CN115040663B - Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma - Google Patents

Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma Download PDF

Info

Publication number
CN115040663B
CN115040663B CN202210672945.2A CN202210672945A CN115040663B CN 115040663 B CN115040663 B CN 115040663B CN 202210672945 A CN202210672945 A CN 202210672945A CN 115040663 B CN115040663 B CN 115040663B
Authority
CN
China
Prior art keywords
cells
cell
gene
multiple myeloma
application
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202210672945.2A
Other languages
Chinese (zh)
Other versions
CN115040663A (en
Inventor
万一柯
侯健
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Renji Hospital Shanghai Jiaotong University School of Medicine
Original Assignee
Renji Hospital Shanghai Jiaotong University School of Medicine
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 Renji Hospital Shanghai Jiaotong University School of Medicine filed Critical Renji Hospital Shanghai Jiaotong University School of Medicine
Priority to CN202210672945.2A priority Critical patent/CN115040663B/en
Publication of CN115040663A publication Critical patent/CN115040663A/en
Application granted granted Critical
Publication of CN115040663B publication Critical patent/CN115040663B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The application relates to the field of biological medicine, in particular to application of a solute carrier family 38member 2 gene or a protein encoded by the solute carrier family 38member 2 gene in preparation of a medicament for treating multiple myeloma. The application also provides application of the reagent interfering with the X-box binding protein 1 in preparing a medicament for treating multiple myeloma. The method for recovering the amino acid metabolism of the T cells is called an 'immune normalization' strategy, and targets the solute carrier family 38member 2 gene, so that the expression of the gene is increased to the level of a normal person, metabolic disturbance in the body of a patient with multiple myeloma is corrected, the states of T cell exhaustion, aging and apoptosis are changed, and the original functions of the T cells are recovered, thereby improving the anti-tumor effect.

