Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21.032
1.
Nat Commun ; 15(1): 3933, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730243

As a strategy to improve the therapeutic success of chimeric antigen receptor T cells (CART) directed against solid tumors, we here test the combinatorial use of CART and IMSA101, a newly developed stimulator of interferon genes (STING) agonist. In two syngeneic tumor models, improved overall survival is observed when mice are treated with intratumorally administered IMSA101 in addition to intravenous CART infusion. Transcriptomic analyses of CART isolated from tumors show elevated T cell activation, as well as upregulated cytokine pathway signatures, in particular IL-18, in the combination treatment group. Also, higher levels of IL-18 in serum and tumor are detected with IMSA101 treatment. Consistent with this, the use of IL-18 receptor negative CART impair anti-tumor responses in mice receiving combination treatment. In summary, we find that IMSA101 enhances CART function which is facilitated through STING agonist-induced IL-18 secretion.


Interleukin-18 , Membrane Proteins , Receptors, Chimeric Antigen , Animals , Interleukin-18/metabolism , Membrane Proteins/agonists , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Humans , Cell Line, Tumor , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Lymphocyte Activation/drug effects , Immunotherapy, Adoptive/methods , Female , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy
2.
Methods Cell Biol ; 186: 131-150, 2024.
Article En | MEDLINE | ID: mdl-38705597

Hypomethylating therapies using decitabine or azacitidine are actively investigated to treat acute myeloid leukemia, myelodysplastic syndromes, as maintenance therapy after allogenic stem cell transplant and hemoglobinopathies. The therapeutic mechanism is to de-repress genes that have been turned off through oncogenesis or development via methylation. The therapy can be non-cytotoxic at low dosage, sparing healthy stem cells and operating on committed precursors. Because the methods of determining maximum tolerated dose are not well suited to this paradigm, and because the mechanism of action, which is depletion of DNA methylase 1 (DNMT1), is complex and dependent on passing through a cell cycle, a pharmacodynamic assay that measures DNMT1 can inform clinical trials aimed at establishing and improving therapy. Herein, we provide an assay that measures DNMT1 relative levels in circulating T cells of peripheral blood.


Azacitidine , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Decitabine , Azacitidine/pharmacology , Humans , Decitabine/pharmacology , DNA Methylation/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism
3.
PLoS One ; 19(5): e0300174, 2024.
Article En | MEDLINE | ID: mdl-38696390

Off-the-shelf immunotherapeutics that suppress tumor growth and provide durable protection against relapse could enhance cancer treatment. We report preclinical studies on a CD33 x CD3 bivalent bispecific diabody, AMV564, that not only suppresses tumor growth, but also facilitates memory responses in a mouse model of acute myelogenous leukemia (AML). Mechanistically, a single 5-day treatment with AMV564 seems to reduce tumor burden by redirection of T cells, providing a time window for allogeneic or other T cells that innately recognize tumor antigens to become activated and proliferate. When the concentration of bispecific becomes negligible, the effector: target ratio has also shifted, and these activated T cells mediate long-term tumor control. To test the efficacy of AMV564 in vivo, we generated a CD33+ MOLM13CG bioluminescent human cell line and optimized conditions needed to control these cells for 62 days in vivo in NSG mice. Of note, not only did MOLM13CG become undetectable by bioluminescence imaging in response to infusion of human T cells plus AMV564, but also NSG mice that had cleared the tumor also resisted rechallenge with MOLM13CG in spite of no additional AMV564 treatment. In these mice, we identified effector and effector memory human CD4+ and CD8+ T cells in the peripheral blood immediately prior to rechallenge that expanded significantly during the subsequent 18 days. In addition to the anti-tumor effects of AMV564 on the clearance of MOLM13CG cells in vivo, similar effects were seen when primary CD33+ human AML cells were engrafted in NSG mice even when the human T cells made up only 2% of the peripheral blood cells and AML cells made up 98%. These studies suggest that AMV564 is a novel and effective bispecific diabody for the targeting of CD33+ AML that may provide long-term survival advantages in the clinic.


