CN112190711A - Application of NLRP3 inhibitor in preparation of anti-DLBCL (dendritic cell death factor) medicines - Google Patents
Application of NLRP3 inhibitor in preparation of anti-DLBCL (dendritic cell death factor) medicines Download PDFInfo
- Publication number
- CN112190711A CN112190711A CN202011196043.3A CN202011196043A CN112190711A CN 112190711 A CN112190711 A CN 112190711A CN 202011196043 A CN202011196043 A CN 202011196043A CN 112190711 A CN112190711 A CN 112190711A
- Authority
- CN
- China
- Prior art keywords
- diffuse large
- cell lymphoma
- inhibitor
- dlbcl
- tnf
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57407—Specifically defined cancers
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57484—Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
- G01N33/57492—Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/705—Assays involving receptors, cell surface antigens or cell surface determinants
- G01N2333/70503—Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
- G01N2333/70521—CD28, CD152
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/705—Assays involving receptors, cell surface antigens or cell surface determinants
- G01N2333/70503—Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
- G01N2333/70532—B7 molecules, e.g. CD80, CD86
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- Biomedical Technology (AREA)
- Hematology (AREA)
- Molecular Biology (AREA)
- Urology & Nephrology (AREA)
- Cell Biology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Analytical Chemistry (AREA)
- Microbiology (AREA)
- Oncology (AREA)
- Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Biochemistry (AREA)
- General Physics & Mathematics (AREA)
- Biotechnology (AREA)
- Pathology (AREA)
- Public Health (AREA)
- Hospice & Palliative Care (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Organic Chemistry (AREA)
Abstract
The invention particularly relates to application of an NLRP3 inhibitor in preparation of anti-DLBCL drugs, and the clinical treatment of diffuse large B cell lymphoma still has the defects of unsatisfactory treatment effect and poor prognosis of a part of patients, and relapse and refractory cases cannot be relieved by conventional therapy. The invention proves that the level of immunosuppressive cells in a DLBCL patient is increased and the anti-tumor immune function of T cells is damaged by analyzing the phenotype of peripheral blood T cells of the DLBCL patient and healthy people. The research of the invention also proves that the expression of PD-1 and TIM-3 is related to poor prognosis of DLBCL, in addition, NLRP3 inflammasome activation exists in a DLBCL patient, the effector cytokine of the DLBCL is positively related to the expression of PD-L1, the expression of co-suppression ligand PD-L1 can be obviously reduced through blocking NLRP3 inflammasome, and the level of the immunosuppressive cell population of a lymphoma mouse is reduced.
Description
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to an application of an NLRP3 inhibitor in preparation of a diffuse large B cell lymphoma drug.
Background
The information in this background section is only for enhancement of understanding of the general background of the invention and is not necessarily to be construed as an admission or any form of suggestion that this information forms the prior art that is already known to a person of ordinary skill in the art.
Diffuse large B-cell lymphoma (DLBCL) accounts for 30-35% of all non-Hodgkin lymphomas (NHL). Although rituximab-based chemotherapy has significantly improved efficacy as a first-line therapy, one-third of patients relapse or are refractory, with a poor clinical prognosis. 90% of relapsed or refractory cases cannot be relieved by conventional rescue immunochemotherapy and autologous stem cell transplantation, so that further elucidation of the pathogenesis of DLBCL, evaluation of high-risk factors, and search for new therapeutic targets are urgently needed.
Malignant tumors can suppress the body's anti-tumor immune response in a variety of ways, escaping the immune surveillance of the host. Immune checkpoints are molecules expressed in immune cells and tumor cells that regulate immune homeostasis, and can achieve immune escape from tumors by inhibiting the function of anti-tumor immune cells such as T cells and NK cells. There is upregulation of the immune checkpoint molecule programmed cell death ligand 1(PD-L1) in a variety of solid tumors and interacts with programmed cell death protein 1(PD-1) expressed on activated T cells to deliver inhibitory signals that inhibit T cell proliferation and cytokine production. In addition, there are various populations of immune-downregulated cells in tumors, including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and regulatory T cells (Tregs), among others, that attenuate the body's anti-tumor immune response, thereby promoting tumor progression, recurrence, and metastasis. In DLBCL, in addition to the occurrence of typical genetic events, there is also immune escape. The research finds that MDSCs and Tregs are amplified in DLBCL peripheral blood, the expression of PD-1 and TIM-3 expressed by T cells is obviously increased, the expression of PD-L1 in DLBCL tumor tissues is obviously up-regulated, meanwhile, the expression of PD-1 and TIM-3 is proved to be related to various adverse prognostic factors, and the effect of immunosuppression in the onset of DLBCL is highlighted. However, the exact mechanism by which immunosuppression exists in DLBCL is not well defined, and knowledge of the underlying immunosuppressive mechanisms in DLBCL helps to identify biomarkers in the tumor microenvironment, reverse the immunosuppressed state, and improve the immunotherapeutic response.