Description

Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma
Technical Field
The application relates to the field of biological medicine, in particular to application of solute carrier family 38member 2 (solute carrier family 38member 2,SLC38A2) in preparing a medicament for treating multiple myeloma.
Background
Multiple Myeloma (MM) is a malignant tumor that originates in plasma cells and is characterized clinically by abnormal proliferation of plasma cells in the bone marrow, massive aggregation and production of monoclonal immunoglobulins or fragments thereof, with extensive osteolytic lesions and bone destruction. The disease accounts for about 1% of all cancers, 10% -15% of all hematological tumors, and is the second most common hematological malignancy. MM is well developed in middle-aged and elderly people, with a median age of onset of approximately 70 years. In recent years, due to the wide application of targeted drugs and the continuous development of hematopoietic stem cell transplantation technology, the disease remission rate and remission depth are improved, and the total survival of MM has been significantly improved in the past two decades. Nonetheless, MM is still an incurable disease and almost all MM patients eventually fail to treat due to relapse.
Proliferation and survival of MM cells is highly dependent on the bone marrow microenvironment (bone marrow microenvironment, BME). MM rarely involves the outside of the marrow unless it reaches the advanced stages of the disease. Immune cells, fibroblasts, endothelial cells, osteoclasts, adipocytes, plasmacytoid dendritic cells and extracellular matrix in BME constitute the soil for MM cell growth. Bone marrow of MM patients contains a large number of T lymphocytes, and these cells which can play an anti-tumor immunity function are often in an immunity-restricted state under the action of MM cells. Zelle-Rieser et al found CD8 in bone marrow of MM patients + T cells show a phenotype of depletion (expression of PD1, CTLA4, 2B4, CD 160) and senescence (expression of CD57, lack of CD 28), associated with reduced proliferation capacity and impaired function. Suen et al believe that dysfunctional T cells in MM are primarily characterized by immune senescence rather than depletion. Its senescence phenotype is KLRG1, CD57 expressing, CD28 not, and this immune senescence is independent of telomere length, suggesting that T cell senescence in MM may be reversible. Current research results show that immune-suppressed T cells in tumors are characterized by up-regulation of inhibitory receptors, reduced cytokine production, loss of proliferative capacity, and impaired cytotoxicity, and the complex mechanisms behind this are urgently needed. (Zelle-Rieser C, thangaiadivel S, biedermann R, brunner A, stoitzner P, willenbacher E, greil R, johrer K.T cells in multiple myeloma display features of exhaustion and senescence at the tumor site [ J)].J Hematol Oncol,2016,9(1):116.Suen H,Brown R,Yang S,Weatherburn C,Ho PJ,Woodland N,Nassif N,Barbaro P,Bryant C,Hart D,Gibson J,Joshua D.Multiple myeloma causes clonal T-cell immunosenescence:identification of potential novel targets for promoting tumour immunity and implications for checkpoint blockade[J].Leukemia,2016,30(8):1716-1724.Yamamoto L,Amodio N,Gulla A,Anderson KC.Harnessing the Immune System Against Multiple Myeloma:Challenges and Opportunities[J].Front Oncol,2020,10:606368.Manier S,Sacco A,Leleu X,Ghobrial IM,Roccaro AM.Bone marrow microenvironment in multiple myeloma progression[J].J Biomed Biotechnol,2012,2012:157496.Kurachi M.CD8(+)T cell exhaustion[J].Semin Immunopathol,2019,41(3):327-337.)
T lymphocytes are key effector cells for anti-tumor immunity. T cells in the MM microenvironment are not only unable to exert immune supervision and effector functions, but may even promote the occurrence and development of myeloma. How to correct the T cell immunosuppression state in MM so that the T cell immunosuppression state can play a normal anti-tumor immune function is a focus of attention in the field of MM research. In recent years, studies have found that the immune system is highly integrated with metabolism. Antitumor immunity requires the full synergy of various types of immune cells, transducing various intracellular signals to regulate their proliferation, differentiation, migration and effector functions. Metabolic activity is critical to ensure accurate performance of these processes.
The fate and function of T cells is in essence closely related to metabolism, by which cells need to produce bioenergy intermediates to support proliferation and effector functions. Glucose, glutamine and fatty acids are the primary nutritional sources of T cell metabolism while T cells can also utilize TCA cycle mechanisms and OXPHOS pathways to produce energy through glutamine metabolism or fatty acid metabolism.
