection was then performed using an In-Vivo MS FX PRO system (Bruker, Billerica, MA).
  • D-luciferin Promega, Madison, WI
  • Luminescence images were captured with an exposure time of 30 seconds and the signal intensity of the tumors was measured by Bruker MI software. The survival time of each mouse was recorded up to 120 days. All animal experiments were carried out in strict accordance with the experimental protocol approved by the Animal Ethics Committee of Shanghai Medical College of Fudan University.
  • the data of the present invention are expressed as mean ⁇ standard deviation unless otherwise stated.
  • significance was determined by a one-tailed Mann Whitney test.
  • Mantel-Cox test was used to compare the survival rates of the two groups of xenograft models. In all analyses, p ⁇ 0.05 was considered to be statistically significant.
  • the total photon flux is used to express the growth of the tumor according to the saline control group, rituximab group, rice bran.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Endocrinology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed is a pharmaceutical composition for treating non-Hodgkin's lymphoma, comprising a protein kinase C inhibitor and rituximab. Also disclosed is a pharmaceutical composition for treating non-Hodgkin's lymphoma, comprising a first preparation formed by a protein kinase C inhibitor and a pharmaceutically acceptable carrier, and a second preparation formed by rituximab and a pharmaceutically acceptable carrier. By combining the discovery that PKC is highly phosphorylated in rituximab-resistant non-Hodgkin's lymphoma cells, and the principle of pro-apoptotic effects of a PKC inhibitor, the present invention proves, by means of researches, that the protein kinase C inhibitor can be used, alone or in combination with rituximab, for treating rituximab-tolerant relapsed/refractory non-Hodgkin's lymphoma and significantly improves the overall survival.