Antibodies, Bispecific , CD3 Complex , Immunologic Memory , Leukemia, Myeloid, Acute , Sialic Acid Binding Ig-like Lectin 3 , Animals , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/drug therapy , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Mice , CD3 Complex/immunology , Immunologic Memory/drug effects , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/drug effects
5.
Front Immunol ; 15: 1388962, 2024.
Article En | MEDLINE | ID: mdl-38720895

Introduction: Chronic inflammation is a hallmark of chronic wounds and inflammatory skin diseases. Due to a hyperactive and prolonged inflammation triggered by proinflammatory immune cells, transitioning to the repair and healing phase is halted. T cells may exacerbate the proinflammatory milieu by secreting proinflammatory cytokines. Chamomilla recutita L. (chamomile) has been suggested for use in several inflammatory diseases, implying a capability to modulate T cells. Here, we have characterized and compared the effects of differently prepared chamomile extracts and characteristic pure compounds on the T cell redox milieu as well as on the migration, activation, proliferation, and cytokine production of primary human T cells. Methods: Phytochemical analysis of the extracts was carried out by LC-MS/MS. Primary human T cells from peripheral blood (PBTs) were pretreated with aqueous or hydroethanolic chamomile extracts or pure compounds. Subsequently, the effects on intracellular ROS levels, SDF-1α induced T cell migration, T cell activation, proliferation, and cytokine production after TCR/CD3 and CD28 costimulation were determined. Gene expression profiling was performed using nCounter analysis, followed by ingenuity pathway analysis, and validation at protein levels. Results: The tested chamomile extracts and pure compounds differentially affected intracellular ROS levels, migration, and activation of T cells. Three out of five differently prepared extracts and two out of three pure compounds diminished T cell proliferation. In line with these findings, LC-MS/MS analysis revealed high heterogeneity of phytochemicals among the different extracts. nCounter based gene expression profiling identified several genes related to T cell functions associated with activation and differentiation to be downregulated. Most prominently, apigenin significantly reduced granzyme B induction and cytotoxic T cell activity. Conclusion: Our results demonstrate an anti-inflammatory effect of chamomile- derived products on primary human T cells. These findings provide molecular explanations for the observed anti-inflammatory action of chamomile and imply a broader use of chamomile extracts in T cell driven chronic inflammatory diseases such as chronic wounds and inflammatory skin diseases. Importantly, the mode of extract preparation needs to be considered as the resulting different phytochemicals can result in differential effects on T cells.


Anti-Inflammatory Agents , Cytokines , Flowers , Lymphocyte Activation , Matricaria , Plant Extracts , T-Lymphocytes , Humans , Plant Extracts/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Matricaria/chemistry , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Flowers/chemistry , Lymphocyte Activation/drug effects , Plant Roots/chemistry , Cells, Cultured , Cell Proliferation/drug effects , Cell Movement/drug effects
6.
Nat Commun ; 15(1): 3664, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38693123

The application of mammalian target of rapamycin inhibition (mTORi) as primary prophylactic therapy to optimize T cell effector function while preserving allograft tolerance remains challenging. Here, we present a comprehensive two-step therapeutic approach in a male patient with metastatic cutaneous squamous cell carcinoma and heart transplantation followed with concomitant longitudinal analysis of systemic immunologic changes. In the first step, calcineurin inhibitor/ mycophenolic acid is replaced by the mTORi everolimus to achieve an improved effector T cell status with increased cytotoxic activity (perforin, granzyme), enhanced proliferation (Ki67) and upregulated activation markers (CD38, CD69). In the second step, talimogene laherparepvec (T-VEC) injection further enhances effector function by switching CD4 and CD8 cells from central memory to effector memory profiles, enhancing Th1 responses, and boosting cytotoxic and proliferative activities. In addition, cytokine release (IL-6, IL-18, sCD25, CCL-2, CCL-4) is enhanced and the frequency of circulating regulatory T cells is increased. Notably, no histologic signs of allograft rejection are observed in consecutive end-myocardial biopsies. These findings provide valuable insights into the dynamics of T cell activation and differentiation and suggest that timely initiation of mTORi-based primary prophylaxis may provide a dual benefit of revitalizing T cell function while maintaining allograft tolerance.