For Teff and Tem cells that function as antitumor effects, the glycolytic and glutaminolytic pathways are critical for proliferation and survival of the cell. Glucose deprivation by tumor cells can impair the anti-tumor response by inhibiting T cell metabolic function. Glucose deficiency has been reported to induce a depleted T Cell phenotype characterized by an increase in expression of inhibitory receptors such as PD1, CTLA4, TIM3, LAG3, TIGIT (Zhang Y, kurupati R, liu L, zhou XY, zhang G, hudaihed A, filisio F, giles-Davis W, xu XW, karakouis GC, schuchter LM, xu W, amaravidi R, xiao M, sadek N, krepler C, herlyn M, freeman GJ, rabinowitz JD, ertl HCJ.Enhancing CD8 (+) T Cell Fatty Acid Catabolism within a Metabolically Challenging Tumor Microenvironment Increases the Efficacy of Melanoma Immunotherapy [ J ]. Cancer Cell,2017,32 (3): 3.+ -. Whery EJ, ram M.molecular and cellular insights into T Cell exhaustion [ J ]. Nature Reviews Immunology,2015,15 (8): 499.Chauvin JM,Pagliano O,Fourcade J,Sun ZJ,Wang H,Sander C,Kirkwood JM,Chen THT,Maurer M,Korman AJ,Zarour HM.TIGIT and PD-1 (+) CD8, 678, 2048..376. Activated T cells are in higher glutamine demand. Activated T cells have 5-10 times the rate of glutamine uptake compared to non-stimulated T cells, while glutamine metabolism dysfunction impairs cell proliferation and cytokine secretion (Song W, li D, tao L, luo Q, chen L.Solute carrier transporters: the metabolic gatekeepers of immune cells [ J ]. Acta Pharm Sin B,2020,10 (1): 61-78.). It was found that decreased availability of glutamine in the tumor microenvironment limited the necessary metabolic functions and biosynthetic pathways of T cells, thereby impairing anti-tumor immunity (Carr EL, kelman A, wu GS, gopal R, senkepitch E, aghvanan A, turay AM, frauwirth KA. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation [ J ]. J Immunol,2010,185 (2): 1037-1044.). The tumor microenvironment forces T cells to undergo metabolic reprogramming associated with immunophenotype, especially changes in glycolysis and glutamine catabolic pathways, which ultimately destroy the anti-tumor function of effector T cells and create a barrier to cancer treatment.
MM is a malignant plasma cell disease, and the antitumor ability of immune cells of the organism is insufficient, so enhancement of T cell-mediated antitumor immune response is currently the mainstream method of MM immunotherapy, such as antibody targeting therapy, chimeric antigen receptor (chimeric antigen receptor, CAR) T cells, etc. Such methods are of specific immunity, which is mediated or activated by a mechanism that stimulates immune activity, thereby enhancing the anti-myeloma effect to some extent. This idea is called immune potentiation therapy.
Although the above-described immune potentiation therapies have good therapeutic effects on MM, the risk of immune-related adverse events is increased, and toxic reactions limit clinical applications. To date, none of these drugs has a broad indication.
Disclosure of Invention
In the face of an enhancement of immune response, MM may be countered by an "immune escape" strategy. The immune monitoring mechanisms in MM are compromised, including: reduced normal antibody production, deregulation of T cell and NK cell proliferation and activation, interruption of antigen presentation processes, upregulation of immune checkpoints and immunosuppressive mediators. Meanwhile, some immunosuppressive molecules exist in the bone marrow microenvironment and can interact with normal immune cells. Thus, blocking the mechanism described above, restoring the anti-myeloma ability of immune cells has become a new strategy, known as "immune normalization". Studies have shown that immune normalization and normalization of bone marrow microenvironment gene expression are important prognostic factors in MM patients, except for MRD levels (fouread D, zhang Q, cog dill TD, wynn AS, steparwald NM, druhan LJ, guo F, rib K, madden KL, symanoski JT, avalos BR, copelan EA, usemani SZ, bhutani m.peripheral Immune Profile and Minimal Residual Disease (MRD) Burden Following Autologous Stem Cell Transplantation (ASCT) in Multiple Myeloma (MM) [ J ]. Blood,2016,128 (22)). This suggests that the bone marrow microenvironment and immune system are important targets for future therapies for MM.
The microenvironment of a variety of cancers, including MM, is characterized by nutrient deficiency, metabolite accumulation, acidosis, and hypoxia. In this harsh environment, T cells develop stress responses. Endoplasmic reticulum is the major organelle in cells responsible for secretory and transmembrane protein folding and assembly. When T cells are affected by BME, unfolded or misfolded proteins in the endoplasmic reticulum increase, and endoplasmic reticulum stress (endoplasmic reticulum stress, ERS) occurs, and T cells lose their ability to self-stabilize, posing a threat to cell survival and function. At the same time, ERS signals can be transmitted through endoplasmic reticulum transmembrane molecules into the nucleus, up-regulating the expression of a range of specific chaperones, and bind to unfolded or misfolded protein molecules for proper folding. ERS can also activate endoplasmic reticulum-related protein degradation pathways, transporting unfolded or misfolded proteins to the cytoplasm and degrading via the ubiquitin-proteinase system. In the ERS state, a series of biochemical reactions that occur by cells to restore homeostasis are called unfolded protein reactions (unfolded protein response, UPR).