Inflammation plays a key role in tumorigenesis, and the inflammatory microenvironment is an important component of tumors. Inflammatory bodies are important factors that lead to the release of inflammatory cytokines that in turn produce the inflammatory cascade. NLRP3 is the most widely studied inflammasome, invasion by foreign pathogens or injury and death of cells in vivo, and can recruit, assemble and activate NLRP3 inflammasome, activate Caspase-1, cleave IL-1 beta and IL-18 precursors, and produce active IL-1 beta and IL-18. On the one hand, NLRP3 inflammasome plays an important role in the body's ability to limit pathogen invasion and clear endogenous lesions; on the other hand, the accumulated effect of the components generated by the activation can form an environment suitable for the growth of the tumor, thereby promoting the growth and the metastasis of the tumor. Previous studies by the inventors found that NLRP3 inflammasome is activated in lymphoma cell lines, indicating its potential as a therapeutic target. However, the mechanism of inflammasome in regulating tumor microenvironment, tumorigenesis and tumor immunity is not clear. Recent studies have shown that NLRP3 inflammasome and its effector cytokines IL-1. beta. and IL-18 are involved in the immune escape of tumors. IL-1. beta. and IL-18 can suppress the anti-tumor immune response of the body by recruiting and activating MDSCs or inducing immune checkpoint expression, etc. Therefore, immunosuppression may be one of the mechanisms by which NLRP3 inflammasome induces tumorigenesis. At present, whether the activation of NLRP3 exists in DLBCL and the relation between the activation and the tumor immunosuppression is not reported.
Disclosure of Invention
Based on the above background, the present invention aims to clarify the mechanism of action of NLRP3 inflammasome-mediated immunosuppression in diffuse large B-cell lymphoma, and to provide new therapeutic ideas and drugs for further improving the treatment of diffuse large B-cell lymphoma.
Aiming at the technical purpose, the invention provides the following technical scheme:
in a first aspect of the invention, there is provided the use of an IFN-gamma and/or TNF-alpha agonist as an active ingredient against diffuse large B-cell lymphoma.
Type II interferon IFN-gamma is widely involved in immune and inflammatory response, and TNF-alpha (tumor necrosis factor) has the ability to directly kill tumor cells. The research result of the invention shows that the generation of IFN-gamma and TNF-alpha by peripheral blood T cells of a DLBCL patient is obviously reduced compared with that of a healthy contrast person, the expression of PD-1 and TIM-3 on the surface of the T cells is obviously up-regulated, and the result shows that the anti-tumor immune function of the T cells of the DLBCL patient is damaged. According to the general research thought in the field, the treatment effect on diffuse large B cell lymphoma is expected to be realized by promoting the expression of IFN-gamma and TNF-alpha in a tumor patient body or inhibiting the expression of PD-1 and/or TIM-3.
In a second aspect of the present invention, there is provided the use of a PD-1 and/or TIM-3 inhibitor as an active ingredient against diffuse large B-cell lymphoma.
In a third aspect of the invention, there is provided the use of PD-1 and/or PD-L1 as a diagnostic marker against diffuse large B-cell lymphoma.
It is well known in the art that Lactate Dehydrogenase (LDH) is one of the important enzyme systems for anaerobic glycolysis of sugars and gluconeogenesis, and is clinically used for diagnosing myocardial infarction, liver disease, and partial malignancy. Ki-67 is a marker that reflects tumor proliferation. LDH and Ki-67 levels are closely correlated with DLBCL prognosis.
According to the results of the present study, PD-1 in the peripheral blood of DLBCL patients+The T cell ratio is positively correlated with serum Lactate Dehydrogenase (LDH) level, and the content of PD-L1 expressed by the tumor tissue of the DLBCL patient is positively correlated with the expression of Ki-67 in the tumor tissue. Based on the findings of this study, one skilled in the art can conclude that PD-1 and/or PD-L1 are expected to be a diagnostic marker for DLBCL based on general research considerations.
In a third aspect of the invention, the use of NLRP3 inflammasome as a target for treatment of diffuse large B-cell lymphoma is provided.
To validate the feasibility of NLRP3 inflammasome as a therapeutic target, the present invention administered an NLRP3 inflammasome inhibitor against a lymphoma disease model, and the results showed that the growth of mouse lymphoma was inhibited; further, the present study also demonstrates that after blocking the NLRP3 inflammasome in vivoCD3 in lymphoma mice+、CD4+And CD8+The proportion of T cells is increased, the level of TNF-alpha is increased, the expression of T cells PD-1 and TIM-3 of a lymphoma mouse can be obviously reduced, the level of immunosuppressive cell population of the lymphoma mouse is reduced, and the diseased level of a diffuse large B cell lymphoma mouse is comprehensively improved. It is suggested that NLRP3 inflammasome can be used as a therapeutic target, and the therapeutic effect on diffuse large B cell lymphoma is realized by inhibiting the expression of NLRP3 inflammasome.