Carcinoma, Squamous Cell , Graft Rejection , Heart Transplantation , Herpesvirus 1, Human , MTOR Inhibitors , Heart Transplantation/adverse effects , Humans , Male , Graft Rejection/prevention & control , Graft Rejection/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/drug therapy , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Biological Products/pharmacology , Biological Products/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Middle Aged , Everolimus/pharmacology , Everolimus/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
7.
Lancet Oncol ; 25(5): e205-e216, 2024 May.
Article En | MEDLINE | ID: mdl-38697166

Multiple myeloma remains an incurable disease, despite the development of numerous drug classes and combinations that have contributed to improved overall survival. Immunotherapies directed against cancer cell-surface antigens, such as chimeric antigen receptor (CAR) T-cell therapy and T-cell-redirecting bispecific antibodies, have recently received regulatory approvals and shown unprecedented efficacy. However, these immunotherapies have unique mechanisms of action and toxicities that are different to previous treatments for myeloma, so experiences from clinical trials and early access programmes are essential for providing specific recommendations for management of patients, especially as these agents become available across many parts of the world. Here, we provide expert consensus clinical practice guidelines for the use of bispecific antibodies for the treatment of myeloma. The International Myeloma Working Group is also involved in the collection of prospective real-time data of patients treated with such immunotherapies, with the aim of learning continuously and adapting clinical practices to optimise the management of patients receiving immunotherapies.


Antibodies, Bispecific , Consensus , Multiple Myeloma , T-Lymphocytes , Humans , Antibodies, Bispecific/therapeutic use , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Multiple Myeloma/drug therapy , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Immunotherapy/methods , Immunotherapy/standards , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/adverse effects
8.
Nutrients ; 16(9)2024 Apr 27.
Article En | MEDLINE | ID: mdl-38732558

Polyunsaturated fatty acids (PUFAs) can alter adipose tissue function; however, the relative effects of plant and marine n3-PUFAs are less clear. Our objective was to directly compare the n3-PUFAs, plant-based α-linolenic acid (ALA) in flaxseed oil, and marine-based eicosapentaenoic acid (EPA) or docosahexaenoic acid (DHA) in high-purity oils versus n6-PUFA containing linoleic acid (LA) for their effects on the adipose tissue and oral glucose tolerance of obese rats. Male fa/fa Zucker rats were assigned to faALA, faEPA, faDHA, and faLA groups and compared to baseline fa/fa rats (faBASE) and lean Zucker rats (lnLA). After 8 weeks, faEPA and faDHA had 11-14% lower body weight than faLA. The oral glucose tolerance and total body fat were unchanged, but faEPA had less mesenteric fat. faEPA and faDHA had fewer large adipocytes compared to faLA and faALA. EPA reduced macrophages in the adipose tissue of fa/fa rats compared to ALA and DHA, while faLA had the greatest macrophage infiltration. DHA decreased (~10-fold) T-cell infiltration compared to faBASE and faEPA, whereas faALA and faLA had an ~40% increase. The n3-PUFA diets attenuated tumour necrosis factor-α in adipose tissue compared to faBASE, while it was increased by LA in both genotypes. In conclusion, EPA and DHA target different aspects of inflammation in adipose tissue.


Adipose Tissue , Docosahexaenoic Acids , Eicosapentaenoic Acid , Macrophages , Obesity , Rats, Zucker , Animals , Eicosapentaenoic Acid/pharmacology , Docosahexaenoic Acids/pharmacology , Obesity/metabolism , Male , Macrophages/metabolism , Macrophages/drug effects , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Rats , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , alpha-Linolenic Acid/pharmacology , Mesentery
9.
Int Immunopharmacol ; 133: 112130, 2024 May 30.
Article En | MEDLINE | ID: mdl-38648712