There are 3 transmembrane proteins on the endoplasmic reticulum that transduce UPR signals: inositol 1 (inositol requiring enzyme, IRE 1), protein kinase receptor like endoplasmic reticulum kinase (PKR-like ER kinase, PERK), transcriptional activator-6 (activating transcription factor-6, ATF 6). These membrane proteins can sense the accumulation of misfolded proteins and become activated, initiating the complete transcription process. UPR is closely related to cell homeostasis and survival, and is involved in a variety of pathophysiological processes. So et al have found that the activation of the 3 signal pathways of the UPR varies depending on the intensity, duration and cell type of the stress source. For T cells in tumors, moderate UPR can enable unfolded or misfolded proteins to fold correctly under the action of chaperones or degrade through endoplasmic reticulum-related protein degradation pathways, thus relieving the damage to T cell survival; however, improper UPR can cause T cell dysfunction. The cube-Ruiz et al study found that in malignant patients, excessive UPR can lead to a suppression of T cell function and promote tumor progression. (So JS. Roles of Endoplasmic Reticulum Stress in Immune Responses [ J ]. Mol Cells,2018,41 (8): 705-716. Cubic-Ruiz JR, bettigel SE, glimcher LH.Tumorigenic and Immunosuppressive Effects of Endoplasmic Reticulum Stress in Cancer [ J ]. Cell,2017,168 (4): 692-706.)
Notably, UPR can also sense and integrate metabolic signals, regulate glucose, glutamine, fatty acid metabolism, and induce reprogramming of cellular metabolism. For example, activation of transcription factor 4 (activating transcription factor, ATF 4) in the PERK pathway induces gene expression of hexokinase, phosphoenolpyruvate carboxykinase, etc. involved in glucose metabolism. IRE1 pathway transcription factor X-box binding protein 1 (X-box binding factor protein 1, XBP 1) can bind to promoters regulating genes for glucose and fatty acid metabolism, mediating gene expression. (Van Der Harg JM, van Heest JC, bangel FN, patiwel S, van Weering JR, scheper W.the UPR reduces glucose metabolism via IRE1 signaling [ J ]. Biochim Biophys Acta Mol Cell Res,2017,1864 (4): 655-665.Hotamisligil GS.Endoplasmic reticulum stress and the inflammatory basis of metabolic disease[J ]. Cell,2010,140 (6): 900-917.Acosta-Alvear D, zhou Y, blais A, tsikitis M, lents NH, arias C, lennon CJ, kluger Y, dynlacht BD.XBP1 controls diverse Cell type-and condition-specific transcriptional regulatory networks [ J ]. Mol Cell,2007,27 (1): 53-66.)
It follows that metabolic reprogramming by poorly adapted UPR may be a key cause of immune dysfunction for T cells in the MM microenvironment.
The application aims to provide a method for recovering CD8 in MM tumor microenvironment + A method of glutamine metabolism in T lymphocytes to increase the anti-tumor immunity of the T cells.
In order to achieve the above purpose, the technical scheme of the application is as follows: bone marrow CD8 in MM patients + The T lymphocyte interferes with the X-box binding protein 1 (X-box binding factor protein 1, XBP 1) or over-expresses the solute carrier family 38member 2 (solute carrier family 38member 2,SLC38A2) gene or the protein encoded by the gene, thereby relieving the T cell immunity inhibition phenotype, enhancing the generation of effector molecules and improving the tumor killing effect of the T cell.
In a first aspect, the application provides the use of the SLC38A2 gene or a protein encoded thereby in the manufacture of a medicament for the treatment of multiple myeloma.
In a second aspect, the application provides the use of an agent that increases the expression of the SLC38A2 gene or a protein encoded thereby in the manufacture of a medicament for the treatment of multiple myeloma.
In a third aspect of the application, there is provided the use of an agent that interferes with XBP1 in the manufacture of a medicament for the treatment of multiple myeloma.
In a fourth aspect of the application, there is providedCD8 in preparation of reagent for interfering XBP1 or improving SLC38A2 gene or coded protein expression + Use of a drug for glutamine metabolism in T lymphocytes.
In a fifth aspect, the present application provides an agent that interferes with XBP1 or increases expression of the SLC38A2 gene or protein encoded thereby for use in the manufacture of a medicament for increasing T cell killing tumor; the tumor is multiple myeloma.
In a sixth aspect of the application, there is provided a method of restoring CD8 in a multiple myeloma tumor microenvironment + The method of glutamine metabolism in T lymphocyte is to make CD8 in bone marrow + The T lymphocytes interfere with XBP1 or over-express the SLC38A2 gene or its encoded protein.
The application has the advantages that:
1. the application provides a method, namely, bone marrow CD8 of MM patient + The T lymphocyte interferes XBP1 or overexpresses SLC38A2 gene or protein encoded by the gene, thereby relieving the immune suppression phenotype of the T cell, enhancing the generation of effector molecules and improving the tumor killing effect of the T cell.
2. The method for recovering the amino acid metabolism of the T cells is called an 'immune normalization' strategy, and targets the SLC38A2 gene, so that the expression of the SLC38A2 gene is increased to the level of a normal person, metabolic disturbance in the MM patient is corrected, the states of the depletion, aging and apoptosis of the T cells are changed, and the original functions of the T cells are recovered, thereby improving the anti-tumor effect.