In a fourth aspect of the present invention, there is provided the use of an inhibitor of IL-18 as an active ingredient against diffuse large B-cell lymphoma.
Relevant studies in the art have shown that the inflammasome is capable of regulating the activation of caspase-1 (caspase-1) and thus promoting the maturation and secretion of the cytokine precursors pro-IL-1 β and pro-IL-18 during the course of the innate immune defense. In order to determine whether the NLRP3 inflammasome has expression in diffuse large B cell lymphoma or not, the detection is carried out aiming at the levels of effector cytokines IL-1 beta and IL-18 generated by the activation of the NLRP3 inflammasome, and the detection result shows that the expression of the IL-18 in DLBCL tumor tissues is obviously increased compared with that of a control group, the expression content of the IL-18 is positively correlated with that of PD-L1, and the inhibition of the expression of the IL-18 is expected to realize the treatment effect on the diffuse large B cell lymphoma.
The beneficial effects of one or more technical schemes are as follows:
according to the research result of the invention, the action mode of the NLRP3 inflammasome in diffuse large B cell lymphoma is further determined, and according to the research result of the invention, the inhibition of the NLRP3 inflammasome can comprehensively realize the treatment effect on the diffuse large B cell lymphoma through immunosuppression and inhibit the growth of tumors. The research result of the invention further defines the action mechanism of NLRP3 inflammasome, is beneficial to the development of clinical treatment drugs for diffuse large B cell lymphoma, and has important clinical significance.
Drawings
The accompanying drawings, which are incorporated in and constitute a part of this specification, are included to provide a further understanding of the invention, and are incorporated in and constitute a part of this specification, illustrate exemplary embodiments of the invention and together with the description serve to explain the invention and not to limit the invention.
FIG. 1 is peripheral blood T cell function of the DLBCL patient described in example 1;
wherein, FIG. 1A shows the IFN-y content in peripheral blood of DLBCL patients;
FIG. 1B is a graph of TNF- α content in peripheral blood of patients with DLBCL.
FIG. 2 is a graph of the expression of peripheral blood T cell co-inhibitory receptors of the DLBCL patients described in example 1;
wherein FIG. 2A is the levels of PD-1 phenotype T cells in the peripheral blood of the control group and the patients;
FIG. 2B is a graph showing the T cell content of the Tim-3 phenotype in the peripheral blood of the control group and the patients;
FIG. 2C is a graph showing the BTLA-phenotype T cell content in the peripheral blood of a control group and patients;
FIG. 2D is a graph of TIGIT phenotype T cell content in peripheral blood of control and patients;
FIG. 2E is a graph showing the levels of T cells of the CD160 phenotype in the peripheral blood of a control group and patients;
FIG. 2F is a graph showing the LAG-3 phenotype T cell content in peripheral blood of control group and patients.
FIG. 3 is a graph of the correlation of PD-1 and TIM-3 expression with serum LDH levels as described in example 1;
FIG. 4 is a graph of PD-1 and TIM-3 expression versus Ann Arbor staging as described in example 1;
FIG. 5 is a graph of PD-1 and TIM-3 expression versus IPI score as described in example 1;
FIG. 6 is the expression level of PD-L1 in DLBCL tumor tissue and normal tissue in example 1;
FIG. 7 is a graph of the relationship between PD-L1 expression and Ki-67 in DLBCL tumor tissue as described in example 1;
FIG. 8 is the ratio of MDSCs and Tregs in DLBCL peripheral blood as described in example 1;
FIG. 9 is the expression levels of IL-1 β and IL-18 in the DLBCL tumor tissue described in example 1;
FIG. 10 is a graph of the expression of IL-1. beta. and IL-18 in DLBCL tumor tissue analyzed with CD10 and MUM1 expression as described in example 1;
FIG. 11 is a correlation analysis of the expression of IL-1. beta. and IL-18 in DLBCL tumor tissue and the expression of PD-L1 as described in example 1;
FIG. 12 is the tumor size and growth curves of the tumor-bearing mice described in example 1;
FIG. 13 is a graph showing the expression of tumor and spleen NLRP 3-related proteins and PD-L1 in MCC 950-treated mice as described in example 1;
FIG. 14 shows the proportion of splenic T cells and the level of secreted TNF-. alpha.in mice treated with MCC950 described in example 1;
FIG. 15 shows surface PD-1 and TIM-3 expression of spleen and lymph node T cells of mice of the control group and MCC950 group described in example 1;
FIG. 16 is a graph of the expression measurements of MDSC, TAM and Treg in peripheral blood, spleen and tumors of mice as described in example 1.