Neutrophils and T lymphocytes are closely related to occurrence of immunosuppression in sepsis. Studies have shown that neutrophil apoptosis decreases and T lymphocyte apoptosis increases in sepsis immunosuppression, but the specific mechanism involved remains unclear. In the present study, we found Toll-like Receptor 2 (TLR2) and programmed death-ligand 1 (PD-L1) were significantly activated in bone marrow neutrophils of wild-type mice after LPS treatment and that they were attenuated by treatment with C29, an inhibitor of TLR2. PD-L1 activation inhibits neutrophil apoptosis, whereas programmed death protein 1 (PD-1)activation promotes apoptosis of T lymphocytes, which leads to immunosuppression. Mechanistically, when sepsis occurs, pro-inflammatory factors and High mobility group box-1 protein (HMGB1) passively released from dead cells cause the up-regulation of PD-L1 through TLR2 on neutrophils. The binding of PD-L1 and PD-1 on T lymphocytes leads to increased apoptosis of T lymphocytes and immune dysfunction, eventually resulting in the occurrence of sepsis immunosuppression. In vivo experiments showed that the HMGB1 inhibitor glycyrrhizic acid (GA) and the TLR2 inhibitor C29 could inhibit the HMGB1/TLR2/PD-L1 pathway, and improving sepsis-induced lung injury. In summary, this study shows that HMGB1 regulates PD-L1 and PD-1 signaling pathways through TLR2, which leads to immunosuppression.


Apoptosis , B7-H1 Antigen , HMGB1 Protein , Mice, Inbred C57BL , Neutrophils , Sepsis , T-Lymphocytes , Toll-Like Receptor 2 , Animals , Toll-Like Receptor 2/metabolism , HMGB1 Protein/metabolism , Sepsis/immunology , Sepsis/metabolism , B7-H1 Antigen/metabolism , Apoptosis/drug effects , Neutrophils/immunology , Neutrophils/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Mice , Male , Immune Tolerance , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/therapeutic use , Lipopolysaccharides/immunology , Signal Transduction , Immunosuppression Therapy
10.
Biochem Biophys Res Commun ; 715: 149995, 2024 Jun 30.
Article En | MEDLINE | ID: mdl-38685185

Immune checkpoint inhibitors have significantly transformed the landscape of cancer therapy. Nevertheless, while these inhibitors are highly effective for certain patient groups, many do not benefit due to primary or acquired resistance. Specifically, these treatments often lack sufficient therapeutic efficacy against cancers with low antigenicity. Thus, the development of an effective strategy to overcome cancers with low antigenicity is imperative for advancing next-generation cancer immunotherapy. Here, we show that small molecule inhibitor of hematopoietic progenitor kinase 1 (HPK1) combined with programmed cell death ligand 1 (PD-L1) blockade can enhance T-cell response to tumor with low antigenicity. We found that treatment of OT-1 splenocytes with HPK1 inhibitor enhanced the activation of signaling molecules downstream of T-cell receptor provoked by low-affinity-antigen stimulation. Using an in vivo OT-1 T-cell transfer model, we demonstrated that combining the HPK1 inhibitor with the anti-PD-L1 antibody significantly suppressed the growth of tumors expressing low-affinity altered peptide ligand of chicken ovalbumin, while anti-PD-L1 antibody monotherapy was ineffective. Our findings offer crucial insights into the potential for overcoming tumors with low antigenicity by combining conventional immune checkpoint inhibitors with HPK1 inhibitor.


B7-H1 Antigen , Mice, Inbred C57BL , Protein Serine-Threonine Kinases , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Mice , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Immunotherapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Humans , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology , 3-Phosphoinositide-Dependent Protein Kinases
11.
Eur J Pharmacol ; 973: 176562, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38588767

In recent years, immunosuppressants have shown significant success in the treatment of autoimmune diseases. Therefore, there is an urgent need to develop additional immunosuppressants that offer more options for patients. Toosendanin has been shown to have immunosuppressive activity in vitro as well as effects on autoimmune hepatitis (AIH) in vivo. Toosendanin did not induce apoptosis in activated T-cells and affect the survival rate of naive T-cells. Toosendanin did not affect the expression of CD25 or secretion of IL-2 by activated T-cells, and not affect the expression of IL-4 and INF-γ. Toosendanin did not affect the phosphorylation of STAT5, ERK, AKT, P70S6K. However, toosendanin inhibited proliferation of anti-CD3/anti-CD28 mAbs-activated T-cells with IC50 of (10 ± 2.02) nM. Toosendanin arrested the cell cycle in the G0/G1 phase, significantly inhibited IL-6 and IL-17A secretion, promoted IL-10 expression, and inhibited the P38 MAPK pathway. Finally, toosendanin significantly alleviated ConA-induced AIH in mice. In Summary, toosendanin exhibited immunosuppressive activity in vivo and in vitro. Toosendanin inhibits the proliferation of activated T-cells through the P38 MAPK signalling pathway, significantly suppresses the expression of inflammatory factors, enhances the expression of anti-inflammatory factors, and effectively alleviates ConA-induced AIH in mice, suggesting that toosendanin may be a lead compound for the development of novel immunomodulatory agents with improved efficacy and reduced toxicity.