3. The application focuses on the metabolic characteristics of T cells in the bone marrow microenvironment of the multiple myeloma, improves the immune suppression phenotype of the T cells through XBP1 and SLC38A2 targets from the immune normalization angle, and enhances the expression of effector molecules of the T cells, thereby being beneficial to the anti-myeloma immune effect of the T cells. Modulation of T cell metabolism will aid in the development of multiple myeloma immunotherapeutic strategies, benefiting patients.
Drawings
Fig. 1: identification of T cell subsets in MM patients and healthy control bone marrow and peripheral blood samples;
a.10 MM patients before and after treatment and 3 patientsDimensionality reduction clustering of 15 (C0-C14) T cell subsets in healthy control bone marrow and peripheral blood samples; colors represent subgroups. Violin map of marker gene expression in 15T cell subpopulations. C.T cell subsets are the proportion of healthy people, patients before and after treatment of bone marrow and peripheral blood sample sources. D. Proportion of each T cell subpopulation in bone marrow before and after treatment of healthy controls and patients; gray represents healthy control, yellow represents patient before treatment, and blue represents patient after treatment; with the use of a rank sum test, * P≤0.05, ** P≤0.01。
fig. 2: CTL subpopulations in bone marrow of MM patients display a senescent and depleted phenotype;
A.T cell subsets are mapped according to the correlation of gene expression; according to the Spearman correlation coefficient analysis, the color represents the correlation coefficient. Transcript maps of senescence-associated genes in bone marrow T cells of mm patients; color represents gene expression. C. Violin shows expression of senescence-associated genes in healthy controls, bone marrow T cell subpopulations before and after patient treatment; gray represents healthy control, yellow represents patient before treatment, and blue represents patient after treatment. D. Violin plots show expression of depletion related genes in healthy controls, and bone marrow T cell subsets before and after patient treatment. Transcript patterns of depletion related genes in bone marrow T cells of mm patients.
Fig. 3: metabolic reprogramming and upregulation of the UPR pathway occurs in the bone marrow CTL subpopulations of MM patients;
gsea showed glycolytic pathway, hypoxia pathway prior to MM patient treatment and healthy control bone marrow CD8 + The degree of enrichment in CTL cells (subpopulations C1, C3, C5, C11), P-values and calibration P-values are noted. Gsea shows the extent of enrichment of OXPHOS pathway in pre-MM patient treatment and healthy control bone marrow subpopulations C5, C12, C13; and the degree of enrichment of the OXPHOS pathway in the C11 and C1 subpopulations of bone marrow prior to MM patient treatment. Gsea shows the extent of enrichment of UPR pathway in pre-MM patient treatment and healthy control bone marrow C3, C5 subpopulations. D. Violin shows the expression of mitochondrial related genes and UPR pathway genes in a healthy control, bone marrow T cell subpopulation before and after patient treatment; gray represents healthy control, yellow represents patient before treatment, and blue represents patient after treatment. E. Box diagram displayExpression of XBP1 in healthy controls, bone marrow C1, C3, C5, C6, C11 subpopulations before and after patient treatment; with rank sum test, the P value is noted.
Fig. 4: CD8 + XBP1 in T cells can directly negatively regulate SLC38A2 and influence T cell functions;
A. violin plots show the expression of SLC38A2 in healthy controls, bone marrow T cell subsets before and after patient treatment; gray represents healthy control, yellow represents patient before treatment, and blue represents patient after treatment. B. Patient pre-treatment bone marrow CD8 + Correlation analysis of SLC38A2 expression in CTLs (subpopulations C1, C3, C5, C11) with senescence, depletion, apoptosis, UPR gene expression; red represents positive correlation and blue represents negative correlation. C. The heat map shows the differences in expression of SLC38A2, XBP1, aging, depletion, inflammatory factors in healthy controls, and in bone marrow samples before and after patient treatment; red represents high expression and blue represents low expression. D. Culturing normal human CD8 in a medium containing or lacking glucose + T cells, qPCR detected XBP1s and SLC38A2 mRNA expression (n=3, mean ± standard deviation). E. Luciferase reporter experiments detected the binding of transcription factor XBP1s to the SLC38A2 promoter (n=3, mean ± standard deviation). qPCR detects XBP1s and SLC38A2 mRNA levels. Interference or overexpression of XBP1 up-or down-regulates SLC38A2, respectively, in cd8+ T cells of healthy donors (interference experiments, n=4; overexpression experiments, n=3; mean ± standard deviation). G. CD8 in healthy donors + Interference or overexpression of XBP1 in T cells, detection of CD8 by flow cytometry + The duty cycle of the effector, senescent and depleting markers of T cells (n=4, median of the quartile range). H. CD8 in healthy donors + Overexpression of SLC38A2 in T cells, expression of SLC38A2 was detected by qPCR; flow cytometry detection of CD8 + The duty cycle of the effector markers of T cells (n=3, mean ± standard deviation). The t-test is adopted to carry out the test, * P≤0.05, ** P≤0.01, *** P≤0.001。