Detailed Description
It is to be understood that the following detailed description is exemplary and is intended to provide further explanation of the invention as claimed. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
It is noted that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of exemplary embodiments according to the invention. As used herein, the singular forms "a", "an" and "the" are intended to include the plural forms as well, and it should be understood that when the terms "comprises" and/or "comprising" are used in this specification, they specify the presence of stated features, steps, operations, devices, components, and/or combinations thereof, unless the context clearly indicates otherwise.
As introduced in the background art, aiming at the phenomena of difficult treatment and poor prognosis of diffuse large B-cell lymphoma in the prior art, the invention aims to further define the pathogenesis of the diffuse large B-cell lymphoma and provide a new treatment target for the treatment of the diffuse large B-cell lymphoma in order to solve the technical problems.
In a first aspect of the invention, there is provided the use of an IFN-gamma and/or TNF-alpha agonist as an active ingredient against diffuse large B-cell lymphoma.
Preferably, the IFN-gamma and/or TNF-alpha agonist is an IFN-gamma agonist, a TNF-alpha agonist or a mixture of IFN-gamma agonists, TNF-alpha agonists, or a substance that agonizes both IFN-gamma and TNF-alpha.
Preferably, the application mode of the active ingredient for resisting the diffuse large B cell lymphoma comprises but is not limited to the application of the IFN-gamma and/or TNF-alpha agonist in preparing a medicine for resisting the diffuse large B cell lymphoma or a health product.
In some embodiments of the above preferred embodiments, the anti-diffuse large B cell lymphoma drug comprises IFN- γ and/or TNF- α agonists and pharmaceutically necessary excipients.
In other embodiments of the above preferred technical solution, the anti-diffuse large B cell lymphoma drug comprises the IFN- γ and/or TNF- α agonist and pharmaceutically necessary excipients, and further comprises other components having anti-diffuse large B cell lymphoma activity.
In a second aspect of the present invention, there is provided the use of a PD-1 and/or TIM-3 inhibitor as an active ingredient against diffuse large B-cell lymphoma.
Preferably, the PD-1 and/or TIM-3 inhibitor is a PD-1 inhibitor, a TIM-3 inhibitor or a composition of a PD-1 inhibitor and a TIM-3 inhibitor, or a substance inhibiting both PD-1 and TIM-3.
In a third aspect of the invention, there is provided the use of PD-1 and/or PD-L1 as a diagnostic marker against diffuse large B-cell lymphoma.
Preferably, the application mode of the PD-1 and/or PD-L1 as the diagnosis marker for the diffuse large B cell lymphoma includes but is not limited to the application of a PD-1 and/or PD-L1 content detection reagent in the preparation of a diagnosis kit for the diffuse large B cell lymphoma.
In a third aspect of the invention, the use of NLRP3 inflammasome as a target for treatment of diffuse large B-cell lymphoma is provided.
Preferably, the application mode of the NLRP3 inflammasome as a target for treating diffuse large B cell lymphoma comprises the application of an NLRP3 inflammasome inhibitor in preparing a drug for resisting diffuse large B cell lymphoma.
Further preferably, the NLRP3 inflammasome inhibitor is MCC 950.
Further preferably, in the anti-diffuse large B cell lymphoma drug, the NLRP3 inflammasome inhibitor is used as CD3+、CD4+And CD8+T cell agonists.
Further preferably, in the anti-diffuse large B-cell lymphoma drug, the NLRP3 inflammasome inhibitor acts as a TNF- α agonist.
Further preferably, in the anti-diffuse large B-cell lymphoma drug, the NLRP3 inflammasome inhibitor is used as a PD-1 and TIM-3 inhibitor.
Further preferably, in the anti-diffuse large B-cell lymphoma drug, the NLRP3 inflammasome inhibitor is used as an immunosuppressive cell population inhibitor.
In a fourth aspect of the present invention, there is provided the use of an inhibitor of IL-18 as an active ingredient against diffuse large B-cell lymphoma.
In order to make the technical solutions of the present invention more clearly understood by those skilled in the art, the technical solutions of the present invention will be described in detail below with reference to specific embodiments.