Cell Proliferation , Drugs, Chinese Herbal , T-Lymphocytes , Triterpenes , p38 Mitogen-Activated Protein Kinases , Animals , Cell Proliferation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Drugs, Chinese Herbal/pharmacology , MAP Kinase Signaling System/drug effects , Lymphocyte Activation/drug effects , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Cytokines/metabolism , Immunosuppressive Agents/pharmacology , Mice, Inbred BALB C , Female
12.
Drug Des Devel Ther ; 18: 1247-1262, 2024.
Article En | MEDLINE | ID: mdl-38645988

Purpose: Sinomenine hydrochloride (SH) is used to treat chronic inflammatory diseases such as rheumatoid arthritis and may also be efficacious against Immunoglobulin A nephropathy (IgAN). However, no trial has investigated the molecular mechanism of SH on IgAN. Therefore, this study aims to investigate the effect and mechanism of SH on IgAN. Methods: The pathological changes and IgA and C3 depositions in the kidney of an IgAN rat model were detected by periodic acid-Schiff (PAS) and direct immunofluorescence staining. After extracting T and B cells using immunomagnetic beads, we assessed their purity, cell cycle phase, and apoptosis stage through flow cytometry. Furthermore, we quantified cell cycle-related and apoptosis-associated proteins by Western blotting. Results: SH reduced IgA and C3 depositions in stage 4 IgAN, thereby decreasing inflammatory cellular infiltration and mesangial injury in an IgAN model induced using heteroproteins. Furthermore, SH arrested the cell cycle of lymphocytes T and B from the spleen of IgAN rats. Regarding the mechanism, our results demonstrated that SH regulated the Cyclin D1 and Cyclin E1 protein levels for arresting the cell cycle and it also regulated Bax and Bcl-2 protein levels, thus increasing Cleaved caspase-3 protein levels in Jurkat T and Ramos B cells. Conclusion: SH exerts a dual regulation on the cell cycle and apoptosis of T and B cells by controlling cell cycle-related and apoptosis-associated proteins; it also reduces inflammatory cellular infiltration and mesangial proliferation. These are the major mechanisms of SH in IgAN.


Apoptosis , B-Lymphocytes , Cell Proliferation , Glomerulonephritis, IGA , Morphinans , T-Lymphocytes , Morphinans/pharmacology , Morphinans/chemistry , Glomerulonephritis, IGA/drug therapy , Glomerulonephritis, IGA/pathology , Animals , Apoptosis/drug effects , Rats , Cell Proliferation/drug effects , B-Lymphocytes/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Male , Dose-Response Relationship, Drug , Disease Models, Animal , Rats, Sprague-Dawley , Structure-Activity Relationship , Protective Agents/pharmacology , Protective Agents/chemistry , Humans , Cells, Cultured
13.
Eur J Pharmacol ; 972: 176565, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38599309

Blockade of PD-1/PD-L1 immune checkpoint is wildly used for multiple types of cancer treatment, while the low response rate for patients is still completely unknown. As nuclear hormone receptor, PPARδ (peroxisome-proliferator-activated receptor) regulates cell proliferation, inflammation, and tumor progression, while the effect of PPARδ on tumor immune escape is still unclear. Here we found that PPARδ antagonist GSK0660 significantly reduced colon cancer cell PD-L1 protein and gene expression. Luciferase analysis showed that GSK0660 decreased PD-L1 gene transcription activity. Moreover, reduced PD-L1 expression in colon cancer cells led to increased T cell activity. Further analysis showed that GSK0660 decreased PD-L1 expression in a PPARδ dependent manner. Implanted tumor model analysis showed that GSK0660 inhibited tumor immune escape and the combined PD-1 antibody with GSK0660 effectively enhanced colorectal cancer immunotherapy. These findings suggest that GSK0660 treatment could be an effective strategy for cancer immunotherapy.