Detailed Description
The following provides a detailed description of embodiments of the present application with reference to examples.
Examples:
in the early work, the present application studied bone marrow and peripheral blood mononuclear cell samples of MM patients with healthy controls after initial and two-course VCD regimens (bortezomib, cyclophosphamide, dexamethasone) using scRNA-seq, identified 15T cell subsets, including 6 CD8 + T cells, 7 CD4 + T cells and 2 γδ T cells can be divided into 9 cytotoxic T cell (cytotoxic T lymphocytes, CTL) subpopulations and 6 non-cytotoxic T cell subpopulations according to their function (fig. 1). These T cells are respectively: CD8 + Teff cells, CD8 + Terminally differentiated Tem (terminally differentiated effector memory T, temra) cells, CD8 + GZMK + Tem cells, CD8 + Primary T cells, CD8 + Depletion of Tem cells, mucosa-associated unchanged T (mucosal-associated invariant T, MAIT) cells, CD4 + Primary T cells, CD4 + TCF7 + Central memory T (Tcm) cells, CD4 + ANXA1 + Tcm cells, CD4 + Teff cells, CD4 + GZMK + Tem cells, treg cells, CD4 + Interferon-responsive T (IFN-responsive T) cells, PRF1 + Gamma delta T cells, GZMK + γδ T cells. All CTL subpopulations in MM patient bone marrow generally exhibited a senescent phenotype, while some CTLs exhibited a characteristic of depletion (fig. 2).
The CTL subpopulation of MM patients showed alterations in upregulation of UPR, metabolic reprogramming, etc. signaling pathways (figure 3). The expression of the key transcription factor XBP1 in the UPR pathway is elevated in nearly all CTL subsets in patients. The glycolytic pathway of the CTL subpopulation was not significantly altered compared to the normal control, but the OXPHOS pathway was down-regulated in some subpopulations, the most significant difference being that all CTL subpopulations in the patient's bone marrow expressed little solute carrier family 38member 2 (solute carrier family 38member 2,SLC38A2), and these T cells had reduced mitochondrial function. Further bioinformatics analysis showed that in patient CD8 + In CTL, expression of SLC38A2 was inversely correlated with expression of senescence, depletion, apoptosis-related genes, XBP1 (fig. 4). The dual luciferase reporter gene showed that XBP1 could bind to the promoter of SLC38 A2.And in vitro experiments showed that in activated CD8 + The XBP1-shRNA plasmid interference XBP1 packaged by slow viruses in T cells can increase the expression of SLC38A2 and reduce the expression of KLRG 1; the expression of SLC38A2 and GZMA can be inhibited by adopting the XBP1-cDNA plasmid of lentivirus package to overexpress XBP 1; overexpression of SLC38A2 using the lentiviral-packaged SLC38A2-cDNA plasmid enhanced expression of CD45, perforin and GZMA. Suggesting that interference with XBP1 pathway or overexpression of SLC38A2 may improve CD8 + T cell senescence and enhanced effector molecule production.
XBP1 is a target gene downstream of IRE1 in the UPR signaling pathway, and activated IRE1 has endonuclease activity, splicing XBP1 to form 371aa of XBP1 splice protein (XBP 1 s). The unspliced XBP1 mRNA forms 267aa of protein (XBP 1 u). Studies have shown that only spliced XBP1s is transcriptionally active, binds to ERS elements into the nucleus and induces transcription and translation of activated UPR target genes. XBP1 can regulate and control the expression of a series of glucose, amino acid and lipid metabolism genes, and has profound effects on cell metabolism and functions.
The SLC38A2 gene encodes sodium ion dependent neutral amino acid transporter 2 (sodium-coupled neutral amino acid transporter, SNAT2). SNAT2 is primarily responsible for the transport of glutamine, methionine and alanine. And the amino acid transporter of the snap family is identified as a key mediator of glutamine uptake in T cells. The catabolism of glutamine can inhibit cell stress response, maintain the integrity of mitochondrial membranes and promote the survival of proliferation cells. In combination with the in vitro research result, XBP1 can directly act on a promoter of SLC38A2 gene to inhibit the expression of SNAT2, thereby regulating glutamine metabolism and inducing the immune damage of CTL in MM.
In conclusion, the severe microenvironment of MM induces ERS in bone marrow CTL, and the UPR pathway of T cells is activated to restore self-stability, wherein the expression of XBP1 can be directly inhibited to express SLC38A2, the uptake of CTL glutamine is limited, metabolic disorder is caused, T cell immunity is inhibited, and MM immune escape is mediated. The application provides a method, namely, bone marrow CD8 of MM patient + Interfering XBP1 or over-expressing SLC38A2 gene or encoded protein in T lymphocyte,thereby relieving the T cell immunity inhibition phenotype, enhancing the generation of effector molecules and improving the tumor killing effect of T cells.
While the preferred embodiments of the present application have been described in detail, the present application is not limited to the embodiments, and various equivalent modifications and substitutions can be made by those skilled in the art without departing from the spirit of the present application, and these equivalent modifications and substitutions are intended to be included in the scope of the present application as defined in the appended claims.