Description

[根据细则37.2由ISA制定的发明名称] 治疗非霍奇金氏淋巴瘤的药物组合物[Name of invention established by ISA according to Rule 37.2] Pharmaceutical composition for treating non-Hodgkin's lymphoma 技术领域Technical field
本发明涉及一种治疗非霍奇金氏淋巴瘤的药物组合物,特别涉及一种治疗B细胞非霍奇金氏淋巴瘤的药物组合物。The present invention relates to a pharmaceutical composition for treating non-Hodgkin's lymphoma, and more particularly to a pharmaceutical composition for treating B cell non-Hodgkin's lymphoma.
背景技术Background technique
伯基特淋巴瘤(Burkitt’s lymphoma,BL)是一种高度侵袭性的B细胞非霍奇金氏淋巴瘤(non-Hodgkin’s lymphoma,NHL),占所有年龄组淋巴瘤的3-5%,占儿童淋巴瘤的40-50%,其特征在于高表达的c-MYC靶标和生发中心相关的B细胞基因,以及低表达的MHC-I分子和NF-κB靶基因。成人BL患者对CHOP(环磷酰胺、多柔比星、长春新碱和泼尼松龙)为基础的治疗方案反应较差,2年和5年总生存率(OS)约为50-65%,若波及到骨髓或中枢神经系统则进一步降低至30%以下。相比之下,强化短期化疗方案将儿童BL患者的存活率显着提高至超过90%,在成人BL患者中,类似的治疗方案提高存活率至70%以上。然而,这些治疗方案却不可避免地会遇到药物毒性和耐药性的障碍。Burkitt's lymphoma (BL) is a highly invasive B-cell non-Hodgkin's lymphoma (NHL), accounting for 3-5% of lymphoma in all age groups, accounting for children. 40-50% of lymphomas are characterized by highly expressed c-MYC targets and germinal center-associated B cell genes, as well as low expressed MHC-I molecules and NF-κB target genes. Adult BL patients have a poor response to CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisolone)-based treatment regimens with a 2-year and 5-year overall survival (OS) of approximately 50-65% If it reaches the bone marrow or central nervous system, it will be further reduced to less than 30%. In contrast, the intensive short-term chemotherapy regimen significantly increased the survival rate of children with BL to more than 90%, and in adult BL patients, a similar treatment regimen improved survival to more than 70%. However, these treatment options inevitably encounter barriers to drug toxicity and drug resistance.
利妥昔单抗联合CHOP化疗(R-CHOP)可使弥漫性大B细胞淋巴瘤(DLBCL)患者的总生存率提高至少20%。同样,许多单臂临床试验测试并进一步证实了利妥昔单抗对于BL强化短期化疗方案的效果。最近的一项III期临床试验表明,化疗加上利妥昔单抗比单独化疗可达到更高的3年无事件生存率(75%对62%,P=0.024)和3年总生存率(83%对70%,P=0.011)。因此,增加利妥昔单抗在未来BL治疗方案的设计中备受期待,从而在该治疗过程中对利妥昔单抗耐药性的产生也将与其治疗DLBCL一样,预期也会出现。Rituximab combined with CHOP chemotherapy (R-CHOP) can increase overall survival in patients with diffuse large B-cell lymphoma (DLBCL) by at least 20%. Similarly, many single-arm clinical trials have further demonstrated the efficacy of rituximab for BL-enhanced short-term chemotherapy regimens. A recent phase III clinical trial showed that chemotherapy plus rituximab achieved a higher 3-year event-free survival rate (75% vs. 62%, P=0.024) and 3-year overall survival rate compared with chemotherapy alone ( 83% vs. 70%, P = 0.011). Therefore, the increase in rituximab is expected in the design of future BL treatment regimens, so that the production of rituximab resistance during this treatment will be the same as the treatment of DLBCL, and it is expected to occur.
尽管利妥昔单抗在治疗各种各样的B细胞淋巴瘤方面取得了巨大的成功,然而对利妥昔单抗的耐药性却是复发/难治性患者的一大挑战,且仍然约有50%的患者对利妥昔单抗的治疗无反应,而最初有应答的患者最终会对进一步的利妥昔单抗的治疗产生耐药性。Although rituximab has achieved great success in treating a wide variety of B-cell lymphomas, resistance to rituximab is a major challenge for relapsed/refractory patients and remains Approximately 50% of patients do not respond to treatment with rituximab, and patients who initially respond will eventually develop resistance to further treatment with rituximab.
通过与膜CD20结合,利妥昔单抗主要通过抗体依赖性细胞毒性(ADCC)和补体依赖性细胞毒性(CDC)杀伤淋巴细胞。对ADCC的耐药可能来源于IgG Fc受体FcgammaRIIIa基因的内在多态性,而对CDC的耐药很可能归因于CD20的表达下调和膜补体调节蛋白(mCRPs)特别是CD59的表达上调。Rituximab kills lymphocytes primarily by antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) by binding to membrane CD20. Resistance to ADCC may be due to the intrinsic polymorphism of the Fc Fc receptor FcgammaRIIIa gene, and resistance to CDC is likely due to down-regulation of CD20 expression and upregulation of membrane complement regulatory proteins (mCRPs), particularly CD59.
为了改善利妥昔单抗的治疗功效,本领域研究人员已经作出相当大的努力来提高CD20的表达,例如通过组蛋白脱乙酰酶抑制剂曲古抑菌素A或合成CpG寡脱氧核苷 酸,或通过修饰的单克隆抗体或细菌毒素衍生的ILYd4抑制CD59的功能。然而,其中只有CpG寡脱氧核苷酸在临床试验(I期)中进一步评估其在B细胞NHL患者中的安全性,并且没有进一步的临床试验报道。因此,需要寻找替代的治疗策略。因此,一方面,为了提高利妥昔单抗的抗肿瘤效果,另一方面,为了逆转对利妥昔单抗的耐药,本领域技术人员致力于开发一种可与利妥昔单抗组合的促凋亡剂用于B细胞淋巴瘤的治疗。In order to improve the therapeutic efficacy of rituximab, researchers in the field have made considerable efforts to improve the expression of CD20, such as by the histone deacetylase inhibitor trichostatin A or synthetic CpG oligodeoxynucleotides. , or the function of CD59 is inhibited by modified monoclonal antibody or bacterial toxin-derived ILYd4. However, only CpG oligodeoxynucleotides were further evaluated for safety in B-cell NHL patients in clinical trials (stage I) and no further clinical trials have been reported. Therefore, there is a need to find alternative treatment strategies. Therefore, on the one hand, in order to improve the antitumor effect of rituximab, on the other hand, in order to reverse the resistance to rituximab, those skilled in the art are working to develop a combination with rituximab. Pro-apoptotic agents are used in the treatment of B-cell lymphoma.
发明内容Summary of the invention
有鉴于现有技术中存在的上述问题,本发明的目的是提供一种治疗复发/难治性非霍奇金氏淋巴瘤(特别是对利妥昔单抗耐药的非霍奇金氏淋巴瘤)的药物组合物。In view of the above problems in the prior art, it is an object of the present invention to provide a treatment for relapsed/refractory non-Hodgkin's lymphoma (especially non-Hodgkin's lymphoma resistant to rituximab) A pharmaceutical composition of a tumor.
为实现上述目的,本发明首先提供了一种治疗非霍奇金氏淋巴瘤的药物组合物,包括蛋白激酶C(PKC)抑制剂和利妥昔单抗。To achieve the above object, the present invention first provides a pharmaceutical composition for treating non-Hodgkin's lymphoma, including a protein kinase C (PKC) inhibitor and rituximab.
进一步,本发明所述治疗非霍奇金氏淋巴瘤的药物组合物还包括药学上可接受的载体或赋形剂。Further, the pharmaceutical composition for treating non-Hodgkin's lymphoma of the present invention further comprises a pharmaceutically acceptable carrier or excipient.
进一步,本发明所述蛋白激酶C抑制剂包括但不限于:星形孢菌素及其类似物、吲哚卡巴唑类化合物、双吲哚马来酰胺类化合物、双吲哚马来酰胺大环化合物、2-烷基吲哚马来酰亚胺类化合物、Balanol类化合物、N-苯基-2-嘧啶胺类衍生物、Rottlerin、H-series抑制剂、磺胺和磺酰苯甲酰衍生物、单吲哚马来酰胺衍生物、腺苷-5’-端羧酸肽衍生物。Further, the protein kinase C inhibitor of the present invention includes, but is not limited to, staurosporin and its analogs, carbamazepines, biguanide maleamides, biguanide maleamide macrocycles Compounds, 2-alkylindole maleimide compounds, Balanol compounds, N-phenyl-2-pyrimidinamine derivatives, Rottlerin, H-series inhibitors, sulfonamides and sulfonylbenzoyl derivatives , a monoterpene maleimide derivative, an adenosine-5'-terminal carboxylic acid peptide derivative.
优选地,所述蛋白激酶C抑制剂选自米哚妥林、其衍生物、其药学上可接受的盐、其溶剂合物和其前药。Preferably, the protein kinase C inhibitor is selected from the group consisting of militalin, a derivative thereof, a pharmaceutically acceptable salt thereof, a solvate thereof, and a prodrug thereof.
米哚妥林(Midostaurin)是一种广谱的PKC抑制剂(PKC412),其来自于被修饰的星形孢菌素,以提高对PKC的选择性,并且进一步可以抑制其它激酶如FLT3、PDGFR、KIT和VEGFR2及其代谢物。在I期临床试验中,米哚妥林慢性口服治疗是安全和可耐受的,并且由FDA在临床治疗中证实;其具有如下式I所示的化学结构:Midostaurin is a broad-spectrum PKC inhibitor (PKC412) derived from modified staurosporins to increase selectivity for PKC and further inhibit other kinases such as FLT3, PDGFR , KIT and VEGFR2 and their metabolites. In Phase I clinical trials, chronic oral treatment with midazolam is safe and tolerable and confirmed by the FDA in clinical treatment; it has the chemical structure shown in Formula I below:
Figure PCTCN2019082102-appb-000001
Figure PCTCN2019082102-appb-000001
2017年4月,米哚妥林被批准用于治疗伴FLT3(Fms样酪氨酸激酶3)突变的急性骨髓性白血病(AML)和晚期系统性肥大细胞增多症(SM)。In April 2017, midazoline was approved for the treatment of acute myeloid leukemia (AML) and advanced systemic mastocytosis (SM) with FLT3 (Fms-like tyrosine kinase 3) mutations.
本发明人在研究中发现米哚妥林单独或与利妥昔单抗联合用药可用于BL患者的 治疗,特别是用于复发/难治性BL患者的治疗,以及进一步可用于其它具有高度活化的PKC的非霍奇金氏淋巴瘤患者的治疗,特别是用于具有高度活化的PKC的复发/难治性B细胞非霍奇金氏淋巴瘤。The present inventors have found in the study that midazolam alone or in combination with rituximab can be used for the treatment of patients with BL, especially for patients with relapsed/refractory BL, and further for other highly active Treatment of patients with non-Hodgkin's lymphoma of PKC, especially for relapsed/refractory B-cell non-Hodgkin's lymphoma with highly activated PKC.
尽管利妥昔单抗对淋巴细胞直接诱导细胞毒性的作用有时是有争议的,但许多研究表明利妥昔单抗在各种NHL细胞中缺乏促凋亡能力。我们的一系列研究揭示利妥昔单抗在包括BL细胞在内的非霍奇金氏淋巴瘤中未能诱导细胞凋亡达到任何可检测的程度,因此,细胞凋亡在利妥昔单抗抗肿瘤活性中所起的作用非常有限。因此,我们尝试通过结合其它具有促凋亡效应的药物来提高利妥昔单抗疗法的敏感性。大分子试剂包括靶向TRAIL-R1的人源化单克隆马帕木单抗(mapatumumab)、基因工程融合蛋白scFvRit:sFasL、Apo2配体(Apo2L)/TRAIL(dulanermin)和抗CD20-白细胞介素-21,以及小分子试剂包括选择性NEDD8活化酶(NAE)抑制剂pevonedistat(MLN4924)、mTOR抑制剂temsirolimus和蛋白酶体抑制剂bortezomib,增强了利妥昔单抗对B细胞NHL细胞的抗肿瘤活性。然而,它们的疗效需要进一步证实,或在临床试验中被证实为无效。Although the effect of rituximab on the direct induction of cytotoxicity by lymphocytes is sometimes controversial, many studies have shown that rituximab lacks pro-apoptotic ability in various NHL cells. Our series of studies have revealed that rituximab fails to induce apoptosis to any detectable extent in non-Hodgkin's lymphoma, including BL cells, and therefore apoptosis in rituximab The role played by antitumor activity is very limited. Therefore, we attempted to increase the sensitivity of rituximab therapy by combining other drugs with pro-apoptotic effects. Macromolecular reagents include humanized monoclonal mapramumab targeting TRAIL-R1, genetically engineered fusion protein scFvRit: sFasL, Apo2L/TRAIL (dulanermin), and anti-CD20-interleukin -21, and small molecule reagents including selective NEDD8 activating enzyme (NAE) inhibitor pevonedistat (MLN4924), mTOR inhibitor temsirolimus and proteasome inhibitor bortezomib, enhance the antitumor activity of rituximab on B cell NHL cells . However, their efficacy needs to be further confirmed or proven to be ineffective in clinical trials.
最终,本发明人在研究中发现米哚妥林通过促进BL细胞中的细胞凋亡而强烈增强利妥昔单抗的细胞毒性作用。Finally, the inventors found in the study that midazolin strongly enhanced the cytotoxic effect of rituximab by promoting apoptosis in BL cells.
考虑到PKC在耐药BL细胞中高度磷酸化,我们测试了米哚妥林在原始和利妥昔单抗耐药的BL细胞中对利妥昔单抗抗肿瘤活性的作用,实验结果证实其在体外和体内都能强烈增强利妥昔单抗的抗肿瘤活性(尤其是在妥昔单抗耐药BL细胞中),进一步我们发现米哚妥林是通过促进细胞凋亡来增强利妥昔单抗的细胞毒性的,其可能是通过改变下游信号传导通路(包括Bad、Bcl-2和NF-κB)的磷酸化来实现的。Considering the high phosphorylation of PKC in resistant BL cells, we tested the effect of midazolam on the antitumor activity of rituximab in naive and rituximab-resistant BL cells, and the experimental results confirmed its The anti-tumor activity of rituximab was strongly enhanced in vitro and in vivo (especially in rituximab-resistant BL cells), and further we found that methotrex enhances rituximab by promoting apoptosis. The cytotoxicity of monoclonal antibodies may be achieved by altering the phosphorylation of downstream signaling pathways, including Bad, Bcl-2 and NF-κB.
基于上述研究结果,本发明还提供了一种治疗非霍奇金氏淋巴瘤的药物组合物,其包括不同规格单位制剂的利妥昔单抗和蛋白激酶C抑制剂及药学上可接受的载体或赋形剂,用于同时、分别或依次给药。Based on the above findings, the present invention also provides a pharmaceutical composition for treating non-Hodgkin's lymphoma comprising rituximab and protein kinase C inhibitors of different specification unit preparations and a pharmaceutically acceptable carrier Or excipients for simultaneous, separate or sequential administration.
进一步,本发明提供了一种治疗非霍奇金氏淋巴瘤的药物组合物,包括蛋白激酶C抑制剂和药学上可接受的载体形成的第一制剂,及利妥昔单抗和药学上可接受的载体形成的第二制剂。Further, the present invention provides a pharmaceutical composition for treating non-Hodgkin's lymphoma, comprising a first formulation of a protein kinase C inhibitor and a pharmaceutically acceptable carrier, and rituximab and a pharmaceutically acceptable A second formulation of the accepted carrier formed.
可选地,上述制剂的剂型为注射给药制剂、经胃肠道给药制剂、呼吸道给药制剂、皮肤给药制剂、粘膜给药制剂或腔道给药制剂。Alternatively, the dosage form of the above preparation is an injection administration preparation, a gastrointestinal administration preparation, a respiratory administration preparation, a dermal administration preparation, a mucosal administration preparation or a channel administration preparation.
其中,本发明所述注射给药制剂包括但不限于静脉注射、肌内注射、皮下注射、皮内注射及腔内注射等;本发明所述经胃肠道给药制剂是指经口服用后进入胃肠道,起局部或经吸收而发挥全身作用的药物剂型,包括但不限于散剂、片剂、颗粒剂、胶囊剂、溶液剂、乳剂、混悬剂等;本发明所述呼吸道给药制剂包括但不限于喷雾剂、气雾剂、粉雾剂等;本发明所述皮肤给药制剂包括但不限于外用溶液剂、洗剂、搽剂、软膏剂、硬膏剂、糊剂、贴剂等;本发明所述粘膜给药剂型包括但不限于滴眼剂、滴 鼻剂、眼用软膏剂、含漱剂、舌下片剂、粘贴片及贴膜剂等;本发明所述腔道给药制剂包括但不限于栓剂、气雾剂、泡腾片、滴剂及滴丸剂等。Wherein, the injection administration preparation of the present invention includes, but is not limited to, intravenous injection, intramuscular injection, subcutaneous injection, intradermal injection, intraluminal injection, and the like; and the gastrointestinal administration preparation of the present invention refers to after oral administration. A pharmaceutical dosage form that enters the gastrointestinal tract and acts locally or absorbed to exert a systemic effect, including but not limited to powders, tablets, granules, capsules, solutions, emulsions, suspensions, and the like; Formulations include, but are not limited to, sprays, aerosols, powders, and the like; the dermal administration preparations of the present invention include, but are not limited to, external solutions, lotions, tinctures, ointments, plasters, pastes, patches The mucosal administration dosage form of the present invention includes, but is not limited to, eye drops, nasal drops, ophthalmic ointments, gargles, sublingual tablets, adhesive tablets and filming agents, etc.; Pharmaceutical preparations include, but are not limited to, suppositories, aerosols, effervescent tablets, drops, and pills.
优选地,所述蛋白激酶C抑制剂选自米哚妥林、其衍生物、其药学上可接受的盐、其溶剂合物和其前药。Preferably, the protein kinase C inhibitor is selected from the group consisting of militalin, a derivative thereof, a pharmaceutically acceptable salt thereof, a solvate thereof, and a prodrug thereof.
在本发明的一种优选实施方式中,所述米哚妥林为口服,每日1-3次,每次剂量为50-100mg,连续服用10-18天为一个疗程;更优选为每日口服2次,每次50-100mg,连续服用14天为一个疗程;进一步,所述米哚妥林的服用剂量为1-4mg/Kg/天。In a preferred embodiment of the present invention, the militalin is orally administered 1-3 times a day, each dose is 50-100 mg, and continuous administration is 10-18 days for one course of treatment; more preferably daily Oral administration twice, 50-100 mg each time, for 14 days for one course of treatment; further, the dosage of the mexetiline is 1-4 mg/Kg/day.
在本发明的另一种优选实施方式中,所述利妥昔单抗为静脉注射,350-400mg/m 2体表面积,更优选为375mg/m 2体表面积,每周注射一次,四周为一个疗程。 In another preferred embodiment of the present invention, the rituximab is intravenous, 350-400 mg/m 2 body surface area, more preferably 375 mg/m 2 body surface area, once a week, one for each week Treatment.
进一步,本发明还提供了蛋白激酶C抑制剂在治疗对利妥昔单抗耐药的非霍奇金氏淋巴瘤中的应用,以及在制备对利妥昔单抗耐药的非霍奇金氏淋巴瘤治疗药物中的应用。Further, the present invention also provides a protein kinase C inhibitor for use in the treatment of rituximab-resistant non-Hodgkin's lymphoma, and in the preparation of non-Hodgkin-resistant rituximab The application of the drug for the treatment of lymphoma.
进一步,本发明还提供了利妥昔单抗和蛋白激酶C抑制剂在治疗非霍奇金氏淋巴瘤中的应用,以及在制备非霍奇金氏淋巴瘤治疗联合用药物中的应用。Further, the present invention also provides the use of rituximab and a protein kinase C inhibitor for the treatment of non-Hodgkin's lymphoma, and for the preparation of a combination therapy for non-Hodgkin's lymphoma.
其中,所述非霍奇金氏淋巴瘤为B细胞非霍奇金氏淋巴瘤,或者所述非霍奇金氏淋巴瘤为对利妥昔单抗耐药的非霍奇金氏淋巴瘤,或者所述非霍奇金氏淋巴瘤为具有高度活化的PKC的非霍奇金氏淋巴瘤。Wherein the non-Hodgkin's lymphoma is a B cell non-Hodgkin's lymphoma, or the non-Hodgkin's lymphoma is a non-Hodgkin's lymphoma resistant to rituximab, Alternatively, the non-Hodgkin's lymphoma is a non-Hodgkin's lymphoma with highly activated PKC.
本发明人在研究中发现蛋白激酶C(PKC)在耐受利妥昔单抗的BL细胞中高度上调并活化,而广谱PKC抑制剂(pan-PKC抑制剂)单独使用或与利妥昔单抗联合使用,可有效触发BL细胞的细胞凋亡并显著改善总生存期,特别是在耐受利妥昔单抗的BL细胞中;该结果可进一步扩展至对利妥昔单抗耐药的非霍奇金氏淋巴瘤,或具有高度活化的PKC的非霍奇金氏淋巴瘤。因此,在本发明的上述结论支持单独或与利妥昔单抗组合使用蛋白激酶C以治疗复发/难治性非霍奇金氏淋巴瘤,我们呼吁对其疗效进行进一步的临床试验评估。The present inventors have found in the study that protein kinase C (PKC) is highly upregulated and activated in BL cells that are resistant to rituximab, while broad-spectrum PKC inhibitors (pan-PKC inhibitors) are used alone or in combination with rituximab. The combination of monoclonal antibodies can effectively trigger apoptosis in BL cells and significantly improve overall survival, especially in BL cells that are resistant to rituximab; this result can be further extended to resistance to rituximab Non-Hodgkin's lymphoma, or non-Hodgkin's lymphoma with highly activated PKC. Thus, the above conclusions of the present invention support the use of protein kinase C alone or in combination with rituximab to treat relapsed/refractory non-Hodgkin's lymphoma, and we call for further clinical trial evaluation of its efficacy.
以下将结合附图对本发明的构思、具体结构及产生的技术效果作进一步说明,以充分地了解本发明的目的、特征和效果。The concept, the specific structure and the technical effects of the present invention will be further described in conjunction with the accompanying drawings in order to fully understand the objects, features and effects of the invention.
附图说明DRAWINGS
图1是Ramos细胞和Ramos640细胞中mCRPs表达的Western印迹图;Figure 1 is a Western blot of mCRPs expression in Ramos cells and Ramos 640 cells;
图2是Ramos细胞和Ramos640细胞中mCRPs表达的FACS分析比较结果:与原始细胞相比,Ramos640细胞中CD20表达降低,CD59表达升高,CD55表达略有降低,CD46表达无差异,数据表示为平均值±SD,n=3,NS=无显著性差异,**P<0.01,***P<0.001,****P<0.0001;Figure 2 is a comparison of FACS analysis of mCRPs expression in Ramos cells and Ramos640 cells: compared with the original cells, the expression of CD20 is decreased, the expression of CD59 is increased, the expression of CD55 is slightly decreased, and the expression of CD46 is not different. Value ± SD, n = 3, NS = no significant difference, ** P < 0.01, *** P < 0.001, **** P < 0.0001;
图3是Raji细胞和Raji32细胞中mCRPs表达的Western印迹图;Figure 3 is a Western blot of mCRPs expression in Raji cells and Raji32 cells;
图4是Raji细胞和Raji32细胞中mCRPs表达的FACS分析比较结果:与原始细 胞相比,Raji32细胞中CD20表达降低,CD59表达升高,CD55表达升高,CD46表达降低,数据表示为平均值±SD,n=3,**P<0.01,***P<0.001,****P<0.0001;Figure 4 is a comparison of FACS analysis of mCRPs expression in Raji cells and Raji32 cells: compared with the original cells, the expression of CD20 was decreased, the expression of CD59 was increased, the expression of CD55 was increased, and the expression of CD46 was decreased in Raji32 cells. SD, n=3, **P<0.01, ***P<0.001, ****P<0.0001;
图5是PI3K抑制剂IPI-145(1μM)处理前后四组细胞中Akt磷酸化水平和mCRPs表达水平的Western印迹图:与其原始细胞相比,Ramos640细胞和Raji32细胞中Akt(S473)的磷酸化水平增加,其可被PI3K抑制剂IPI-145处理抑制,导致CD20和CD55的表达降低;Figure 5 is a Western blot of Akt phosphorylation and mCRPs expression in four groups of cells before and after PI3K inhibitor IPI-145 (1 μM): phosphorylation of Akt (S473) in Ramos640 cells and Raji32 cells compared to their original cells An increase in level, which is inhibited by treatment with the PI3K inhibitor IPI-145, resulting in decreased expression of CD20 and CD55;
图6是在Ramos细胞和Ramos640细胞中加入IPI-145(1μM)对利妥昔单抗介导的CDC的影响:单独的IPI-145不能增加原始细胞和耐药细胞的细胞死亡率,在利妥昔单抗中添加IPI-145反而降低了原始细胞和耐药细胞中的利妥昔单抗介导的CDC,数据表示为平均值±SD,n=3,NS=无显著性差异,***P<0.001,并且****P<0.0001;RTX:利妥昔单抗(640μg/mL);NHS:正常人血清(20%v/v);Figure 6 shows the effect of IPI-145 (1 μM) on rituximab-mediated CDC in Ramos cells and Ramos 640 cells: IPI-145 alone does not increase the cell death rate of primordial and drug-resistant cells. Addition of IPI-145 to rituximab reduced rituximab-mediated CDC in primordial and drug-resistant cells, and the data were expressed as mean ± SD, n = 3, NS = no significant difference, * **P<0.001, and ****P<0.0001; RTX: rituximab (640 μg/mL); NHS: normal human serum (20% v/v);
图7是在Raji细胞和Raji32细胞中加入IPI-145(1μM)对利妥昔单抗介导的CDC的影响:单独添加和在利妥昔单抗中添加IPI-145均增加了耐药细胞的细胞死亡率,但未增加原始细胞的细胞死亡率,这种联合用药显著增加了Raji32细胞中利妥昔单抗介导的CDC,数据表示为平均值±SD,n=3,NS=无显著性差异,***P<0.001,并且****P<0.0001;RTX:利妥昔单抗(32μg/mL);NHS:正常人血清(20%v/v);Figure 7 is the effect of IPI-145 (1 μM) on rituximab-mediated CDC in Raji cells and Raji32 cells: addition of IPI-145 alone and addition of rituximab increased drug-resistant cells Cellular mortality, but did not increase cell death in blast cells, this combination significantly increased rituximab-mediated CDC in Raji32 cells, expressed as mean ± SD, n = 3, NS = none Significant difference, ***P<0.001, and ****P<0.0001; RTX: rituximab (32 μg/mL); NHS: normal human serum (20% v/v);
图8是在磷酸化抗体微阵列中上调大于1.5倍的磷酸化蛋白及其磷酸化位点的列图;Figure 8 is a bar graph of up-regulated greater than 1.5-fold phosphorylated protein and its phosphorylation site in a phosphorylated antibody microarray;
图9是图8中上调蛋白的IPA(Ingenuity Pathway Analysis)分析结果,实线表示直接调节,虚线表示间接调节;Figure 9 is an IPA (Ingenuity Pathway Analysis) analysis result of the up-regulated protein of Figure 8, the solid line indicates direct adjustment, and the broken line indicates indirect regulation;
图10是Ramos640细胞与Ramos细胞RNA-Seq数据比较后获得的抗凋亡TNFs:NF-κB:Bcl-2通路的GSEA富集特征图,FDR<0.25被认为是显著的;Figure 10 is a GSEA enrichment profile of anti-apoptotic TNFs:NF-κB:Bcl-2 pathway obtained after comparison of Ramos640 cells with Ramos cell RNA-Seq data, FDR < 0.25 is considered significant;
图11是Ramos640细胞与Ramos细胞RNA-Seq数据比较后获得的p53通路的GSEA富集特征图,FDR<0.25被认为是显著的;Figure 11 is a GSEA enrichment characteristic map of the p53 pathway obtained by comparing Ramos640 cells with Ramos cell RNA-Seq data, and FDR < 0.25 is considered significant;
图12是PKCα/β1/β2/γ/η在四种BL细胞中表达及PKC与其下游的抗凋亡蛋白的磷酸化以及PCK抑制剂米哚妥林(1μM)对它们影响的Western印迹图,该图表明PKC亚型在耐药细胞中上调导致PKC磷酸化升高,并且激活了下游的抗凋亡蛋白,而米哚妥林抑制了PKC及其下游信号;Figure 12 is a Western blot of PKCα/β1/β2/γ/η expression in four BL cells and phosphorylation of PKC and its downstream anti-apoptotic proteins and the effect of PCK inhibitor methotrexate (1 μM) on them. This figure shows that up-regulation of PKC isoforms in drug-resistant cells leads to an increase in PKC phosphorylation and activates downstream anti-apoptotic proteins, while midazolin inhibits PKC and its downstream signaling;
图13是Ramos细胞和Ramos640细胞中pan-PKC抑制剂米哚妥林诱导细胞凋亡的结果图,其中Control:培养基对照,RTX:利妥昔单抗(640μg/mL),Mido:米哚妥林(1μM);该图表明pan-PKC抑制剂有效地诱导了原始和耐药Ramos细胞的细胞凋亡,并且将利妥昔单抗添加到米哚妥林中能显著增强Ramos640细胞中的促凋亡效应,数据表示为平均值±SD,n=3,**P<0.01,***P<0.001和****P<0.0001;Figure 13 is a graph showing the results of apoptosis induced by the pan-PKC inhibitor militalin in Ramos cells and Ramos 640 cells, where Control: medium control, RTX: rituximab (640 μg/mL), Mido: rice bran Tolin (1 μM); this figure demonstrates that pan-PKC inhibitors effectively induce apoptosis in na[iota]ve and resistant Ramos cells, and the addition of rituximab to midazoline significantly enhances Ramos640 cells. Pro-apoptotic effect, data are expressed as mean ± SD, n = 3, ** P < 0.01, *** P < 0.001 and **** P < 0.0001;
图14是Raji细胞和Raji32细胞中pan-PKC抑制剂米哚妥林诱导细胞凋亡的结果图,其中Control:培养基对照,RTX:利妥昔单抗(32μg/mL),Mido:米哚妥林(1 μM);该图表明pan-PKC抑制剂有效地诱导了原始和耐药Raji细胞的细胞凋亡,数据表示为平均值±SD,n=3,**P<0.01,***P<0.001和****P<0.0001;Figure 14 is a graph showing the results of apoptosis induced by the pan-PKC inhibitor methotrexate in Raji cells and Raji32 cells, in which Control: medium control, RTX: rituximab (32 μg/mL), Mido: rice bran Tolin (1 μM); this figure demonstrates that pan-PKC inhibitors effectively induce apoptosis in naive and drug-resistant Raji cells, and the data are expressed as mean ± SD, n = 3, **P < 0.01, ** *P<0.001 and ****P<0.0001;
图15是PKC抑制剂米哚妥林(1μM)对四种细胞中mCRPs表达影响的Western印迹图,该图表明米哚妥林显著降低了原始和耐药细胞中的CD20表达以及耐药细胞中的CD59表达,但未改变原始细胞或耐药细胞中CD55和CD46的表达;Figure 15 is a Western blot of the effect of the PKC inhibitor, mitutoline (1 μM), on the expression of mCRPs in four cells, which shows that midazolin significantly reduces CD20 expression in primary and drug-resistant cells as well as in resistant cells. CD59 expression, but did not alter the expression of CD55 and CD46 in protocell or resistant cells;
图16是PKC抑制剂米哚妥林对Ramos细胞和Ramos640细胞中利妥昔单抗介导的CDC的作用效果,其中Control:培养基对照,RTX:利妥昔单抗(640μg/mL),Mido:米哚妥林(1μM),NHS:正常人血清(20%v/v);该图表明在原始Ramos细胞中,加入米哚妥林未能增强利妥昔单抗介导的CDC,然而在耐药细胞中增加了CDC,数据表示为平均值±SD,n=3,***P<0.001和****P<0.0001;Figure 16 is a graph showing the effect of the PKC inhibitor, midazoline, on rituximab-mediated CDC in Ramos cells and Ramos 640 cells, where Control: medium control, RTX: rituximab (640 μg/mL), Mido: midazolam (1 μM), NHS: normal human serum (20% v/v); this figure indicates that in the original Ramos cells, the addition of militalin did not enhance rituximab-mediated CDC, However, CDC was increased in drug-resistant cells, and the data were expressed as mean ± SD, n = 3, *** P < 0.001 and **** P < 0.0001;
图17是PKC抑制剂米哚妥林对Raji细胞和Raji32细胞中利妥昔单抗介导的CDC的作用效果,其中Control:培养基对照,RTX:利妥昔单抗(32μg/mL),Mido:米哚妥林(1μM),NHS:正常人血清(20%v/v);该图表明在原始Raji细胞中,加入米哚妥林未能增强利妥昔单抗介导的CDC,然而在耐药细胞中增加了CDC,数据表示为平均值±SD,n=3,***P<0.001和****P<0.0001;Figure 17 is a graph showing the effect of the PKC inhibitor, mitolide, on rituximab-mediated CDC in Raji cells and Raji32 cells, in which Control: medium control, RTX: rituximab (32 μg/mL), Mido: midazolam (1 μM), NHS: normal human serum (20% v/v); this figure shows that in the original Raji cells, the addition of methotrex failed to enhance rituximab-mediated CDC, However, CDC was increased in drug-resistant cells, and the data were expressed as mean ± SD, n = 3, *** P < 0.001 and **** P < 0.0001;
图18是在接种Raji32-Luc细胞后第50天肿瘤的生长情况,肿瘤质量由萤火虫荧光素酶活性强度表示,RTX:利妥昔单抗,Mido:米哚妥林;Figure 18 is the growth of tumors on day 50 after inoculation of Raji32-Luc cells, the tumor mass is represented by the intensity of firefly luciferase activity, RTX: rituximab, Mido: methotrexate;
图19是图18中总光子通量的定量结果,数据表示为平均值±SEM(n=7),*P<0.05,**P<0.01和***P<0.001;Figure 19 is a quantitative result of total photon flux in Figure 18, data expressed as mean ± SEM (n = 7), * P < 0.05, ** P < 0.01 and *** P < 0.001;
图20是在接种Raji32-Luc细胞后第70天肿瘤的生长情况,肿瘤质量由萤火虫荧光素酶活性强度表示,“X”代表小鼠死亡,RTX:利妥昔单抗,Mido:米哚妥林;Figure 20 is the growth of tumors on day 70 after inoculation of Raji32-Luc cells, the tumor mass is represented by the intensity of firefly luciferase activity, "X" represents mouse death, RTX: rituximab, Mido: rice bran forest;
图21是图19中总光子通量的定量结果,数据表示为平均值±SEM(n=7),*P<0.05,**P<0.01和***P<0.001;Figure 21 is a quantitative result of total photon flux in Figure 19, data expressed as mean ± SEM (n = 7), * P < 0.05, ** P < 0.01 and *** P < 0.001;
图22是在接种Raji32-Luc细胞后第90天肿瘤的生长情况,肿瘤质量由萤火虫荧光素酶活性强度表示,“X”代表小鼠死亡,RTX:利妥昔单抗,Mido:米哚妥林;Figure 22 shows tumor growth on day 90 after inoculation of Raji32-Luc cells. Tumor quality is expressed by the intensity of firefly luciferase activity, "X" represents death of mice, RTX: rituximab, Mido: rice bran forest;
图23是图21中总光子通量的定量结果,数据表示为平均值±SEM(n=7),*P<0.05,**P<0.01和***P<0.001;Figure 23 is a quantitative result of the total photon flux in Figure 21, the data is expressed as mean ± SEM (n = 7), * P < 0.05, ** P < 0.01 and *** P < 0.001;
图24是接种Raji32-Luc细胞后用药物处理的不同组小鼠的存活曲线,数据表示为平均值±SEM(n=7)。*P<0.05,**P<0.01和***P<0.001。Figure 24 is a survival curve of different groups of mice treated with drugs after inoculation of Raji32-Luc cells, and the data are expressed as mean ± SEM (n = 7). *P<0.05, **P<0.01 and ***P<0.001.
具体实施方式Detailed ways
实施例1对利妥昔单抗介导的CDC耐受的Raji细胞和Ramos细胞的构建Example 1 Construction of Raji Cells and Ramos Cells Against Rituximab-Mediated CDC Tolerance
利妥昔单抗介导的ADCC由个体患者的免疫细胞进行,其对ADCC的耐受性由免疫细胞的内在特征引起,如FcγRIIIa基因多态性。而细胞凋亡在利妥昔单抗抗肿瘤活性中只起着微不足道的作用。因此,我们构建了两种BL细胞系Ramos640和Raji32, 它们分别在利妥昔单抗浓度640和32μg/mL时对利妥昔单抗介导的补体依赖性细胞毒性耐受。其构建方法具体如下:Rituximab-mediated ADCC is carried out by immune cells from individual patients whose tolerance to ADCC is caused by the intrinsic characteristics of immune cells, such as the FcyRIIIa gene polymorphism. Apoptosis plays only a negligible role in the antitumor activity of rituximab. Therefore, we constructed two BL cell lines, Ramos640 and Raji32, which are resistant to rituximab-mediated complement-dependent cytotoxicity at rituximab concentrations of 640 and 32 μg/mL, respectively. The construction method is as follows:
两种BL细胞系Raji和Ramos均购自美国模式培养物集存库(American type culture collection,ATCC)(Manassas,VA),细胞培养在含有10%(体积含量)胎牛血清(GIBCO BRL公司,Grand Island,NY)和1%(体积含量)青霉素/链霉素(Ambion公司,Austin,TX)的RPMI 1640培养基中。Both BL cell lines Raji and Ramos were purchased from American type culture collection (ATCC) (Manassas, VA), and the cells were cultured in 10% (volume) fetal bovine serum (GIBCO BRL, Grand Island, NY) and 1% (by volume) penicillin/streptomycin (Ambion, Austin, TX) in RPMI 1640 medium.
作为补体资源,从10名健康人体中采集获得的正常人血清(NHS),合并后作为提供补体来源的试剂。上述血清等分后在-80℃下储存直至使用。另外,上述血清在65℃水浴中孵育30分钟制备的热灭活人血清(IHS)作为阴性对照。As a complement resource, normal human serum (NHS) was collected from 10 healthy humans and combined to serve as a source of complement. The above serum was aliquoted and stored at -80 ° C until use. In addition, heat-killed human serum (IHS) prepared by incubating the above serum for 30 minutes in a 65 ° C water bath was used as a negative control.
用浓度逐渐升高的利妥昔单抗(Roche公司,Basel,Switzerland)分别处理原始的Raji细胞和Ramos细胞。在20%(体积含量)NHS存在下,浓度分别从4或40μg/mL经依次以2倍浓度的倍数升高至32或640μg/mL,得到的耐药细胞分别被称为Raji32和Ramos640。它们每21天分别用含20%(体积含量)NHS的32μg/mL和640μg/mL的利妥昔单抗处理以维持对利妥昔单抗的耐药性。Raw Raji cells and Ramos cells were separately treated with increasing concentrations of rituximab (Roche, Basel, Switzerland). In the presence of 20% (by volume) of NHS, the concentrations were increased from 4 or 40 μg/mL, respectively, to a multiple of 2 or more to 32 or 640 μg/mL, and the resulting drug-resistant cells were called Raji32 and Ramos 640, respectively. They were treated with 32 μg/mL and 640 μg/mL rituximab containing 20% (by volume) of NHS every 21 days to maintain resistance to rituximab.
实施例2免疫印迹分析和CDC效果测定Example 2 Immunoblot analysis and CDC effect assay
本发明根据标准方法进行了免疫印迹分析。The present invention was subjected to immunoblot analysis according to standard methods.
CDC效果用荧光激活细胞分选(FACS)分析检测碘化丙啶(PI)染色阳性细胞来测定。具体地,用PBS洗涤后,将细胞与荧光素缀合的抗体温育30分钟,然后漂洗并重新悬浮于PBS中。流式细胞分析在Cytomics FC500 MPL流式细胞仪(Beckman Coulter公司,Brea,CA)上进行并用FlowJo软件(Ashland公司,OR)进行分析。我们用MoFlo XDP仪(Beckman Coulter公司,Brea,CA)根据相关的荧光进行细胞分选,使用PE膜联蛋白V凋亡检测试剂盒(BD Pharmingen公司,San Diego,CA)按照其制造商的说明书进行细胞凋亡分析。The CDC effect was determined by fluorescence-activated cell sorting (FACS) analysis to detect propidium iodide (PI) staining positive cells. Specifically, after washing with PBS, the cells were incubated with fluorescein-conjugated antibody for 30 minutes, then rinsed and resuspended in PBS. Flow cytometric analysis was performed on a Cytomics FC500 MPL flow cytometer (Beckman Coulter, Brea, CA) and analyzed using FlowJo software (Ashland, OR). We used the MoFlo XDP instrument (Beckman Coulter, Brea, CA) for cell sorting based on the relevant fluorescence, using the PE annexin V apoptosis assay kit (BD Pharmingen, Inc., San Diego, CA) according to the manufacturer's instructions. Perform apoptosis analysis.
实施例3 CD20下调和CD59上调导致BL细胞对利妥昔单抗介导的CDC的耐受Example 3 Downregulation of CD20 and Upregulation of CD59 Lead to BL Cells to Rituximab-Mediated CDC Tolerance
使用免疫印迹分析(图1和图3)和FACS(图2和图4),我们发现与其原始细胞相比,在两种耐药细胞中CD20的表达都降低,而CD59的表达都升高(图1-图4)。然而,另外两种膜补体调节蛋白(mCRP)CD55和CD46在两种耐药细胞中的表达并不一致。CD55在Ramos640中的表达降低,而在Raji32中的表达升高;而CD46在Ramos640中的表达没有改变,但在Raji32中的表达下降(图1-图4)。这些结果与先前的报道一致,即降低的CD20表达和升高的CD59表达导致BL细胞对利妥昔单抗介导的CDC的耐受性。但是,由于前人在提高CD20表达和抑制CD59功能的研究中均未获得很好的结果,因此,本发明尝试了其他的治疗策略。Using immunoblot analysis (Figures 1 and 3) and FACS (Figures 2 and 4), we found that CD20 expression was reduced in both drug-resistant cells compared to the original cells, while CD59 expression was elevated ( Figure 1 - Figure 4). However, the expression of two other membrane complement regulatory proteins (mCRP), CD55 and CD46, was inconsistent in both resistant cells. The expression of CD55 was reduced in Ramos 640, while the expression in Raji32 was elevated; whereas the expression of CD46 in Ramos 640 was unchanged, but the expression was decreased in Raji32 (Figs. 1 - 4). These results are consistent with previous reports that reduced CD20 expression and elevated CD59 expression result in BL cells being resistant to rituximab-mediated CDC. However, since the predecessors did not obtain good results in the study of increasing CD20 expression and inhibiting CD59 function, the present invention has attempted other therapeutic strategies.
实施例4 PI3K/Akt通路的强烈富集,但对其的抑制未能逆转耐药性Example 4 Strong enrichment of the PI3K/Akt pathway, but its inhibition failed to reverse drug resistance
本发明通过蛋白质信号通路分析对耐药机理做了进一步的研究,本实施例使用磷酸化抗体微阵列来识别在Ramos640细胞中被激活的功能性蛋白质和信号传导通路。The present invention further studies the mechanism of drug resistance by protein signaling pathway analysis. This example uses a phosphorylated antibody microarray to identify functional proteins and signaling pathways that are activated in Ramos 640 cells.
我们用磷酸化抗体芯片(FullMoon Microsystems,Catalog#CSP100,Sunnyvale,CA)分析了蛋白的磷酸化,其中包含针对131个蛋白质磷酸化位点的269个抗体,并根据其建立的方法由Wayen生物科技公司(上海,中国)进行分析。We analyzed phosphorylation of the protein with a phosphorylated antibody chip (FullMoon Microsystems, Catalog #CSP100, Sunnyvale, CA) containing 269 antibodies against 131 protein phosphorylation sites and according to its established method by Wayen Biotech The company (Shanghai, China) conducted an analysis.
荧光分析显示与Ramos细胞相比,Ramos640中35个蛋白的磷酸化水平上调(31/35)或下调(4/35)超过1.4倍。这些蛋白的KEGG信号通路分析证明PI3K/AKT信号传导通路含有最多的磷酸化蛋白位点,为13个,而细胞凋亡通路显示出最大的富集倍数为47.76(如表1所示)。Fluorescence analysis showed that the phosphorylation levels of 35 proteins in Ramos640 were up-regulated (31/35) or down-regulated (4/35) by more than 1.4-fold compared to Ramos cells. Analysis of the KEGG signaling pathway of these proteins demonstrated that the PI3K/AKT signaling pathway contained the most phosphorylated protein sites in 13 and the apoptotic pathway showed the largest enrichment factor of 47.76 (as shown in Table 1).
表1用磷酸化抗体微阵列测定的Ramos640细胞相对于Ramos细胞的富集通路Table 1 Enrichment pathways of Ramos 640 cells relative to Ramos cells as determined by phosphorylated antibody microarrays
Figure PCTCN2019082102-appb-000002
Figure PCTCN2019082102-appb-000002
Figure PCTCN2019082102-appb-000003
Figure PCTCN2019082102-appb-000003
接下来,使用PI3K抑制剂IPI-145来鉴定对PI3K的抑制是否可以提高耐药BL细胞对利妥昔单抗治疗的敏感性。PI3K抑制剂IPI-145购自Selleck Chemicals公司(Houston,TX)。Next, the PI3K inhibitor IPI-145 was used to identify whether inhibition of PI3K can increase the sensitivity of resistant BL cells to rituximab treatment. The PI3K inhibitor IPI-145 was purchased from Selleck Chemicals (Houston, TX).
具体地,将原始细胞和耐药细胞以1×10 4个细胞/100μL/孔的密度接种到96孔板中,并分别分为四组,第一组用培养基培养48小时作为对照,第二组用含IPI-145浓度为1μM的培养基处理48小时,第三组用含20%(体积含量)NHS且利妥昔单抗浓度为640μg/mL或32μg/mL的培养基处理48小时,第四组用含20%(体积含量)NHS且利妥昔单抗浓度为640μg/mL或32μg/mL、IPI-145浓度为1μM的培养基处理48小时。 Specifically, the original cells and the drug-resistant cells were seeded into 96-well plates at a density of 1×10 4 cells/100 μL/well, and were divided into four groups, and the first group was cultured for 48 hours as a control, The two groups were treated with a medium containing IPI-145 at a concentration of 1 μM for 48 hours, and the third group was treated with a medium containing 20% (by volume) of NHS and rituximab at a concentration of 640 μg/mL or 32 μg/mL for 48 hours. The fourth group was treated with a medium containing 20% (by volume) of NHS and having a rituximab concentration of 640 μg/mL or 32 μg/mL and an IPI-145 concentration of 1 μM for 48 hours.
使用CytoTox-Glo TM细胞毒性测定试剂盒(Promega公司,Madison,WI)根据其技术公报所述的步骤进行细胞毒性分析,并根据下式计算细胞毒性: Using CytoTox-Glo TM cytotoxicity assay kit (Promega Corporation, Madison, WI) cytotoxicity assay performed according to its technical bulletin steps and cytotoxicity was calculated according to the following formula:
细胞毒性(%)=死细胞发光量/总发光量×100%Cytotoxicity (%) = dead cell luminescence / total luminescence × 100%
除非另有说明,本发明数据以平均值±标准差表示。两组之间的显著差异使用未配对数据的双尾学生t检验来确定。在所有分析中,p<0.05被认为是统计学显著的。The data of the present invention are expressed as mean ± standard deviation unless otherwise stated. Significant differences between the two groups were determined using a two-tailed Student's t-test of unpaired data. In all analyses, p < 0.05 was considered to be statistically significant.
实验结果首先证实了PI3K/Akt信号通路在Ramos640和Raji32细胞中与其原始细胞相比被激活,并且IPI-145有效地削弱了Akt的磷酸化(S473)(图5)。然而,在原始细胞和耐药细胞中,IPI-145的处理明显抑制了CD20的表达,而其它三种mCRP包括CD59、CD55和CD46的表达几乎没有改变(图5)。这些结果随后导致IPI-145对原始和耐药细胞中利妥昔单抗介导的CDC无效甚至有略微相反的作用(Raji32细胞除外)(图6和图7)。在Raji32细胞中,单独或联合利妥昔单抗的IPI-145分别与对照或单独的利妥昔单抗相比略微增加了细胞的死亡率(图7)。这些结果表明,PI3K/Akt可能不是促进利妥昔单抗治疗的有价值的药物靶标。The experimental results first confirmed that the PI3K/Akt signaling pathway was activated in Ramos640 and Raji32 cells compared to its original cells, and IPI-145 effectively attenuated Akt phosphorylation (S473) (Fig. 5). However, in both primary and resistant cells, treatment with IPI-145 significantly inhibited expression of CD20, while expression of the other three mCRPs including CD59, CD55 and CD46 showed little change (Fig. 5). These results subsequently led to IPI-145 having a slightly opposite effect on rituximab-mediated CDC in the original and drug-resistant cells (except for Raji32 cells) (Figures 6 and 7). In Raji32 cells, IPI-145 alone or in combination with rituximab slightly increased cell mortality compared to control or rituximab alone (Figure 7). These results suggest that PI3K/Akt may not be a valuable drug target for the promotion of rituximab treatment.