B7-H1 Antigen , Immunotherapy , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Humans , Animals , Immunotherapy/methods , Mice , Cell Line, Tumor , PPAR delta/genetics , PPAR delta/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Colonic Neoplasms/genetics , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Escape/drug effects , Mice, Inbred BALB C
14.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38673976

Antagonist peptides (ANTs) of vasoactive intestinal polypeptide receptors (VIP-Rs) are shown to enhance T cell activation and proliferation in vitro, as well as improving T cell-dependent anti-tumor response in acute myeloid leukemia (AML) murine models. However, peptide therapeutics often suffer from poor metabolic stability and exhibit a short half-life/fast elimination in vivo. In this study, we describe efforts to enhance the drug properties of ANTs via chemical modifications. The lead antagonist (ANT308) is derivatized with the following modifications: N-terminus acetylation, peptide stapling, and PEGylation. Acetylated ANT308 exhibits diminished T cell activation in vitro, indicating that N-terminus conservation is critical for antagonist activity. The replacement of residues 13 and 17 with cysteine to accommodate a chemical staple results in diminished survival using the modified peptide to treat mice with AML. However, the incorporation of the constraint increases survival and reduces tumor burden relative to its unstapled counterpart. Notably, PEGylation has a significant positive effect, with fewer doses of PEGylated ANT308 needed to achieve comparable overall survival and tumor burden in leukemic mice dosed with the parenteral ANT308 peptide, suggesting that polyethylene glycol (PEG) incorporation enhances longevity, and thus the antagonist activity of ANT308.


Leukemia, Myeloid, Acute , Receptors, Vasoactive Intestinal Peptide , Animals , Mice , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors , Humans , Peptides/chemistry , Peptides/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cell Line, Tumor
15.
Phytomedicine ; 128: 155497, 2024 Jun.
Article En | MEDLINE | ID: mdl-38640855

BACKGROUND: Colorectal cancer (CRC) is a significant public health issue, ranking as one of the predominant cancer types globally in terms of incidence. Intriguingly, Arenobufagin (Are), a compound extracted from toad venom, has demonstrated the potential to inhibit tumor growth effectively. PURPOSE: This study aimed to explore Are's molecular targets and unravel its antitumor mechanism in CRC. Specifically, we were interested in its impact on immune checkpoint modulation and correlations with HSP90ß-STAT3-PD-L1 axis activity. METHODS: We investigated the in vivo antitumor effects of Are by constructing a colorectalcancer subcutaneous xenograft mouse model. Subsequently, we employed single-cell multi-omics technology to study the potential mechanism by which Are inhibits CRC. Utilizing target-responsive accessibility profiling (TRAP) technology, we identified heatshock protein 90ß (HSP90ß) as the direct target of Are, and confirmed this through a microscale thermophoresis experiment (MST). Further downstream mechanisms were explored through techniques such as co-immunoprecipitation, Western blotting, qPCR, and immunofluorescence. Concurrently, we arrived at the same research conclusion at the organoid level by co-cultivating with immune cells. RESULTS: We observed that Are inhibits PD-Ll expression in CRC tumor xenografts at low concentrations. Moreover, TRAP revealed that HSP90ß's accessibility significantly decreased upon Are binding. We demonstrated a decrease in the activity of the HSP90ß-STAT3-PD-Ll axis following low-concentration Are treatment in vivo. The PDO analysis showed improved enrichment of lymphocytes, particularly T cells, on the PDOs following Are treatment. CONCLUSION: Contrary to previous research focusing on the direct cytotoxicity of Are towards tumor cells, our findings indicate that it can also inhibit tumor growth at lower concentrations through the modulation of immune checkpoints. This study unveils a novel anti-tumor mechanism of Are and stimulates contemplation on the dose-response relationship of natural products, which is beneficial for the clinical translational application of Are.