Claims (1)

1. shRNA interfering X-box binding protein 1 gene and preparation method of CD8 in recovery of multiple myeloma tumor microenvironment + The application of the T lymphocyte glutamine metabolism to the medicine for improving the effect of T cell killing tumor.
CN202210672945.2A 2022-06-15 2022-06-15 Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma Active CN115040663B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210672945.2A CN115040663B (en) 2022-06-15 2022-06-15 Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210672945.2A CN115040663B (en) 2022-06-15 2022-06-15 Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma

Publications (2)

Publication Number Publication Date
CN115040663A CN115040663A (en) 2022-09-13
CN115040663B true CN115040663B (en) 2023-12-12

Family

ID=83161368

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210672945.2A Active CN115040663B (en) 2022-06-15 2022-06-15 Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma

Country Status (1)

Country Link
CN (1) CN115040663B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015048331A1 (en) * 2013-09-25 2015-04-02 Cornell University Compounds for inducing anti-tumor immunity and methods thereof
CN105377288A (en) * 2012-11-05 2016-03-02 达纳-法伯癌症研究所股份有限公司 Xbp1, cd138, and cs1 peptides, pharmaceutical compositions that include the peptides, and methods of using such peptides and compositions
CN106420791A (en) * 2016-09-23 2017-02-22 中国人民解放军第二军医大学第二附属医院 Application of miR-145-3p in preparing medicines for preventing or treating multiple myeloma disease
WO2018115189A1 (en) * 2016-12-21 2018-06-28 Cellectis Stably enginereed proteasome inhibitor resistant immune cells for immunotherapy
CN114480645A (en) * 2022-01-13 2022-05-13 上海交通大学医学院附属仁济医院 Multiple myeloma exhausted NK cell subgroup, characteristic gene and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2300034B1 (en) * 2008-06-02 2018-08-08 Dana-Farber Cancer Institute, Inc. Pharmaceutical composition for use in treating or preventing multiple myeloma or waldenstrom's macroglobulinemia
WO2019084493A1 (en) * 2017-10-27 2019-05-02 The Trustees Of The University Of Pennsylvania Methods and compositions for treating diseases associated with exhausted t cells