Example 1
Characterization of the anti-tumor immune response in patients with DLBCL
1.1 expression of PD-1 and TIM-3 in DLBCL patients and their relationship to clinical characteristics
To determine whether the anti-tumor immune response in DLBCL patients was impaired, 59 first-diagnosed DLBCL patients and 59 healthy controls were collected in this example, and the phenotype and function of peripheral blood T cells were examined by flow cytometry. The results show that: peripheral blood T cells from DLBCL patients produced significantly less IFN- γ (P ═ 0.0153) (fig. 1A) and TNF- α (P ═ 0.0183) (fig. 1B) than healthy controls. Detecting the expression of T cell surface immune check point molecules, and displaying that: compared with the control group, the expression of PD-1(P ═ 0.0003) and TIM-3(P <0.0001) on the surface of T cells was significantly up-regulated, and the other co-inhibitory receptors BTLA, TIGIT, CD160 and LAG-3 were not significantly different (P >0.05) compared to the control group (fig. 2). Thus, the T cell anti-tumor immune function of DLBCL patients is impaired.
1.2 relationship of PD-1 and TIM-3 expression to clinical characteristics and prognosis
The results show that PD-1 in peripheral blood of the patient with DLBCL at the initial diagnosis+The T cell proportion is positively correlated with serum Lactate Dehydrogenase (LDH) levels (r: 0.4999; P)<0.0001), whereas TIM-3 has no clear correlation with LDH levels (r ═ 0.1462; p-0.2693) (fig. 3). The expression of PD-1 and TIM-3 by T cells is closely related to lymphoma staging and the International Prognostic Index (IPI): initial DLBCL patients at Ann Arbor III/IV phase expressed PD-1(P ═ 0.0027) and TIM-3(P ═ 0.0051) significantly higher T cells than Ann Arbor I/II phase patients (fig. 4). In addition, patients scored 3-5 for IPI expressed PD-1(P ═ 0.0025) and TIM-3 significantly higher in T cells than patients scored 0-2 for IPI (P ═ 0.0185) (fig. 5). It can be seen that PD-1 and TIM-3 expression correlate with poor prognosis of DLBCL.
1.3DLBCL tumor tissue co-suppression ligand PD-L1 expression
In this example, 35 cases of primary diagnosis DLBCL tumor tissues and 35 cases of normal lymphoid tissues were collected to prepare microarray tissue chips. Immunohistochemical staining was performed and scored for staining intensity and positive cell proportion. The results show that: the expression of DLBCL tumor tissue PD-L1 is obviously up-regulated compared with normal lymph tissue (P)<0.0001) (fig. 6). Further research shows that the expression of PD-L1 is positively correlated with the expression of Ki-67 in the tumor tissue of DLBCL patients (r is 0.4880; P is 0.0029), and the DLBCL patients are divided into Ki-67 patients by taking 60 percent of Ki-67 expression as a boundarylowAnd Ki-67highTwo groups, found Ki-67highPD-L1 expression was significantly increased in group (P ═ 0.0079) (fig. 7).
1.4 levels of immunosuppressive cells in patients with DLBCL
Further analysis of the number and distribution of Myeloid-derived suppressor cells (MDSCs) and Tregs in DLBCL patients revealed a significant increase in the ratio of MDSCs (P <0.0001) to Tregs (P ═ 0.0068) in peripheral blood of DLBCL naive patients compared to healthy controls (fig. 8).
In conclusion, the expression of the co-suppression ligand at the tumor part of the DLBCL patient is obviously up-regulated, and the level of the in vivo immunosuppressive cell population is obviously increased.
2. Detection of NLRP3 inflammasome in DLBCL patients
To clarify the expression of NLRP3 inflammasome in DLBCL, this example examined the levels of the effector cytokines IL-1. beta. and IL-18 produced by its activation. The immunohistochemical staining of IL-1 β and IL-18 using the tissue chip showed that the expression level of IL-18 was significantly increased in DLBCL tumor tissue compared to the control (P. 0.0440), whereas IL-1 β expression was not significantly different between tumor tissue and control (P >0.05) (fig. 9). Further studies found that IL-18 was expressed at a higher level in non-GCB type DLBCL (P ═ 0.0261) compared to GCB type DLBCL patients (fig. 9). In addition, IL-18 and IL-1 β were expressed at significantly lower levels in CD10(+) patients than in CD10(-) patients (P ═ 0.0381), while IL-18 and IL-1 β were expressed at significantly higher levels in MUM1(+) patients than in MUM1(-) patients (P ═ 0.0267) (fig. 10). The correlation between IL-18 and IL-1 β and PD-L1 expression was analyzed and IL-18 levels in DLBCL tumor tissues were found to be positively correlated with PD-L1 expression (r 0.4750; P <0.0001) and IL-1 β was not significantly correlated with PD-L1 expression (r 0.1484; P0.2348) (fig. 11). In conclusion, the DLBCL patient has NLRP3 inflammasome activation, and the effector cytokine is positively correlated with the expression of PD-L1.