Bufanolides , Colorectal Neoplasms , HSP90 Heat-Shock Proteins , STAT3 Transcription Factor , Xenograft Model Antitumor Assays , Bufanolides/pharmacology , Animals , Colorectal Neoplasms/drug therapy , Humans , Mice , STAT3 Transcription Factor/metabolism , T-Lymphocytes/drug effects , Cell Line, Tumor , B7-H1 Antigen , Mice, Nude , Mice, Inbred BALB C , Amphibian Venoms/pharmacology , Female
16.
Biomed Pharmacother ; 174: 116565, 2024 May.
Article En | MEDLINE | ID: mdl-38603888

Neural cell adhesion molecule L1 (L1CAM) is a cell-surface glycoprotein involved in cancer occurrence and migration. Up to today, L1CAM-targeted therapy appeared limited efficacy in clinical trials although quite a few attempts by monoclonal antibody (mAb) or chimeric antigen receptor T-cell therapy (CAR-T) have been reported. Therefore, the development of new effective therapies targeting L1CAM is highly desirable. It has been demonstrated that T cell-engaging bispecific antibody (TCE) plays an effective role in cancer immunotherapy by redirecting the cytotoxic activity of CD3+ T cells to tumor cells, resulting in tumor cell death. In this study, we designed and characterized a novel bispecific antibody (CE7-TCE) based on the IgG-(L)-ScFv format, which targets L1CAM and CD3 simultaneously. In vitro, CE7-TCE induced specific killing of L1CAM-positive tumor cells through T cells. In vivo, CE7-TCE inhibited tumor growth in human peripheral blood mononuclear cell/tumor cell co-grafting models. To overcome the adaptive immune resistance (AIR) that impairs the efficacy of TCEs, we conducted a combination therapy of CE7-TCE with Pembrolizumab (anti-PD1 mAb), which enhanced the anti-tumor activity of CE7-TCE. Our results confirmed the feasibility of using L1CAM as a TCE target for the treatment of solid tumors and revealed the therapeutic potential of CE7-TCE combined with immune checkpoint inhibitors.


Antibodies, Bispecific , Neural Cell Adhesion Molecule L1 , T-Lymphocytes , Animals , Female , Humans , Mice , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Antineoplastic Agents, Immunological/pharmacology , CD3 Complex/immunology , Cell Line, Tumor , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Neural Cell Adhesion Molecule L1/immunology , Neural Cell Adhesion Molecule L1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
17.
Bioconjug Chem ; 35(5): 593-603, 2024 May 15.
Article En | MEDLINE | ID: mdl-38592684

Ferritin is a multivalent, self-assembling protein scaffold found in most human cell types, in addition to being present in invertebrates, higher plants, fungi, and bacteria, that offers an attractive alternative to polymer-based drug delivery systems (DDS). In this study, the utility of the ferritin cage as a DDS was demonstrated within the context of T cell agonism for tumor killing. Members of the tumor necrosis factor receptor superfamily (TNFRSF) are attractive targets for the development of anticancer therapeutics. These receptors are endogenously activated by trimeric ligands that occur in transmembrane or soluble forms, and oligomerization and cell-surface anchoring have been shown to be essential aspects of the targeted agonism of this receptor class. Here, we demonstrated that the ferritin cage could be easily tailored for multivalent display of anti-OX40 antibody fragments on its surface and determined that these arrays are capable of pathway activation through cell-surface clustering. Together, these results confirm the utility, versatility, and developability of ferritin as a DDS.


Ferritins , Humans , Ferritins/chemistry , Ferritins/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Drug Delivery Systems
18.
Int Immunopharmacol ; 133: 112069, 2024 May 30.
Article En | MEDLINE | ID: mdl-38643710

Epigallocatechin-3-gallate (EGCG) is an important tea polyphenol with anti-tumor potential. Our previous studies revealed that EGCG was a promising immune checkpoint inhibitor (ICI) as it could downregulate expression of programmed cell death 1 ligand 1 (PD-L1) in tumor cells, thereby resulting tumor killing effect. In particular, EGCG can effectively avoid the inflammatory storm caused by anti-tumor therapy, which is a healthy green capacity absent from many ICIs. However, the relationship between EGCG and programmed cell death 1 (PD-1) of T cells remains unclear. In this work, we explored the effect of EGCG on T cells and found that EGCG suppressed PD-1 via inhibiting NF-κB phosphorylation and nuclear translocation. Furtherly, the capability of EGCG was confirmed in tumor-bearing mice to inhibit PD-1 expression in T cells and enhance apoptosis in tumor cells. These results implied that EGCG could inhibit the expression of PD-1 in T cells, thereby promoting anti-tumor effects of T cells. EGCG will be a promising candidate in anti-tumor therapy.


Catechin , NF-kappa B , Programmed Cell Death 1 Receptor , T-Lymphocytes , Catechin/analogs & derivatives , Catechin/pharmacology , Animals , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice , Humans , Apoptosis/drug effects , Cell Line, Tumor , Mice, Inbred C57BL , Female , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Active Transport, Cell Nucleus/drug effects
19.
Transpl Int ; 37: 12720, 2024.
Article En | MEDLINE | ID: mdl-38655204

Infectious complications, including widespread human cytomegalovirus (CMV) disease, frequently occur after hematopoietic stem cell and solid organ transplantation due to immunosuppressive treatment causing impairment of T-cell immunity. Therefore, in-depth analysis of the impact of immunosuppressants on antiviral T cells is needed. We analyzed the impact of mTOR inhibitors sirolimus (SIR/S) and everolimus (EVR/E), calcineurin inhibitor tacrolimus (TAC/T), purine synthesis inhibitor mycophenolic acid (MPA/M), glucocorticoid prednisolone (PRE/P) and common double (T+S/E/M/P) and triple (T+S/E/M+P) combinations on antiviral T-cell functionality. T-cell activation and effector molecule production upon antigenic stimulation was impaired in presence of T+P and triple combinations. SIR, EVR and MPA exclusively inhibited T-cell proliferation, TAC inhibited activation and cytokine production and PRE inhibited various aspects of T-cell functionality including cytotoxicity. This was reflected in an in vitro infection model, where elimination of CMV-infected human fibroblasts by CMV-specific T cells was reduced in presence of PRE and all triple combinations. CMV-specific memory T cells were inhibited by TAC and PRE, which was also reflected with double (T+P) and triple combinations. EBV- and SARS-CoV-2-specific T cells were similarly affected. These results highlight the need to optimize immune monitoring to identify patients who may benefit from individually tailored immunosuppression.


Cytomegalovirus Infections , Cytomegalovirus , Everolimus , Immunosuppressive Agents , Mycophenolic Acid , Sirolimus , T-Lymphocytes , Tacrolimus , Humans , Cytomegalovirus Infections/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Cytomegalovirus/immunology , Sirolimus/pharmacology , Sirolimus/therapeutic use , Lymphocyte Activation/drug effects , Prednisolone/therapeutic use , Organ Transplantation , Cell Proliferation/drug effects
20.
Pharmacol Res ; 203: 107158, 2024 May.
Article En | MEDLINE | ID: mdl-38599467

Cancer treatment is one of the fundamental challenges in clinical setting, especially in relapsed/refractory malignancies. The novel immunotherapy-based treatments bring new hope in cancer therapy and achieve various treatment successes. One of the distinguished ways of cancer immunotherapy is adoptive cell therapy, which utilizes genetically modified immune cells against cancer cells. Between different methods in ACT, the chimeric antigen receptor T cells have more investigation and introduced a promising way to treat cancer patients. This technology progressed until it introduced six US Food and Drug Administration-approved CAR T cell-based drugs. These drugs act against hematological malignancies appropriately and achieve exciting results, so they have been utilized widely in cell therapy clinics. In this review, we introduce all CAR T cells-approved drugs based on their last data and investigate them from all aspects of pharmacology, side effects, and compressional. Also, the efficacy of drugs, pre- and post-treatment steps, and expected side effects are introduced, and the challenges and new solutions in CAR T cell therapy are in the last speech.


Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Animals , Receptors, Chimeric Antigen/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Receptors, Antigen, T-Cell/immunology
...