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105377288A (en) * 2012-11-05 2016-03-02 达纳-法伯癌症研究所股份有限公司 Xbp1, cd138, and cs1 peptides, pharmaceutical compositions that include the peptides, and methods of using such peptides and compositions
WO2015048331A1 (en) * 2013-09-25 2015-04-02 Cornell University Compounds for inducing anti-tumor immunity and methods thereof
CN106420791A (en) * 2016-09-23 2017-02-22 中国人民解放军第二军医大学第二附属医院 Application of miR-145-3p in preparing medicines for preventing or treating multiple myeloma disease
WO2018115189A1 (en) * 2016-12-21 2018-06-28 Cellectis Stably enginereed proteasome inhibitor resistant immune cells for immunotherapy
CN114480645A (en) * 2022-01-13 2022-05-13 上海交通大学医学院附属仁济医院 Multiple myeloma exhausted NK cell subgroup, characteristic gene and application thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Deletion of Amino Acid Transporter ASCT2 (SLC1A5) Reveals an Essential Role for Transporters SNAT1 (SLC38A1) and SNAT2 (SLC38A2) to Sustain Glutaminolysis in Cancer Cells;Bröer A;J Biol Chem;13194-13205 *
Identification of novel myeloma-specific XBP1 peptides able to generate cytotoxic T lymphocytes: a potential therapeutic application in multiple myeloma;J Bae;Leukemia;1610–1619 *
IRE1α-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity;Song M;Nature;423-428 *
IRE1α-XBP1 signaling pathway, a potential therapeutic target in multiple myeloma;Chen L;Leuk Res;7-12 *
Pallett LJ. A glutamine 'tug-of-war': targets to manipulate glutamine metabolism for cancer immunotherapy.Immunother Adv.2021,第2页右栏第3段、第3页右栏第1段. *
Single-cell RNA sequencing reveals XBP1-SLC38A2 axis as a metabolic regulator in cytotoxic T lymphocytes in multiple myeloma;Wan Yike;Cancer Lett;1-14 *
中性氨基酸转运蛋白SNAT2拓扑结构初探;李洋;中国优秀硕士学位论文全文数据库;A006-117 *
氨基酸及其转运体对肿瘤细胞和T细胞作用的研究进展;张峥;肿瘤代谢与营养电子杂志;271-275 *

Also Published As

Publication number Publication date
CN115040663A (en) 2022-09-13

Similar Documents

Publication Publication Date Title
Kumagai et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments
Wang et al. The deubiquitinase USP22 regulates PD-L1 degradation in human cancer cells
Banta et al. Mechanistic convergence of the TIGIT and PD-1 inhibitory pathways necessitates co-blockade to optimize anti-tumor CD8+ T cell responses
Yarosz et al. The role of reactive oxygen species in regulating T cell-mediated immunity and disease
Cha et al. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1
Ma et al. Cholesterol induces CD8+ T cell exhaustion in the tumor microenvironment
Raninga et al. Therapeutic cooperation between auranofin, a thioredoxin reductase inhibitor and anti‐PD‐L1 antibody for treatment of triple‐negative breast cancer
Galore-Haskel et al. A novel immune resistance mechanism of melanoma cells controlled by the ADAR1 enzyme
EP2817619B1 (en) Prediction of responsiveness to treatment with immunomodulatory therapeutics and method of monitoring abscopal effects during such treatment
Paul et al. Coenzyme A fuels T cell anti-tumor immunity
Liu et al. The EGFR‐P38 MAPK axis up‐regulates PD‐L1 through miR‐675‐5p and down‐regulates HLA‐ABC via hexokinase‐2 in hepatocellular carcinoma cells
Pucci et al. Caveolin-1, breast cancer and ionizing radiation
Zhang et al. Potential function of CTLA‑4 in the tumourigenic capacity of melanoma stem cells
Wu et al. Role of kynurenine in promoting the generation of exhausted CD8+ T cells in colorectal cancer
Zhang et al. Amino acids and RagD potentiate mTORC1 activation in CD8+ T cells to confer antitumor immunity
CN115040663B (en) Application of solute carrier family 38member 2 in preparing medicament for treating multiple myeloma
WO2017025962A1 (en) Prediction of response to immunotherapy based on tumor biomarkers
Li et al. ACADL suppresses PD-L1 expression to prevent cancer immune evasion by targeting HIPPO/YAP signaling in lung adenocarcinoma
WO2019164870A1 (en) Expression of signature mrnas for identifying patients responsive to anti-pd-l1 antibody therapy
US9567394B2 (en) Anti-malignant tumor agent
Udumula et al. Intermittent Fasting induced ketogenesis inhibits mouse epithelial ovarian tumors by promoting anti-tumor T cell response
EP3356551B1 (en) Methods for determining the metabolic status of b-lymphomas
CN111773380A (en) Application of PLPP1 in preparation of T cell immune tumor related medicament
Schofield et al. Acod1 expression in cancer cells promotes immune evasion through the generation of inhibitory peptides
CN113134090B (en) Anti-tumor medicine composition

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
GR01 Patent grant
GR01 Patent grant