Effect of NLRP3 inflammasome on lymphoma mouse model pathogenesis
A tumor-bearing mouse model is constructed by using a BALB/c mouse syngeneic lymphoma cell strain A20, a tumor growth curve is drawn, the growth conditions of the lymphoma of mice in a control group (PBS treatment group) and an NLRP3 inhibition group (MCC950 treatment group) are compared, the size of the lymphoma of the mice in the NLRP3 inhibition group is remarkably reduced (P is 0.0308) compared with that of the control group, the lymphoma of the mice is inhibited, and the tumor progress is delayed compared with that of the control group (figure 12). At the same time, Western Blot verifies the blocking effect of MCC950 on NLRP3 inflammasome in tumor-bearing mice, and shows that MCC950 significantly down-regulates the expression of NLRP 3-related proteins (NLRP3, Caspase-1, cleared Caspase-1, Pro-IL-1 beta, cleared IL-1 beta and IL-18) in tumor tissues and spleen, and the activation marks that the protein levels of cleared IL-1 beta and IL-18 in tumors are obviously reduced (FIG. 13). Meanwhile, Western Blot detection of mouse tumor tissues shows that the MCC950 can block NLRP3 inflammatory mice to remarkably down-regulate the expression of co-suppression ligand PD-L1 (figure 13).
Effect of NLRP3 inflammasome on anti-tumor immune response in lymphoma mouse model
4.1 in vivo blockade of the effects of NLRP3 inflammasome on T cell proportion and function
And (3) displaying a flow detection result: compared with a control group, the NLRP3 inhibits spleen CD3 of tumor-bearing mice of the group+(P=0.0089)、CD4+(P ═ 0.0050) and CD8+(P ═ 0.0177) the proportion of T cells was significantly increased; CD3+(P ═ 0.0033) and CD4+(P ═ 0.0052) T cells secreted significantly increased levels of TNF- α (fig. 14).
4.2 in vivo blockade of the Effect of NLRP3 inflammatory bodies on T cell surface PD-1 and TIM-3 expression
And (3) displaying a flow detection result: spleen CD3+(P ═ 0.0094) and CD4+(P-0.0045) the proportion of T cells expressing PD-1 was significantly reduced, and lymph node CD3+(P ═ 0.0318) and CD4+(P-0.0241) the percentage of T cells decreased significantly; spleen CD3+(P=0.0009)、CD4+(P-0.0001) and CD8+(P ═ 0.0136) T cells expressed TIM-3 significantly reduced; lymph node CD3+(P-0.0476) and CD4+(P ═ 0.0278) the proportion of T cells expressing TIM-3 was significantly reduced (fig. 15). In conclusion, MCC950 blocks NLRP3 inflammatory bodies and can remarkably reduce the expression of PD-1 and TIM-3 of T cells of lymphoma mice.
4.3 in vivo blockade of the effects of NLRP3 inflammasome on MDSCs, TAMs and Tregs levels
And (3) displaying a flow detection result: tumor-infiltrating MDSCs (Gr-1)+CD11b+)(P=0.0514)、TAMs(CD11b+F4/80+)(P<0.0001) and Tregs (CD 4)+CD25+Foxp3+) (P ═ 0.0003) was significantly reduced; spleen MDSCs (P ═ 0.0061), TAMs (P ═ 0.0046) and Tregs (P ═ 0.0018) were significantly reduced; MDSCs (P ═ 0.0096) and TAMs (P ═ 0.0077) were significantly reduced in peripheral blood (fig. 16). Taken together, MCC950 blocking NLRP3 inflammasome significantly reduced the level of immunosuppressive cell populations in lymphoma mice.
The above description is only a preferred embodiment of the present invention and is not intended to limit the present invention, and various modifications and changes may be made by those skilled in the art. Any modification, equivalent replacement, or improvement made within the spirit and principle of the present invention should be included in the protection scope of the present invention.
Claims (10)
- Use of an IFN-gamma and/or TNF-alpha agonist as an active ingredient against diffuse large B-cell lymphoma.
- 2. Use of an IFN-gamma and/or TNF-alpha agonist as defined in claim 1 as active ingredient against diffuse large B-cell lymphoma, wherein said IFN-gamma and/or TNF-alpha agonist is an IFN-gamma agonist, a TNF-alpha agonist or a mixture of IFN-gamma agonists, TNF-alpha agonists or a substance which simultaneously agonizes IFN-gamma and TNF-alpha.
- 3. The use of an IFN- γ and/or TNF- α agonist as defined in claim 1 as an active ingredient against diffuse large B-cell lymphoma, wherein said use as an active ingredient against diffuse large B-cell lymphoma is carried out in a manner including, but not limited to, the use of said IFN- γ and/or TNF- α agonist in the preparation of a medicament, or nutraceutical, against diffuse large B-cell lymphoma;preferably, the anti-diffuse large B cell lymphoma drug consists of the IFN-gamma and/or TNF-alpha agonist and pharmaceutically necessary auxiliary materials;preferably, the anti-diffuse large B cell lymphoma drug comprises the IFN-gamma and/or TNF-alpha agonist and pharmaceutically necessary auxiliary materials, and also comprises other components with anti-diffuse large B cell lymphoma activity.