实施例5 PKC介导的细胞凋亡通路在耐药性细胞中高度活化Example 5 PKC-mediated apoptotic pathway is highly activated in drug-resistant cells
考虑到利妥昔单抗诱导B-NHL(B细胞型非霍奇金氏淋巴瘤)细胞凋亡的效应可忽略不计,因此向利妥昔单抗添加可诱导细胞凋亡的试剂可能是一种理想的治疗策略。Considering that the effect of rituximab in inducing apoptosis in B-NHL (B-cell non-Hodgkin's lymphoma) cells is negligible, the addition of an agent that induces apoptosis to rituximab may be a An ideal treatment strategy.
尽管细胞凋亡通路在磷酸化抗体微阵列中被鉴定为富集倍数最高的通路,但该通路中仅包含6种蛋白(表1),这阻碍了通路调节剂的选择。因此,我们通过将磷酸化抗体微阵列中上调和下调的磷酸化蛋白位点的倍数变化提高到1.5倍来缩小查找范围,结果显示总共16个蛋白质位点上调了1.5倍(图8)。Although the apoptotic pathway was identified as the most abundant pathway in the phosphorylated antibody microarray, it contained only six proteins (Table 1), which hindered the selection of pathway modulators. Therefore, we narrowed the search by increasing the fold change of the up- and down-regulated phosphorylated protein sites in the phosphorylated antibody microarray to 1.5-fold, and the results showed that a total of 16 protein sites were up-regulated by 1.5-fold (Fig. 8).
通过DAVID生物信息数据库筛选具有倍数变化>1.5的差异蛋白以进行功能注释。有趣的是,通过IPA通路分析软件(Ingenuity Pathway Analysis)(QIAGEN公司,Duesseldorf,Germany)确定了所有这些上调的蛋白/位点都参与了细胞凋亡(图9);更重要的是,IPA还揭示了PKC(蛋白激酶C)信号调节所有这些上调蛋白,尽管PKC磷酸化的检测不包括在磷酸化抗体微阵列集中,通过IPA分析获得的PKC亚基与差异蛋白之间的相互作用网络如图9所示。Functional annotations with a fold change >1.5 were screened by the DAVID Bioinformatics database for functional annotation. Interestingly, all of these up-regulated proteins/sites were involved in apoptosis through the IPA pathway analysis software (QIAGEN, Duesseldorf, Germany); more importantly, IPA also It is revealed that PKC (protein kinase C) signaling regulates all of these up-regulated proteins, although the detection of PKC phosphorylation is not included in the phosphorylated antibody microarray set, the interaction network between the PKC subunit and the differential protein obtained by IPA analysis is shown in Fig. 9 is shown.
接着,我们用RNA-seq对原始和耐药Ramos细胞进行了RNA测序并进一步做了基因富集分析。Next, we performed RNA sequencing of the original and resistant Ramos cells with RNA-seq and further performed gene enrichment analysis.
RNA测序方法:RNA sequencing method:
用Trizol试剂(Invitrogen公司,Grand Island,NY)分别从Ramos和Ramos640细胞中提取总RNA。将各组中来自3个不同代细胞的总RNA分别合并在一起。用生物分析仪2200(Agilent Technologies公司,Santa,Clara,CA)鉴定RNA质量并保存在-80℃中。RNA完整数(RIN)>8.0的RNA对于cDNA文库构建是可接受的。由上海Novelbio有限公司进行RNA-seq的测序,使用Ion Tatal RNA-seq试剂盒v2.0(Life Technologies公司,Gaithersburg,MD)为每个合并的RNA样品构建cDNA文库,然后进行质子测序过程。在读取映射之前,通过去除接头序列从原始读取结果中获得干净读取结果,然后使用MapSplice程序(v2.1.6)与人类基因组(版本:GRCh37NCBI)比对。我们应用DEseq算法筛选差异表达基因,显著性分析和错误发现率(FDR)分析的标准如下:(1)倍数变化>1.5或<0.667;(2)FDR<0.05。Total RNA was extracted from Ramos and Ramos 640 cells using Trizol reagent (Invitrogen, Grand Island, NY). Total RNA from 3 different generation cells in each group was pooled separately. RNA quality was identified using Bioanalyzer 2200 (Agilent Technologies, Santa, Clara, CA) and stored at -80 °C. RNA with an RNA complete number (RIN) > 8.0 is acceptable for cDNA library construction. RNA-seq was sequenced by Shanghai Novelbio Co., Ltd., and a cDNA library was constructed for each pooled RNA sample using Ion Tatal RNA-seq kit v2.0 (Life Technologies, Gaithersburg, MD), followed by a proton sequencing process. A clean reading was obtained from the original reading by removing the linker sequence before reading the map, and then aligned with the human genome (version: GRCh37NCBI) using the MapSplice program (v2.1.6). We applied the DEseq algorithm to screen differentially expressed genes. The criteria for significance analysis and false discovery rate (FDR) analysis were as follows: (1) fold change >1.5 or <0.667; (2) FDR<0.05.
基因集富集分析方法:Gene set enrichment analysis method:
本发明使用基因集富集分析GSEA软件(麻省理工学院Broad研究所)来鉴定RNA-seq中发现的差异表达基因的功能。该软件的预定版本被用于鉴定显著富集的通路,并且FDR<0.25的富集通路被认为是显著的。本发明中使用的抗凋亡TNFs:NF-κB:Bcl-2通路基因集由来自PathCards通路统一数据库(4.6.0.37版,Weizmann科学院)中的“凋亡和存活抗凋亡TNFs:NF-κB:Bcl-2通路SuperPath”的42个基因组成。本发明使用的p53通路基因集由来自PathCards通路统一数据库中的“p53通路(RnD)SuperPath”的132个基因组成。The present invention uses gene set enrichment analysis GSEA software (the Massachusetts Institute of Technology Broad Institute) to identify the function of differentially expressed genes found in RNA-seq. A predetermined version of the software was used to identify significantly enriched pathways, and an enrichment pathway with FDR < 0.25 was considered significant. Anti-apoptotic TNFs used in the present invention: NF-κB: Bcl-2 pathway gene set is derived from the PathCards Pathway Uniform Database (version 4.0.6.37, Weizmann Academy of Sciences) "Apoptosis and Survival Anti-apoptotic TNFs: NF-κB : Bcl-2 pathway SuperPath" consists of 42 genes. The p53 pathway gene set used in the present invention consists of 132 genes from the "p53 pathway (RnD) SuperPath" in the unified database of the PathCards pathway.
通过RNA-seq检测原始和耐药Ramos细胞以及GSEA分析,我们进一步发现,与Ramos细胞相比,Ramos640细胞中TNFs:NF-κB:Bcl-2通路的基因显著升高,而p53通路的基因则显著降低(图10和图11)。它们还表明了抗凋亡基因上调而促凋亡基因下调。因此,这些数据表明PKC磷酸化多个下游蛋白,导致Ramos640细胞中产生了抗细胞凋亡作用。By detecting the original and resistant Ramos cells by RNA-seq and GSEA analysis, we further found that the TNFs:NF-κB:Bcl-2 pathway gene was significantly increased in Ramos640 cells compared to Ramos cells, while the p53 pathway gene was significantly increased. Significantly reduced (Figures 10 and 11). They also indicate up-regulation of anti-apoptotic genes and down-regulation of pro-apoptotic genes. Thus, these data indicate that PKC phosphorylates multiple downstream proteins, resulting in an anti-apoptotic effect in Ramos 640 cells.
实施例6 pan-PKC抑制剂米哚妥林(Midostaurin)显著促进细胞凋亡Example 6 pan-PKC inhibitor Midostaurin significantly promotes apoptosis
米哚妥林是一种多激酶抑制剂,最初旨在抑制PKC,目前已被批准用于伴FLT3(Fms样酪氨酸激酶3)突变的急性骨髓性白血病(AML)和晚期系统性肥大细胞增多症(SM)。本发明所用PKC抑制剂米哚妥林购自Selleck Chemicals公司(Houston,TX)。Mitalin is a multi-kinase inhibitor originally designed to inhibit PKC and is currently approved for acute myeloid leukemia (AML) and advanced systemic mast cells with FLT3 (Fms-like tyrosine kinase 3) mutations. Hyperplasia (SM). The PKC inhibitor mitutolin used in the present invention was purchased from Selleck Chemicals (Houston, TX).
本实施例首先比较了原始BL细胞和耐药BL细胞中5种PKC亚型的表达水平,发现PKCα/β2/γ/η在Ramos640细胞中,PKCα/β1/β2/γ/η在Raji32细胞中,与其原始细胞相比过表达,更重要的是,两种耐药细胞中所有PKC亚型的磷酸化水平均显著升高(图12)。此外,我们检测了米哚妥林对调节PKC磷酸化的作用。我们观察到,它在耐药BL细胞和原始Ramos细胞中强烈抑制PKC的磷酸化水平,但在原始Raji细胞中没有,并且它降低了Ramos细胞中PKCβ2/η、Ramos640细胞中PKCα/η以及Raji32细胞中PKCβ1/η的表达水平(图12)。In this example, we first compared the expression levels of five PKC isoforms in naive BL cells and resistant BL cells, and found that PKCα/β2/γ/η in Ramos640 cells, PKCα/β1/β2/γ/η in Raji32 cells. , overexpression compared to its original cells, and more importantly, phosphorylation levels of all PKC isoforms in both drug-resistant cells were significantly elevated (Figure 12). In addition, we examined the effect of midazolam on the regulation of PKC phosphorylation. We observed that it strongly inhibited the phosphorylation of PKC in resistant BL cells and naive Ramos cells, but not in the original Raji cells, and it reduced PKCβ2/η in Ramos cells, PKCα/η in Ramos640 cells, and Raji32. The expression level of PKCβ1/η in the cells (Fig. 12).
接下来,我们检测了几种PKC下游信号分子的磷酸化水平,包括Bad、Bcl-2和NF-κB亚基p65。我们首先发现它们的磷酸化水平显著升高(图12)。此外在两种耐药细胞中,Bad的表达水平显著降低,p65的表达水平显着升高(图12)。所有上述变化都可能对细胞凋亡抑制有很大贡献,从而导致Ramos640细胞和Raji32细胞对利妥昔单抗的耐药性的发展。另外,在所有原始细胞和耐药细胞中,米哚妥林强烈抑制了Bad、Bcl-2和p65的磷酸化水平(图12)。进一步,米哚妥林分别明显地增加或减少了所有四种原始细胞或耐药细胞中的Bad或p65的表达,并且仅在原始的和耐药的Raji细胞中降低了Bcl-2的表达(图12)。因此,米哚妥林抑制PKC及其下游信号,这可能在不同程度上导致Ramos和Raji细胞中的促凋亡作用。其中Raji细胞似乎比Ramos细胞更能抵抗米哚妥林介导的PKC磷酸化,因为米哚妥林未能抑制PKC磷酸化。Next, we examined the phosphorylation levels of several downstream PKC signaling molecules, including the Bad, Bcl-2, and NF-κB subunits p65. We first found that their phosphorylation levels were significantly elevated (Figure 12). In addition, in both drug-resistant cells, the expression level of Bad was significantly decreased, and the expression level of p65 was significantly increased (Fig. 12). All of the above changes may contribute significantly to inhibition of apoptosis, leading to the development of resistance to rituximab in Ramos640 cells and Raji32 cells. In addition, midazolin strongly inhibited the phosphorylation levels of Bad, Bcl-2 and p65 in all of the original cells and drug-resistant cells (Fig. 12). Further, midazolin significantly increased or decreased the expression of Bad or p65 in all four primordial or drug-resistant cells, and reduced Bcl-2 expression only in naive and resistant Raji cells ( Figure 12). Thus, militalin inhibits PKC and its downstream signaling, which may lead to pro-apoptotic effects in Ramos and Raji cells to varying degrees. Among them, Raji cells appear to be more resistant to midazolin-mediated PKC phosphorylation than Ramos cells because midazolin failed to inhibit PKC phosphorylation.
此外,我们检测了单独使用pan-PKC抑制剂米哚妥林(1μM)或与利妥昔单抗(针对Raji细胞的用药浓度为32μg/mL,针对Ramos细胞的用药浓度为640μg/mL)联用对原始和耐药BL细胞的促凋亡作用。In addition, we tested the use of the pan-PKC inhibitor militalin (1 μM) alone or in combination with rituximab (32 μg/mL for Raji cells and 640 μg/mL for Ramos cells). Pro-apoptotic effects on naive and resistant BL cells.
具体地,将原始细胞和耐药细胞以1×10 4个细胞/100μL/孔的密度接种到96孔板中,并分别分为四组,第一组用培养基培养48小时作为对照,第二组用利妥昔单抗浓度为640μg/mL或32μg/mL的培养基处理48小时,第三组用含米哚妥林浓 度为1μM的培养基处理48小时,第四组用含利妥昔单抗浓度为640μg/mL或32μg/mL、米哚妥林浓度为1μM的培养基处理48小时。 Specifically, the original cells and the drug-resistant cells were seeded into 96-well plates at a density of 1×10 4 cells/100 μL/well, and were divided into four groups, and the first group was cultured for 48 hours as a control, The two groups were treated with rituximab at a concentration of 640 μg/mL or 32 μg/mL for 48 hours, and the third group was treated with a medium containing 1 μM of miltazone for 48 hours. The medium was treated with a concentration of 640 μg/mL or 32 μg/mL and a midazorine concentration of 1 μM for 48 hours.
使用CytoTox-Glo TM细胞毒性测定试剂盒(Promega公司,Madison,WI)根据其技术公报所述的步骤进行细胞毒性分析,并根据下式计算细胞毒性: Using CytoTox-Glo TM cytotoxicity assay kit (Promega Corporation, Madison, WI) cytotoxicity assay performed according to its technical bulletin steps and cytotoxicity was calculated according to the following formula:
细胞毒性(%)=死细胞发光量/总发光量×100%Cytotoxicity (%) = dead cell luminescence / total luminescence × 100%
除非另有说明,本发明数据以平均值±标准差表示。两组之间的显著差异使用未配对数据的双尾学生t检验来确定。在所有分析中,p<0.05被认为是统计学显著的。The data of the present invention are expressed as mean ± standard deviation unless otherwise stated. Significant differences between the two groups were determined using a two-tailed Student's t-test of unpaired data. In all analyses, p < 0.05 was considered to be statistically significant.
正如我们所预料的那样,利妥昔单抗单独治疗在全部四种原始或耐药BL细胞中均未能诱导的细胞凋亡(图13和图14)。然而,与培养基对照相比,单独使用米哚妥林显著诱导了Ramos细胞(从4.7%至63.3%)、Ramos640细胞(从10.8%至50.2%)和Raji32细胞(从6.4%至25.4%)的细胞凋亡,而Raji细胞只是略微上升(从6.6%到10.9%),尽管p值达到了统计学显著性(p=0.0038)(图13和图14)。这些结果也从功能上确定了,一般来说,Raji细胞比Ramos细胞更能抵抗米哚妥林诱导的细胞凋亡。此外,米哚妥林与利妥昔单抗联用仅在原始细胞而非耐药Ramos细胞和Raji细胞中呈现轻微的进一步促凋亡效应(图13和图14),进一步证实了利妥昔单抗的促细胞凋亡效应(如果存在)是可忽略的。As we expected, rituximab alone treated apoptosis that was not induced in all four naive or resistant BL cells (Figures 13 and 14). However, compared with the medium control, the use of militalin alone significantly induced Ramos cells (from 4.7% to 63.3%), Ramos 640 cells (from 10.8% to 50.2%), and Raji32 cells (from 6.4% to 25.4%). Apoptosis, while Raji cells only increased slightly (from 6.6% to 10.9%), although the p value reached statistical significance (p = 0.0038) (Figure 13 and Figure 14). These results are also functionally determined. In general, Raji cells are more resistant to ampicillin-induced apoptosis than Ramos cells. In addition, the combination of midazolam and rituximab showed a slight further pro-apoptotic effect only in primordial cells but not in resistant Ramos cells and Raji cells (Figures 13 and 14), further confirming rituximab The pro-apoptotic effect of the monoclonal antibody, if any, is negligible.
实施例7米哚妥林加强了耐药BL细胞对利妥昔单抗介导的CDC的易感性Example 7 Mitalantin enhances susceptibility of resistant BL cells to rituximab-mediated CDC
在本实施例中我们使用pan-PKC抑制剂米哚妥林来鉴定对PKC的抑制是否可以提高耐药BL细胞对利妥昔单抗治疗的敏感性。In this example we used the pan-PKC inhibitor, midazolin, to identify whether inhibition of PKC can increase the sensitivity of resistant BL cells to rituximab treatment.
具体地,将原始细胞和耐药细胞以1×10 4个细胞/100μL/孔的密度接种到96孔板中,并分别分为四组,第一组用培养基培养48小时作为对照,第二组用含20%(体积含量)NHS且利妥昔单抗浓度为640μg/mL或32μg/mL的培养基处理48小时,第三组用含米哚妥林浓度为1μM的培养基处理48小时,第四组用含20%(体积含量)NHS且利妥昔单抗浓度为640μg/mL或32μg/mL、米哚妥林浓度为1μM的培养基处理48小时。 Specifically, the original cells and the drug-resistant cells were seeded into 96-well plates at a density of 1×10 4 cells/100 μL/well, and were divided into four groups, and the first group was cultured for 48 hours as a control, The two groups were treated with a medium containing 20% (by volume) of NHS and a concentration of rituximab of 640 μg/mL or 32 μg/mL for 48 hours, and the third group was treated with a medium containing 1 μM of metoprolin. For the hour, the fourth group was treated with a medium containing 20% (by volume) of NHS and having a rituximab concentration of 640 μg/mL or 32 μg/mL and a myristicin concentration of 1 μM for 48 hours.
使用CytoTox-Glo TM细胞毒性测定试剂盒(Promega公司,Madison,WI)根据其技术公报所述的步骤进行细胞毒性分析,并根据下式计算细胞毒性: Using CytoTox-Glo TM cytotoxicity assay kit (Promega Corporation, Madison, WI) cytotoxicity assay performed according to its technical bulletin steps and cytotoxicity was calculated according to the following formula:
细胞毒性(%)=死细胞发光量/总发光量×100%Cytotoxicity (%) = dead cell luminescence / total luminescence × 100%
除非另有说明,本发明数据以平均值±标准差表示。两组之间的显著差异使用未配对数据的双尾学生t检验来确定。在所有分析中,p<0.05被认为是统计学显著的。The data of the present invention are expressed as mean ± standard deviation unless otherwise stated. Significant differences between the two groups were determined using a two-tailed Student's t-test of unpaired data. In all analyses, p < 0.05 was considered to be statistically significant.
除了CD20之外,利妥昔单抗介导的CDC可以通过mCRP的表达来调节,例如CD46、CD55、特别是CD59。我们观察到,米哚妥林(1μM)显著降低了所有四种 BL细胞中CD20的表达水平,而CD59的表达水平的降低则主要在耐药细胞中;而对CD55和CD46的表达没有影响(图15)。这些结果表明,由于原始BL细胞中仅有CD20的表达降低,所以米哚妥林可能阻碍了利妥昔单抗介导的CDC,而由于耐药BL细胞中CD20和CD59两者的表达都降低了,因此其对耐药BL细胞的作用需要进一步检测。此外,这些结果显示了IPI-145和米哚妥林对调节CD20和CD59表达的影响之间的差异,其中IPI-145仅在耐药BL细胞中减少CD20的表达但不减少CD59的表达(图5)。In addition to CD20, rituximab-mediated CDC can be regulated by expression of mCRP, such as CD46, CD55, and especially CD59. We observed that midazolam (1 μM) significantly reduced the expression of CD20 in all four BL cells, whereas the decrease in CD59 expression was predominant in drug-resistant cells; it had no effect on the expression of CD55 and CD46 ( Figure 15). These results indicate that midazolin may block rituximab-mediated CDC due to decreased expression of CD20 in naive BL cells, whereas expression of both CD20 and CD59 is reduced in resistant BL cells. Therefore, its effect on resistant BL cells requires further testing. In addition, these results show the difference between the effects of IPI-145 and midazolam on the regulation of CD20 and CD59 expression, in which IPI-145 only reduces CD20 expression in resistant BL cells but does not reduce CD59 expression (Fig. 5).
利妥昔单抗(针对Raji细胞的用药浓度为32μg/mL,针对Ramos细胞的用药浓度为640μg/mL)加NHS(20%,v/v)有效诱导了原始Ramos细胞和Raji细胞中的CDC,然而,添加米哚妥林(1μM)未能增强利妥昔单抗介导的CDC的易感性,尽管它可促进细胞凋亡(图16和图17)。这可能是由于利妥昔单抗单独治疗导致的CD20表达降低和已经很高的细胞死亡率。相反,利妥昔单抗的添加显著增强了原始BL细胞中米哚妥林的细胞毒性作用(图16和图17),表明在原始BL细胞中利妥昔单抗介导的CDC的作用大于的米哚妥林诱导的细胞凋亡。有趣的是,我们发现利妥昔单抗和米哚妥林联用的细胞毒性作用在耐药的Ramos640细胞和Raji32细胞中具有协同效果。如图16所示,利妥昔单抗和米哚妥林分别诱导了17.1%和54.3%的细胞死亡,而它们的组合诱导了72.8%的细胞死亡。类似地,如图17所示,利妥昔单抗和米哚妥林分别诱导28.0%和25.8%的细胞死亡,而它们的组合诱导62.9%的细胞死亡。该结果可能源于利妥昔单抗和米哚妥林独特的抗肿瘤机制。Rituximab (a concentration of 32 μg/mL for Raji cells and 640 μg/mL for Ramos cells) plus NHS (20%, v/v) effectively induced CDC in naive Ramos cells and Raji cells. However, the addition of militalin (1 μM) failed to enhance the susceptibility of rituximab-mediated CDC, although it promoted apoptosis (Figures 16 and 17). This may be due to a decrease in CD20 expression and a already high cell death rate due to rituximab alone. In contrast, the addition of rituximab significantly enhanced the cytotoxic effect of militalin in the original BL cells (Figure 16 and Figure 17), indicating that the effect of rituximab-mediated CDC is greater in primary BL cells. Mitotalin induces apoptosis. Interestingly, we found that the cytotoxic effects of rituximab in combination with midazolam have synergistic effects in resistant Ramos640 cells and Raji32 cells. As shown in Figure 16, rituximab and midazoline induced 17.1% and 54.3% cell death, respectively, and their combination induced 72.8% cell death. Similarly, as shown in Figure 17, rituximab and midazoline induced 28.0% and 25.8% cell death, respectively, and their combination induced 62.9% cell death. This result may be due to the unique anti-tumor mechanism of rituximab and midazolam.
实施例8利妥昔单抗与米哚妥林联用显著抑制了耐药细胞的肿瘤生长Example 8 Rituximab in combination with Mentrolidine significantly inhibited tumor growth in drug-resistant cells
鉴于米哚妥林的明显促细胞凋亡效应可能有助于完善利妥昔单抗的抗肿瘤活性,我们在接种了由表达萤光素酶的质粒转染的更具耐药性的Raji32细胞的免疫缺陷小鼠中,测试了利妥昔单抗与米哚妥林联用的疗效。Given that the apparent pro-apoptotic effects of midazolin may contribute to the improvement of the anti-tumor activity of rituximab, we inoculated a more resistant Raji32 cell transfected with a plasmid expressing luciferase. The efficacy of rituximab in combination with midazolam was tested in immunodeficient mice.
质粒构建和慢病毒转导Plasmid construction and lentiviral transduction
萤火虫萤光素酶基因的CDS(coding sequence,蛋白质编码区)通过PCR扩增从pGL3-Basic质粒获得,并通过EcoRI和BamHI内切酶位点插入到pCDH cDNA克隆和表达载体中。萤火虫萤光素酶CDS扩增的引物如下所示:The CDS (coding sequence) of the firefly luciferase gene was obtained from the pGL3-Basic plasmid by PCR amplification, and inserted into the pCDH cDNA clone and expression vector by EcoRI and BamHI endonuclease sites. The primers for firefly luciferase CDS amplification are as follows:
正向引物5'-ATGGAAGACGCCAAAAACATAAAG-3'Forward primer 5'-ATGGAAGACGCCAAAAACATAAAG-3'
反向引物5'-TTACACGGCGATCTTTCCGCCCTT-3'Reverse primer 5'-TTACACGGCGATCTTTCCGCCCTT-3'
将pCDH质粒与pMD.2G和psPAX2质粒在293FT细胞中共转染以产生萤火虫荧光素酶过表达慢病毒。随后将该慢病毒加入到Raji32细胞培养基中孵育48小时。本发明中用慢病毒转染的所有细胞均用GFP及MoFlo XDP仪(Beckman Coulter公司,Brea,CA)进行分类,称为Raji32-Luc细胞。The pCDH plasmid was co-transfected with the pMD.2G and psPAX2 plasmids in 293FT cells to generate firefly luciferase overexpressing lentivirus. The lentivirus was then added to Raji32 cell culture medium for 48 hours. All cells transfected with lentivirus in the present invention were classified using GFP and MoFlo XDP instruments (Beckman Coulter, Brea, CA) and referred to as Raji32-Luc cells.
异种移植模型Xenograft model
8周龄雌性SCID小鼠购自SLAC(Shanghai Laboratory Animal Center,上海实验动物中心)。将Raji32-Luc细胞重悬于PBS中,然后给每只小鼠腹膜内注射1.5×10 7个细胞。基于给药药物的不同,即生理盐水、利妥昔单抗、米哚妥林和利妥昔单抗加米哚妥林,将小鼠分成4组(每组7只小鼠)。肿瘤接种后第8、12和16天,腹腔注射118.4mg/kg利妥昔单抗,以及肿瘤接种后第8、9、10、11、12、13、14、15、16、17、18、19、20和21天,用米哚妥林20mg/kg灌胃。盐水给药的注射量时与利妥昔单抗相同。肿瘤接种后的第50、70和90天通过生物发光监测肿瘤的生长,将D-荧光素(Promega公司,Madison,WI)腹膜注射到小鼠(150mg/kg)体内,十分钟后,腹膜注射戊巴比妥(50mg/kg)麻醉小鼠,然后使用In-Vivo MS FX PRO系统(Bruker公司,Billerica,MA)进行生物发光检测。用30秒的曝光时间捕获发光图像,并通过Bruker MI软件测量肿瘤的信号强度。记录每只小鼠的存活时间直到120天。所有动物实验均严格按照复旦大学上海医学院动物伦理委员会批准的实验方案进行。 Eight-week-old female SCID mice were purchased from SLAC (Shanghai Laboratory Animal Center, Shanghai Experimental Animal Center). Raji32-Luc cells were resuspended in PBS, and then each mouse was intraperitoneally injected with 1.5 x 10 7 cells. Mice were divided into 4 groups (7 mice per group) based on the difference in drug administration, namely saline, rituximab, midazoline and rituximab plus militalin. On the 8th, 12th and 16th day after tumor inoculation, 118.4 mg/kg rituximab was intraperitoneally injected, and 8, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18 after tumor inoculation. On days 19, 20 and 21, the rats were intragastrically administered with miltazone 20 mg/kg. The amount of saline administered was the same as that of rituximab. Tumor growth was monitored by bioluminescence on days 50, 70, and 90 after tumor inoculation, and D-luciferin (Promega, Madison, WI) was intraperitoneally injected into mice (150 mg/kg), and ten minutes later, intraperitoneal injection was performed. Mice were anesthetized with pentobarbital (50 mg/kg) and bioluminescence detection was then performed using an In-Vivo MS FX PRO system (Bruker, Billerica, MA). Luminescence images were captured with an exposure time of 30 seconds and the signal intensity of the tumors was measured by Bruker MI software. The survival time of each mouse was recorded up to 120 days. All animal experiments were carried out in strict accordance with the experimental protocol approved by the Animal Ethics Committee of Shanghai Medical College of Fudan University.
除非另有说明,本发明数据以平均值±标准差表示。对于动物模型的总光子通量,通过单尾Mann Whitney检验来确定显著性。我们应用Mantel-Cox检验来比较两组异种移植模型的存活率。在所有分析中,p<0.05被认为是统计学显著的。The data of the present invention are expressed as mean ± standard deviation unless otherwise stated. For the total photon flux of the animal model, significance was determined by a one-tailed Mann Whitney test. We used the Mantel-Cox test to compare the survival rates of the two groups of xenograft models. In all analyses, p < 0.05 was considered to be statistically significant.
肿瘤接种后第50天,我们检测了肿瘤的生长情况,结果如图18和图19所示,用总光子通量表示肿瘤的生长,按照生理盐水对照组、利妥昔单抗组、米哚妥林组、利妥昔单抗+米哚妥林组的顺序排列,其中所有相邻两组在肿瘤质量中均显示出统计学显著差异。On the 50th day after tumor inoculation, we examined the growth of the tumor. The results are shown in Figure 18 and Figure 19. The total photon flux is used to express the growth of the tumor according to the saline control group, rituximab group, rice bran. The order of the tolin group, the rituximab + miltazone group, in which all adjacent groups showed statistically significant differences in tumor mass.
肿瘤接种后第70天的结果如图20和图21所示,生理盐水组的7只小鼠中有3只死亡,利妥昔单抗组的7只小鼠中有1只死亡,而利妥昔单抗+米哚妥林组的所有7只小鼠均存活(图20)。存活小鼠中的肿瘤块表现出相似的结果,即生理盐水组或利妥昔单抗组的肿瘤生长速度远高于米哚妥林或联合治疗组中的肿瘤(图21)。The results on the 70th day after tumor inoculation are shown in Fig. 20 and Fig. 21, and 3 of the 7 mice in the saline group died, and 1 of the 7 mice in the rituximab group died. All 7 mice in the rituximab + miltazone group survived (Figure 20). Tumor blocks in surviving mice showed similar results, ie the tumor growth rate of the saline group or the rituximab group was much higher than that of the midazolin or combination treatment group (Fig. 21).
肿瘤接种后第90天的结果如图22和图23所示,在生理盐水组、利妥昔单抗组、米哚妥林组和联合治疗组中分别有1只、1只、4只和6只小鼠存活(图22)。用生理盐水处理的存活小鼠中的肿瘤块大于利妥昔单抗处理的存活小鼠中的肿瘤块(图23)。The results on the 90th day after tumor inoculation are shown in Figure 22 and Figure 23. In the saline group, the rituximab group, the metformin group, and the combination treatment group, there were 1, 1 and 4, respectively. Six mice survived (Figure 22). Tumor blocks in surviving mice treated with saline were larger than tumor blocks in rituximab-treated surviving mice (Fig. 23).
此外,图24的生存曲线分析表明,与单用利妥昔单抗相比,单独使用米哚妥林或与利妥昔单抗联合使用显著延长了存活率,表明由米哚妥林诱导的促凋亡作用对BL的治疗可能是必需和有益的,米哚妥林可作为补充的治疗药物使用,尤其是在利妥昔单抗耐药性BL的治疗方案中。In addition, the survival curve analysis of Figure 24 showed that the use of midazolin alone or in combination with rituximab significantly prolonged survival compared to rituximab alone, indicating induction by midazolam. Pro-apoptotic effects may be necessary and beneficial for the treatment of BL, and militalin may be used as a supplemental therapeutic, especially in the treatment of rituximab-resistant BL.
以上详细描述了本发明的较佳具体实施例。应当理解,本领域的普通技术无需创造性劳动就可以根据本发明的构思作出诸多修改和变化。因此,凡本技术领域中技术 人员依本发明的构思在现有技术的基础上通过逻辑分析、推理或者有限的实验可以得到的技术方案,皆应在由权利要求书所确定的保护范围内。The above has described in detail the preferred embodiments of the invention. It should be understood that many modifications and variations can be made in the present invention without departing from the scope of the invention. Therefore, any technical solution that can be obtained by a person skilled in the art based on the prior art based on the prior art by logic analysis, reasoning or limited experimentation should be within the scope of protection determined by the claims.

Claims (10)

  1. 一种治疗非霍奇金氏淋巴瘤的药物组合物,其特征在于,包括蛋白激酶C抑制剂和利妥昔单抗。A pharmaceutical composition for treating non-Hodgkin's lymphoma, comprising a protein kinase C inhibitor and rituximab.
  2. 如权利要求1所述的药物组合物,其特征在于,还包括药学上可接受的载体或赋形剂。The pharmaceutical composition of claim 1 further comprising a pharmaceutically acceptable carrier or excipient.
  3. 如权利要求1或2所述的药物组合物,其特征在于,所述蛋白激酶C抑制剂选自米哚妥林、其衍生物、其药学上可接受的盐、其溶剂合物和其前药中的一种或多种。The pharmaceutical composition according to claim 1 or 2, wherein the protein kinase C inhibitor is selected from the group consisting of militalin, a derivative thereof, a pharmaceutically acceptable salt thereof, a solvate thereof and a former thereof One or more of the medicines.
  4. 一种治疗非霍奇金氏淋巴瘤的药物组合物,其特征在于,包括蛋白激酶C抑制剂和药学上可接受的载体形成的第一制剂,及利妥昔单抗和药学上可接受的载体形成的第二制剂。A pharmaceutical composition for treating non-Hodgkin's lymphoma, comprising a first formulation comprising a protein kinase C inhibitor and a pharmaceutically acceptable carrier, and rituximab and a pharmaceutically acceptable A second formulation of the carrier.
  5. 如权利要求4所述的药物组合物,其特征在于,所述制剂的剂型为注射给药制剂、经胃肠道给药制剂、呼吸道给药制剂、皮肤给药制剂、粘膜给药制剂或腔道给药制剂。The pharmaceutical composition according to claim 4, wherein the preparation is in the form of an injection administration preparation, a gastrointestinal administration preparation, a respiratory administration preparation, a dermal administration preparation, a mucosal administration preparation or a cavity. Dosage preparation.
  6. 如权利要求4所述的药物组合物,其特征在于,所述蛋白激酶C抑制剂选自米哚妥林、其衍生物、其药学上可接受的盐、其溶剂合物和其前药。The pharmaceutical composition according to claim 4, wherein the protein kinase C inhibitor is selected from the group consisting of militalin, a derivative thereof, a pharmaceutically acceptable salt thereof, a solvate thereof, and a prodrug thereof.
  7. 利妥昔单抗和蛋白激酶C抑制剂在制备非霍奇金氏淋巴瘤治疗联合用药物中的应用。The use of rituximab and protein kinase C inhibitors in the preparation of a combination therapy for non-Hodgkin's lymphoma.
  8. 如权利要求7所述的应用,其特征在于,所述非霍奇金氏淋巴瘤为B细胞非霍奇金氏淋巴瘤。The use according to claim 7, wherein the non-Hodgkin's lymphoma is a B cell non-Hodgkin's lymphoma.
  9. 如权利要求7所述的应用,其特征在于,所述非霍奇金氏淋巴瘤为对利妥昔单抗耐药的非霍奇金氏淋巴瘤。The use according to claim 7, wherein said non-Hodgkin's lymphoma is a non-Hodgkin's lymphoma resistant to rituximab.
  10. 如权利要求7所述的应用,其特征在于,所述非霍奇金氏淋巴瘤为具有高度活化的PKC的非霍奇金氏淋巴瘤。The use according to claim 7, wherein the non-Hodgkin's lymphoma is a non-Hodgkin's lymphoma with highly activated PKC.
PCT/CN2019/082102 2018-05-17 2019-04-10 Pharmaceutical composition for treating non-hodgkin's lymphoma WO2019218810A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810475374.7A CN110496223B (en) 2018-05-17 2018-05-17 Pharmaceutical composition for treating non-Hodgkin's lymphoma
CN201810475374.7 2018-05-17

Publications (1)

Publication Number Publication Date
WO2019218810A1 true WO2019218810A1 (en) 2019-11-21

Family

ID=68539411

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/082102 WO2019218810A1 (en) 2018-05-17 2019-04-10 Pharmaceutical composition for treating non-hodgkin's lymphoma

Country Status (2)

Country Link
CN (1) CN110496223B (en)
WO (1) WO2019218810A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102821759A (en) * 2010-03-30 2012-12-12 诺华有限公司 PKC inhibitors for the treatment of B-cell lymphoma having chronic active B-cell-receptor signalling
WO2018045240A1 (en) * 2016-09-01 2018-03-08 Denovo Biopharma Llc Methods and composition for the prediction of the activity of enzastaurin

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100346830C (en) * 2005-12-28 2007-11-07 上海交通大学医学院附属瑞金医院 Medicinal composition for treating B cell lymph tumour
JP6631616B2 (en) * 2014-07-26 2020-01-15 ノース・アンド・サウス・ブラザー・ファーマシー・インベストメント・カンパニー・リミテッド 2-Amino-pyrido [2,3-d] pyrimidin-7 (8H) -one derivatives as CDK inhibitors and uses thereof
JO3589B1 (en) * 2014-08-06 2020-07-05 Novartis Ag Protein kinase c inhibitors and methods of their use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102821759A (en) * 2010-03-30 2012-12-12 诺华有限公司 PKC inhibitors for the treatment of B-cell lymphoma having chronic active B-cell-receptor signalling
WO2018045240A1 (en) * 2016-09-01 2018-03-08 Denovo Biopharma Llc Methods and composition for the prediction of the activity of enzastaurin

Also Published As

Publication number Publication date
CN110496223B (en) 2021-09-10
CN110496223A (en) 2019-11-26

Similar Documents

Publication Publication Date Title
Chen et al. Recent update of HDAC inhibitors in lymphoma
US10869868B2 (en) Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia
EP3888659A1 (en) Combination therapy for treating cancer
US20130164283A1 (en) Antibody induced cell membrane wounding
US20190070183A1 (en) Targeting Casein Kinase-1 and PI3K/AKT/mTOR Pathways for Treatment of c-Myc-Overexpressing Cancers, Organ Transplant Associated Complications and Autoimmune Diseases
KR102613106B1 (en) Cerdulatinib for the treatment of b-cell malignancies
CN110958881A (en) Immunomodulatory retinoids and REXINOID compounds for use in cancer immunotherapy in combination with immunomodulators
CN108367006B (en) Cerdulatinib for treating hematologic cancers
BR112019017851A2 (en) method for treating cancer in an individual who needs it, method for identifying an individual who has cancer as a candidate for treatment with a smarca2 antagonist, method for identifying a cancer cell as sensitive to treatment with a smarca2 antagonist, antagonist of smarca2 for use in the treatment of cancer in an individual who needs it, antagonist of smarca2 for use as a medicine in the treatment of cancer in an individual who needs it and use of the antagonist of smarca2 in the manufacture of a medicine in the treatment of cancer in an individual who needs the same
CN113301896A (en) EP4 inhibitors and synthesis thereof
WO2020132259A1 (en) Compositions and methods of treating cancers by administering a phenothiazine-related drug that activates protein phosphatase 2a (pp2a)
JP2022523423A (en) 2&#39;3&#39;-Cyclic dinucleotide and prodrugs thereof
JP7350872B2 (en) 3&#39;3&#39;-cyclic dinucleotide and its prodrug
Hudson et al. Crizotinib induces apoptosis and gene expression changes in ALK+ anaplastic large cell lymphoma cell lines; brentuximab synergizes and doxorubicin antagonizes
WO2019218810A1 (en) Pharmaceutical composition for treating non-hodgkin&#39;s lymphoma
WO2020007087A1 (en) Pharmaceutical composition for treating b cell lybphoma
CA3149112A1 (en) Novel therapeutic use
TW202241455A (en) Combination therapy by using akr1c3-activated compound with immune checkpoint inhibitor
WO2012078982A2 (en) Xzh-5 inhibits constitutive and interleukin-6-induced stat3 phosphorylation in human hepatocellular carcinoma cells
KR20220004025A (en) Tumor-selective combination therapy
US20150004613A1 (en) Methods for treating hematopoietic malignancies
WO2015009879A1 (en) Compositions for modulating nrf2-are activity and their methods of use
US20230338374A1 (en) Mdm2 inhibitors for use in the treatment or prevention of hematologic neoplasm relapse after hematopoietic cell transplantation
Hawkins Radioresistance and radiosensitization in high-risk Group 3 pediatric medulloblastoma
JP2019511544A (en) Pharmaceutical use of resminostat in Asian patients

Legal Events

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

Ref document number: 19804269

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19804269

Country of ref document: EP

Kind code of ref document: A1