- Use of a PD-1 and/or TIM-3 inhibitor as an active ingredient against diffuse large B-cell lymphoma; preferably, the PD-1 and/or TIM-3 inhibitor is a PD-1 inhibitor, a TIM-3 inhibitor or a composition of a PD-1 inhibitor and a TIM-3 inhibitor, or a substance inhibiting both PD-1 and TIM-3.
- The application of PD-1 and/or PD-L1 as a diagnostic marker for diffuse large B cell lymphoma; preferably, the application mode of the PD-1 as the diagnosis marker for the diffuse large B cell lymphoma includes but is not limited to the application of a PD-1 and/or PD-L1 content detection reagent in the preparation of a diagnosis kit for the diffuse large B cell lymphoma.
- Use of NLRP3 inflammasome as a target for treatment of diffuse large B-cell lymphoma.
- 7. The use of the NLRP3 inflammasome according to claim 6 as a target for treatment of diffuse large B-cell lymphoma, wherein the NLRP3 inflammasome is used as a target for treatment of diffuse large B-cell lymphoma, and the NLRP3 inflammasome inhibitor is used for preparing an anti-diffuse large B-cell lymphoma drug.
- 8. The use of the NLRP3 inflammasome of claim 7 as a target for treatment of diffuse large B-cell lymphoma, wherein the NLRP3 inflammasome inhibitor is MCC 950;or the medicament for resisting diffuse large B cell lymphoma, the NLRP3 inflammasome inhibitor is used as CD3+、CD4+And CD8+A T cell agonist;or the anti-diffuse large B cell lymphoma drug, the NLRP3 inflammasome inhibitor is used as a TNF-alpha agonist.
- 9. The use of the NLRP3 inflammasome of claim 7 as a target for treatment of diffuse large B-cell lymphoma, wherein in the anti-diffuse large B-cell lymphoma medicament, the NLRP3 inflammasome inhibitor acts as a PD-L1 inhibitor;or the anti-diffuse large B cell lymphoma drug, the NLRP3 inflammatory body inhibitor is used as an immunosuppressive cell population inhibitor.
- Use of an inhibitor of IL-18 as an active ingredient against diffuse large B-cell lymphoma.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202011196043.3A CN112190711A (en) | 2020-10-30 | 2020-10-30 | Application of NLRP3 inhibitor in preparation of anti-DLBCL (dendritic cell death factor) medicines |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202011196043.3A CN112190711A (en) | 2020-10-30 | 2020-10-30 | Application of NLRP3 inhibitor in preparation of anti-DLBCL (dendritic cell death factor) medicines |
Publications (1)
Publication Number | Publication Date |
---|---|
CN112190711A true CN112190711A (en) | 2021-01-08 |
Family
ID=74010626
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202011196043.3A Pending CN112190711A (en) | 2020-10-30 | 2020-10-30 | Application of NLRP3 inhibitor in preparation of anti-DLBCL (dendritic cell death factor) medicines |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN112190711A (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113406334A (en) * | 2021-06-02 | 2021-09-17 | 浙江省人民医院 | DLBCL (digital Living chromosome binding protein) related biomarker composition, application thereof and DLBCL prognosis effect prediction model |
CN114509567A (en) * | 2022-02-28 | 2022-05-17 | 成都福实生物科技有限公司 | Application of reagent for detecting spatial distribution of immune check points in preparation of tumor treatment and prognosis diagnosis products |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20180223368A1 (en) * | 2015-03-18 | 2018-08-09 | Memorial Sloan Ketteting Cancer Center | Methods for diagnosing and treating follicular lymphoma |
US20190194760A1 (en) * | 2016-05-25 | 2019-06-27 | Curevac Ag | Novel biomarkers |
-
2020
- 2020-10-30 CN CN202011196043.3A patent/CN112190711A/en active Pending
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20180223368A1 (en) * | 2015-03-18 | 2018-08-09 | Memorial Sloan Ketteting Cancer Center | Methods for diagnosing and treating follicular lymphoma |
US20190194760A1 (en) * | 2016-05-25 | 2019-06-27 | Curevac Ag | Novel biomarkers |
Non-Patent Citations (9)
Title |
---|
LU FEI等: "NLRP3 inflammasome upregulates PD-L1 expression and contributes to immune suppression in lymphoma", 《CANCER LETTERS》 * |
尚晋等: "弥漫大B细胞淋巴瘤血浆sPD-L1与IFN-γ、IL-2的相关性研究", 《福建医药杂志》 * |
徐亚文等: "PD-1/PD-L1抑制剂在血液肿瘤中的应用", 《安徽医科大学学报》 * |
曲通等: "免疫检查点抑制剂在恶性淋巴瘤中的应用研究进展", 《中国肿瘤临床》 * |
杨巍巍: "不同类型淋巴瘤患者肿瘤坏死因子的表达意义", 《齐齐哈尔医学院学报》 * |
林晓骥等: "PD-1在弥漫性大B细胞淋巴瘤患者外周血CD4~+T细胞和CD8~+T细胞上的表达和临床意义", 《中国卫生检验杂志》 * |
赵丽东等: "程序性死亡及其配体1信号通路在弥漫大B细胞淋巴瘤中研究进展", 《中国药物与临床》 * |
阿丽亚?热哈提等: "不同类型淋巴瘤患者血浆TNF-α测定及其临床意义", 《新疆医科大学学报》 * |
陈少华等: "TIM-3、LAG-3和BTLA介导的血液肿瘤免疫耐受及其逆转研究", 《中国实验血液学杂志》 * |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113406334A (en) * | 2021-06-02 | 2021-09-17 | 浙江省人民医院 | DLBCL (digital Living chromosome binding protein) related biomarker composition, application thereof and DLBCL prognosis effect prediction model |
CN114509567A (en) * | 2022-02-28 | 2022-05-17 | 成都福实生物科技有限公司 | Application of reagent for detecting spatial distribution of immune check points in preparation of tumor treatment and prognosis diagnosis products |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Hao et al. | Landscape of myeloid-derived suppressor cell in tumor immunotherapy | |
Wagner et al. | Dose intensification of TRAIL-inducing ONC201 inhibits metastasis and promotes intratumoral NK cell recruitment | |
Yin et al. | Checkpoint inhibitor pneumonitis induced by anti-PD-1/PD-L1 therapy in non-small-cell lung cancer: occurrence and mechanism | |
Wheeler et al. | Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients | |
Speiser et al. | TNF receptor p55 controls early acute graft-versus-host disease. | |
Lin et al. | High-dose per fraction radiotherapy induces both antitumor immunity and immunosuppressive responses in prostate tumors | |
Cascio et al. | Cancer-associated MSC drive tumor immune exclusion and resistance to immunotherapy, which can be overcome by Hedgehog inhibition | |
Chang et al. | Targeting tumor‐infiltrating Ly6G+ myeloid cells improves sorafenib efficacy in mouse orthotopic hepatocellular carcinoma | |
Zhao et al. | IL‐17‐producing T cells contribute to acute graft‐versus‐host disease in patients undergoing unmanipulated blood and marrow transplantation | |
Zeimet et al. | Ascitic interleukin-12 is an independent prognostic factor in ovarian cancer. | |
Duong et al. | Macrophage depletion in elderly mice improves response to tumor immunotherapy, increases anti-tumor T cell activity and reduces treatment-induced cachexia | |
Fiorcari et al. | BTK inhibition impairs the innate response against fungal infection in patients with chronic lymphocytic leukemia | |
JP7389151B2 (en) | Use of CaMK4 inhibitors in the manufacture of drugs for the treatment of psoriasis | |
Pachocki et al. | Current perspectives on diffuse midline glioma and a different role for the immune microenvironment compared to glioblastoma | |
CN112190711A (en) | Application of NLRP3 inhibitor in preparation of anti-DLBCL (dendritic cell death factor) medicines | |
Wang et al. | Therapeutic effects of C-28 methyl ester of 2-cyano-3, 12-dioxoolean-1, 9-dien-28-oic acid (CDDO-Me; bardoxolone methyl) on radiation-induced lung inflammation and fibrosis in mice | |
Fowler et al. | Clinical “cytokine storm” as revealed by monocyte intracellular flow cytometry: correlation of tumor necrosis factor α with severe gut graft-versus-host disease | |
Linz et al. | Innate immune cell recovery is positively regulated by NLRP12 during emergency hematopoiesis | |
KR101426689B1 (en) | Composition for Prevention or Treatment of Immune Disease Comprising Nutlin-3a | |
Wang et al. | The bone marrow edema links to an osteoclastic environment and precedes synovitis during the development of collagen induced arthritis | |
Carballido et al. | Clinical significance of natural killer activity in patients with transitional cell carcinoma of the bladder | |
JP6791951B2 (en) | Methods for Predicting Survival Time in Patients with Lung Cancer | |
JP2022544169A (en) | Treatment and prevention of cardiovascular disease | |
Kii et al. | STAT1-mediated induction of Ly6c-expressing macrophages are involved in the pathogenesis of an acute colitis model | |
Liu et al. | Inhibited experimental corneal neovascularization by neutralizing anti-SDF-1α antibody |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |