Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell Commun Signal ; 22(1): 173, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38462636

ABSTRACT

BACKGROUND: Targeting the tumor microenvironment (TME) has emerged as a promising strategy in cancer treatment, particularly through the utilization of immune checkpoint blockade (ICB) agents such as PD-1/PD-L1 inhibitors. Despite partial success, the presence of tumor-associated macrophages (TAMs) contributes to an immunosuppressive TME that fosters tumor progression, and diminishes the therapeutic efficacy of ICB. Blockade of the CD47/SIRPα pathway has proven to be an effective intervention, that restores macrophage phagocytosis and yields substantial antitumor effects, especially when combined with PD-1/PD-L1 blockade. Therefore, the identification of small molecules capable of simultaneously blocking CD47/SIRPα and PD-1/PD-L1 interactions has remained imperative. METHODS: SMC18, a small molecule with the capacity of targeting both SIRPα and PD-L1 was obtained using MST. The efficiency of SMC18 in interrupting CD47/SIRPα and PD-1/PD-L1 interactions was tested by the blocking assay. The function of SMC18 in enhancing the activity of macrophages and T cells was tested using phagocytosis assay and co-culture assay. The antitumor effects and mechanisms of SMC18 were investigated in the MC38-bearing mouse model. RESULTS: SMC18, a small molecule that dual-targets both SIRPα and PD-L1 protein, was identified. SMC18 effectively blocked CD47/SIRPα interaction, thereby restoring macrophage phagocytosis, and disrupted PD-1/PD-L1 interactions, thus activating Jurkat cells, as evidenced by increased secretion of IL-2. SMC18 demonstrated substantial inhibition of MC38 tumor growths through promoting the infiltration of CD8+ T and M1-type macrophages into tumor sites, while also priming the function of CD8+ T cells and macrophages. Moreover, SMC18 in combination with radiotherapy (RT) further improved the therapeutic efficacy. CONCLUSION: Our findings suggested that the small molecule compound SMC18, which dual-targets the CD47/SIRPα and PD-1/PD-L1 pathways, could be a candidate for promoting macrophage- and T-cell-mediated phagocytosis and immune responses in cancer immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes , CD47 Antigen/metabolism , B7-H1 Antigen , Phagocytosis , Immunotherapy , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment
2.
Small ; 18(20): e2107001, 2022 05.
Article in English | MEDLINE | ID: mdl-35434938

ABSTRACT

Radiotherapy (RT) has been shown to cause immunogenic cell death (ICD) of cancer cells, which promote the release of tumor-associated antigens, and trigger the cancer-immunity cycle (CIC). However, ICD induced by RT usually does not occur in hypoxic tumor cells due to their resistance to radiation. Moreover, RT also induces programmed death ligand 1 (PD-L1) upregulation on tumor cells, which has an inhibitory effect on T lymphocytes. Therefore, therapy based on CIC must selectively target the restricted steps of antitumor immunity. Herein, the authors design a versatile three-in-one assembling nanoparticle that can simultaneously execute these obstacles. The amphiphilic peptide drug conjugate NIA-D1, containing the hydrophobic radio-sensitizer 2-(2-nitroimidazol-1-yl) acetic acid (NIA), a peptide substrate of matrix metalloproteinase-2, and a hydrophilic PD-L1 antagonist D PPA-1, is constructed and co-assembled with hydrophobic Toll-like receptor (TLR) 7/8 agonist R848 to form nanoparticle NIA-D1@R848. The NIA-D1@R848 nanoparticles combined with RT can trigger the apoptosis of tumor cells and initiate the CIC. In the presence of R848, it promotes the maturation of dendritic cells, which together with protein programmed cell death protein 1 (PD-1) and its ligand PD-L1  blockade to relieve T cell suppression, and amplify the antitumor immune cycle. In conclusion, a functionalized three-in-one nanoparticle NIA-D1@R848 is successfully constructed, which can induce strong systemic antitumor immune response.


Subject(s)
Nanoparticles , Neoplasms , Toll-Like Receptor 8/agonists , Adjuvants, Immunologic , B7-H1 Antigen/metabolism , Cell Line, Tumor , Humans , Immunity , Immunotherapy , Matrix Metalloproteinase 2 , Nanoparticles/chemistry , Neoplasms/drug therapy , Peptides/therapeutic use , Toll-Like Receptor 7
3.
Pharmacol Res ; 182: 106343, 2022 08.
Article in English | MEDLINE | ID: mdl-35798286

ABSTRACT

Although the blockade of immune checkpoint PD-1/PD-L1 has achieved great success, the lack of tumor-infiltrating immune cells and PD-L1 expression in the tumor microenvironment results in a limited response in certain tumor types. Thus, rational and optimal combination strategies were urgently needed. The combination of PD-1/PD-L1 blockade and anti-angiogenic therapy has been reported to have great potential. Here, a chimeric peptide OGS was designed by conjugating the peptides OPBP-1 (8-12) and DA7R targeting PD-L1 and VEGFR2, respectively. OGS could bind to both human and mouse PD-L1 with high affinity and block the PD-1/PD-L1 interaction, and also inhibit the migration and tube formation of HUVEC cells in wound healing and tube formation assays. To further prolong the half-life of OGS, it was modified by coupling with peptide DSP which has a high binding affinity to both human serum albumin (HSA) and mouse serum albumin (MSA) to form the peptide DSPOGS. DSPOGS could not directly affect the viability, apoptosis, and cell cycle of tumor cells in vitro, while significantly inhibiting the tumor growth in the MC38 mouse model. DSPOGS could elicit a potent anti-tumor immune response and inhibit tumor angiogenesis, with the enhancement of tumor infiltrating CD8+ T cells and the IFN-γ secreting CD8+ T cells in the spleen and tumor-draining lymph node. Further, the combination of radiotherapy with DSPOGS could dramatically improve the therapeutic efficacy. Our study could provide a promising paradigm for the combination of immune checkpoint blockade, anti-angiogenesis, and radiotherapy.


Subject(s)
B7-H1 Antigen , Neoplasms , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Humans , Immunotherapy/methods , Mice , Neoplasms/drug therapy , Neoplasms/radiotherapy , Peptides/pharmacology , Peptides/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment
4.
Toxicol Appl Pharmacol ; 420: 115523, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33838154

ABSTRACT

Metformin, as the first-line drug for the treatment of type 2 diabetes mellitus, has been shown to possess a capability to activate or inhibit the production of reactive oxygen species (ROS) in different ways. However, the detailed mechanisms of the opposite effect are poorly understood. Here we provide evidence that metformin induces accumulation of ROS by inhibiting the expression of a core antioxidant transcription factor nuclear factor erythroid 2 like 1 (NFE2L1/Nrf1) in human hepatocellular carcinoma HepG2 cells. In the present study, we originally found that the increased ROS induced by metformin was blunted in NFE2L1 knockdown cell line. Furtherly by examining the effects of metformin on endogenous and exogenous NFE2L1, we also found metformin could not only inhibit the transcription of NFE2L1 gene, but also promote the degradation of NFE2L1 protein at the post-transcriptional level, whereas this effect can be reversed by high glucose. The inhibitory effect of metformin on NFE2L1 was investigated to occur through the N-terminal domain (NTD) of NFE2L1 protein, and its downregulation by metformin was in an AMP-activated protein kinase (AMPK)-independent manner. But the activation of AMPK signaling pathway by metformin in NFE2L1 knockdown HepG2 cells is reversed, indicating that NFE2L1 may be an important regulator of AMPK signal. Altogether, this work provides a better understanding of the relationship between metformin and oxidative stress, and hence contributes to translational study of metformin through its hypoglycemic and tumor suppressive effects.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Metformin/pharmacology , NF-E2-Related Factor 1/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , NF-E2-Related Factor 1/genetics , Signal Transduction
5.
Cell Commun Signal ; 19(1): 12, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33557880

ABSTRACT

BACKGROUND: TIGIT, as a novel immune checkpoint molecule involved in T cell and NK cell anergy, could induce the immune tolerance and escape through binding with its ligand PVR. Blockade of TIGIT/PVR is considered as a promising strategy in cancer immunotherapy. However, to facilitate the design of inhibitors targeting TIGIT/PVR, the structural characteristics and binding mechanism still need to be further studied. METHODS: In this study, molecular dynamics (MD) simulations and in silico mutagenesis were used to analyze the interaction between TIGIT and its ligand PVR. Then, PVR mutants were designed and their activities were determined by using TIGIT overexpressed Jurkat cells. RESULTS: The results suggested that the loops of PVR (CC' loop, C'C″ loop, and FG loop) underwent a large intra-molecular rearrangement, and more hydrogen bond crosslinking between PVR and TIGIT were formed during MD simulations. The potential residues for PVR to interact with TIGIT were identified and utilized to predict high affinity PVR mutants. Through the biological activity evaluation, four PVR mutants (PVRS72W, PVRS72R, PVRG131V and PVRS132Q) with enhanced affinity to TIGIT were discovered, which could elicit more potent inhibitory effects compared with the wild type PVR. CONCLUSIONS: The MD simulations analysis provided new insights into the TIGIT/PVR interaction model, and the identified PVR mutants (PVRS72W, PVRS72R, PVRG131V and PVRS132Q) could serve as new candidates for immunotherapy to block TIGIT/PVR. Video Abstract.


Subject(s)
Receptors, Immunologic/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Animals , CHO Cells , Coculture Techniques , Computer-Aided Design , Cricetulus , HEK293 Cells , Humans , Jurkat Cells , Molecular Dynamics Simulation , Mutation , Protein Binding , Receptors, Immunologic/chemistry , Receptors, Virus/chemistry
6.
Cell Commun Signal ; 18(1): 142, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32894141

ABSTRACT

BACKGROUND: Inhibitors targeting immune checkpoint were proved effective in cancer immunotherapy, such as PD-1/PD-L1 blockade. The novel immune checkpoint TIGIT/PVR plays critical roles in suppressing the anti-tumor effects of CD8+ T and NK cells, and dual blockade of TIGIT/PVR and PD-1/PD-L1 by antibody can elicit synergistic effects in tumor models and clinical trials. However, small molecules for TIGIT/PVR blockade have not been investigated. METHODS: The expression of PVR in tumors were analyzed by using TCGA, Oncomine and GEO database, and in cancer cell lines examined by flow cytometry. Natural product compounds were docked to PVR for virtual screening by using the software Molecular Operating Environment (MOE). Candidate compounds were further tested by biolayer interferometry-based binding assay, microscale thermophoresis assay and cell based blocking assay. The in vitro activity of the candidate compound was determined by MTT, peripheral blood mononuclear cells (PBMCs) activation assay and coculture assay. The anti-tumor effects and mechanism were also investigated by using MC38 tumor-bearing mice model and immune cell depletion tumor model. RESULTS: PVR was over-expressed in many tumor tissues and cancer cell lines, making it a promising therapeutic target. Through virtual screening, binding, and blocking assay, liothyronine was discovered to bind PVR and block the interaction of TIGIT/PVR. Liothyronine could enhance the function of CD4+ and CD8+ T cells in PBMCs. Besides, in the Jurkat-hTIGIT and CHOK1-hPVR coculture assay, liothyronine could reverse the IL-2 secretion inhibition resulted by TIGIT/PVR ligation. Although had no influence on the proliferation of tumor cells in vitro, liothyronine could significantly inhibit tumor growth when administrated in vivo, by enhancing CD8+ T cell infiltration and immune responses in the tumor bearing mice. The immune cell depletion model showed that the anti-tumor effects of liothyronine depends on CD4+ T cells, CD8+ T cells and NK cells. CONCLUSIONS: A small molecule liothyronine was discovered to serve as a potential candidate for cancer immunotherapy by blocking the immune checkpoint TIGIT/PVR. Video abstract.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/therapy , Receptors, Immunologic/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Triiodothyronine/therapeutic use , Animals , Cell Line, Tumor , Drug Repositioning , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Receptors, Immunologic/immunology , Receptors, Virus/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Triiodothyronine/pharmacology
7.
Angew Chem Int Ed Engl ; 59(35): 15114-15118, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32386245

ABSTRACT

The low response rate and adaptive resistance of PD-1/PD-L1 blockade demands the studies on novel therapeutic targets for cancer immunotherapy. We discovered that a novel immune checkpoint TIGIT expressed higher than PD-1 in many tumors especially anti-PD-1 resistant tumors. Here, mirror-image phage display bio-panning was performed using the d-enantiomer of TIGIT synthesized by hydrazide-based native chemical ligation. d-peptide D TBP-3 was identified, which could occupy the binding interface and effectively block the interaction of TIGIT with its ligand PVR. D TBP-3 showed proteolytic resistance, tumor tissue penetrating ability, and significant tumor suppressing effects in a CD8+ T cell dependent manner. More importantly, D TBP-3 could inhibit tumor growth and metastasis in anti-PD-1 resistant tumor model. This is the first d-peptide targeting TIGIT, which could serve as a potential candidate for cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Peptides/metabolism , Receptors, Immunologic/metabolism , Humans
8.
Cell Commun Signal ; 16(1): 25, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29879980

ABSTRACT

BACKGROUND: Programmed cell death protein 1 (PD-1), a negative co-stimulatory molecule, plays crucial roles in immune escape. Blockade of the interaction between PD-1 and PD-L1 shows exciting clinical responses in a fraction of cancer patients and the success makes PD-1 as a valuable target in immune checkpoint therapy. For the rational design of PD-1 targeting modulators, the ligand binding mechanism of PD-1 should be well understood in prior. METHODS: In this study, we applied 50 ns molecular dynamics simulations to observe the structural properties of PD-1 molecule in both apo and ligand bound states, and we studied the structural features of PD-1 in human and mouse respectively. RESULTS: The results showed that the apo hPD-1 was more flexible than that in PD-L1 bound state. We unexpectedly found that K135 was important for binding energy although it was not at the binding interface. Moreover, the residues which stabilized the interactions with PD-L1 were distinguished. Taking the dynamic features of these residues into account, we identified several residual sites where mutations may gain the function of ligand binding. The in vitro binding experiments revealed the mutants M70I, S87 W, A129L, A132L, and K135 M were better in ligand binding than the wild type PD-1. CONCLUSIONS: The structural information from MD simulation combined with in silico mutagenesis provides guidance to design engineered PD-1 mutants to modulate the PD-1/PD-L1 pathway.


Subject(s)
Molecular Dynamics Simulation , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Protein Engineering , Amino Acid Sequence , Animals , Apoproteins/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Binding Sites , Humans , Ligands , Mice , Mutagenesis , Mutant Proteins/chemistry , Programmed Cell Death 1 Receptor/chemistry , Protein Domains
9.
Materials (Basel) ; 17(16)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39203125

ABSTRACT

During the process of chip formation, the chip is subjected to extrusion pressure, friction, heat, and a strong chemical reaction. The chip's macro and micro morphology, to a certain extent, reflect the condition of the tool during the cutting procedure. Therefore, researching the macroscopic and microscopic morphology of the chip's surface in response to different tool wear conditions is of great significance to reproducing the cutting condition and analyzing the tool wear mechanism. This paper focuses on the chips formed by milling the difficult-to-machine material 508III high-strength steel. Firstly, the 508III steel milling experiment is carried out at the actual machining site to collect chip data under different tool wear conditions. Next, the free surface morphology of chips and the bottom surface morphology of chips are analyzed. Further, the chip edges are investigated, and their causes are analyzed. Finally, heavy milling 508III steel chip curl morphology analysis is performed. The research results play important roles in revealing the mechanism of tool wear and the relationship between chip morphology and tool wear. This information can be used to provide theoretical and technical support for monitoring the tool wear status based on chip morphology.

10.
J Colloid Interface Sci ; 677(Pt B): 417-428, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39153245

ABSTRACT

Synthesis of catalysts with high catalytic degradation activity for formaldehyde (HCHO) at room temperature is highly desirable for indoor air quality control. Herein, a novel K-Mn3O4@CeO2 catalyst with excellent catalytic oxidation activity toward HCHO at near room temperature was reported. In particular, the K addition in K-Mn3O4@CeO2 considerably enhanced the oxidation activity, and importantly, 99.3 % conversion of 10 mL of a 40 mg/L HCHO solution at 30 °C for 14 h was achieved, with simultaneous strong cycling stability. Moreover, the addition of K species considerably influenced the chemical valence state of Mn from +4 (ε-MnO2) to +8/3 (Mn3O4) on the surface of CeO2, which obviously changed the tunnel structure and the number of oxygen vacancies. One part of K species is uniformly dispersed on K-Mn3O4@CeO2, and the other part exists in the tunnel structure of Mn3O4@CeO2, which is mainly used to balance the negative charge of the tunnel and prevent collapse of the structure, providing enough active sites for the catalytic oxidation of HCHO. We observed a phase transition from tunneled KMnO2 to Mn3O4 to tunneled MnO2 with the decreasing K+ content, in which K-Mn3O4@CeO2 exhibited higher HCHO oxidation activity. In addition, K-Mn3O4@CeO2 exhibited lower oxygen vacancy formation and HCHO adsorption energies in aqueous solution based on density functional theory calculations. This is because the K species provide more active oxygen species and richer oxygen vacancies on the surface of K-Mn3O4@CeO2, promote the mobility of lattice oxygen and the room-temperature reduction properties of oxygen species, and enhance the ability of the catalyst to replenish the consumed oxygen species. Finally, a possible HCHO catalytic oxidation pathway on the surface of K-Mn3O4@CeO2 catalyst is proposed.

11.
Biochem Pharmacol ; 223: 116162, 2024 05.
Article in English | MEDLINE | ID: mdl-38527557

ABSTRACT

Immune checkpoint inhibitors have unveiled promising clinical prospects in cancer treatment. Nonetheless, their effectiveness remains restricted, marked by consistently low response rates and affecting only a subset of patients. The co-blockade of TIGIT with PD-1 has exhibited substantial anti-tumor effects. Notably, there is a dearth of reports on small-molecule inhibitors concurrently targeting both TIGIT and PD-1. In this study, we employed Microscale Thermophoresis (MST) to screen our laboratory's existing repository of small molecules. Our findings illuminated Gln(TrT) 's affinity for both TIGIT and PD-1, affirming its potential to effectively inhibit TIGIT/PVR and PD-1/PD-L1 pathways. In vitro co-culture experiments substantiated Gln(TrT)'s proficiency in restoring Jurkat T-cell functionality by blocking both TIGIT/PVR and PD-1/PD-L1 interactions. In the MC38 murine tumor model, Gln(TrT) emerges as a pivotal modulator, promoting the intratumoral infiltration and functional competence of CD8+ T cells. Furthermore, whether used as a monotherapy or in conjunction with radiotherapy, Gln(TrT) substantially impedes MC38 tumor progression, significantly extending the survival of murine subjects.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Animals , Mice , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/metabolism , Receptors, Immunologic , Immunotherapy , Neoplasms/drug therapy , Neoplasms/metabolism
12.
J Control Release ; 373: 568-582, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067792

ABSTRACT

Cancer vaccine is regarded as an effective immunotherapy approach mediated by dendritic cells (DCs) which are crucial for antigen presentation and the initiation of adaptive immune responses. However, lack of DC-targeting properties significantly hampers the efficacy of cancer vaccines. Here, by using the phage display technique, peptides targeting the endocytic receptor DEC-205 primarily found on cDC1s were initially screened. An optimized hydrolysis-resistant peptide, hr-8, was identified and conjugated to PLGA-loaded antigen (Ag) and CpG adjuvant nanoparticles, resulting in a DC-targeting nanovaccine. The nanovaccine hr-8-PLGA@Ag/CpG facilitates dendritic cell maturation and improves antigen cross-presentation. The nanovaccine can enhance the antitumor immune response mediated by CD8+ T cells by encapsulating the nanovaccine with either exogenous OVA protein antigen or endogenous gp100/E7 antigenic peptide. As a result, strong antitumor effects are observed in both anti-PD-1 responsive B16-OVA and anti-PD-1 non-responsive B16 and TC1 immunocompetent tumor models. In summary, this study presents the initial documentation of a nanovaccine that targets dendritic cells via the novel DEC-205 binding peptide. This approach offers a new method for developing cancer vaccines that can potentially improve the effectiveness of cancer immunotherapy.


Subject(s)
Cancer Vaccines , Dendritic Cells , Immunotherapy , Lectins, C-Type , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Nanoparticles , Peptides , Receptors, Cell Surface , Dendritic Cells/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Animals , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , Immunotherapy/methods , Nanoparticles/chemistry , Peptides/chemistry , Peptides/administration & dosage , Minor Histocompatibility Antigens/immunology , Cell Line, Tumor , Ovalbumin/immunology , Ovalbumin/administration & dosage , Female , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Mice , Antigens, CD/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , CD8-Positive T-Lymphocytes/immunology , Nanovaccines
13.
Sci China Life Sci ; 67(5): 996-1009, 2024 May.
Article in English | MEDLINE | ID: mdl-38324132

ABSTRACT

The immune checkpoint TIGIT/PVR blockade exhibits significant antitumor effects through activation of NK and CD8+ T cell-mediated cytotoxicity. Immune checkpoint blockade (ICB) could induce tumor ferroptosis through IFN-γ released by immune cells, indicating the synergetic effects of ICB with ferroptosis in inhibiting tumor growth. However, the development of TIGIT/PVR inhibitors with ferroptosis-inducing effects has not been explored yet. In this study, the small molecule Hemin that could bind with TIGIT to block TIGIT/PVR interaction was screened by virtual molecular docking and cell-based blocking assay. Hemin could effectively restore the IL-2 secretion from Jurkat-hTIGIT cells. Hemin reinvigorated the function of CD8+ T cells to secrete IFN-γ and the elevated IFN-γ could synergize with Hemin to induce ferroptosis in tumor cells. Hemin inhibited tumor growth by boosting CD8+ T cell immune response and inducing ferroptosis in CT26 tumor model. More importantly, Hemin in combination with PD-1/PD-L1 blockade exhibited more effective antitumor efficacy in anti-PD-1 resistant B16 tumor model. In summary, our finding indicated that Hemin blocked TIGIT/PVR interaction and induced tumor cell ferroptosis, which provided a new therapeutic strategy to combine immunotherapy and ferroptosis for cancer treatment.


Subject(s)
Ferroptosis , Hemin , Immunotherapy , Receptors, Immunologic , Hemin/pharmacology , Receptors, Immunologic/metabolism , Animals , Humans , Ferroptosis/drug effects , Mice , Immunotherapy/methods , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Molecular Docking Simulation , Jurkat Cells , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/pharmacology , Drug Synergism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Receptors, Virus/metabolism , Mice, Inbred BALB C
14.
Acta Pharm Sin B ; 14(3): 1150-1165, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38486998

ABSTRACT

Aside from antibodies, peptides show great potential as immune checkpoint inhibitors (ICIs) due to several advantages, such as better tumor penetration and lower cost. Lymphocyte-activation gene 3 (LAG-3) is an immune checkpoint which can induce T cell dysfunction through interaction with its soluble ligand fibrinogen like protein-1 (FGL1). Here, we found that LAG-3 expression was higher than programmed cell death protein 1 (PD-1) in multiple human cancers by TCGA databases, and successfully identified a LAG-3 binding peptide LFP-6 by phage display bio-panning, which specifically blocks the interaction of LAG-3/FGL1 but not LAG-3/MHC-II. Subsequently, d-amino acids were introduced to substitute the N- and C-terminus of LFP-6 to obtain the proteolysis-resistant peptide LFP-D1, which restores T cell function in vitro and inhibits tumor growth in vivo. Further, a bispecific peptide LFOP targeting both PD-1/PD-L1 and LAG-3/FGL1 was designed by conjugating LFP-D1 with PD-1/PD-L1 blocking peptide OPBP-1(8-12), which activates T cell with enhanced proliferation and IFN-γ production. More importantly, LFOP combined with radiotherapy significantly improve the T cell infiltration in tumor and elevate systemic antitumor immune response. In conclusion, we developed a novel peptide blocking LAG-3/FGL1 which can restore T cell function, and the bispecific peptide synergizes with radiotherapy to further enhance the antitumor immune response.

15.
Cancer Immunol Immunother ; 62(11): 1723-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24077852

ABSTRACT

PIWIL2, a member of PIWI/AGO family, is expressed in germline stem cells and precancerous stem cells, but not in adult somatic cells. PIWIL2 plays an important role in tumor development. It is considered as a cancer­testis antigen (CT80). It has been reported that the spliced fragment of PIWIL2, PL2L60, was widely expressed in cancer cell lines. In this study, HLA-A2-restricted epitopes from PL2L60 were predicted by online tools. To improve the activity of the native epitope, a candidate peptide P281 with potent binding affinity was chosen to investigate the modification strategy. A series of aromatic amino acids were introduced to substitute the first residue of P281. Then, we tested the binding affinity and stability of the peptide analogs and their ability to elicit specific immune responses both in vitro and in vivo. Our results indicated that the cytotoxic T lymphocytes (CTLs) induced by [4-Cl-Phe1]P281 could elicit more potent activities than that of P281 and other analogs. The CTLs induced by this analog could lyze target cells in HLA-A2-restricted and antigen-specific manners. [4-Cl-Phe1]P281 also showed the best resistance against degradation in human serum. In conclusion, the introduction of the unnatural amino acid, 4-Cl-Phe, into the first position could enhance the activity of the native epitope to induce cytotoxic T lymphocytes. It might be a good strategy to modify other promising native epitopes. The novel epitopes identified in this study could be used as novel candidates to the immunotherapy of HLA-A2 positive patients with tumors expressing PL2L60.


Subject(s)
Argonaute Proteins/immunology , Epitopes, T-Lymphocyte/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Alternative Splicing , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Blotting, Western , Cell Line , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Gene Expression Regulation, Neoplastic , HLA-A2 Antigen/immunology , HT29 Cells , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , MCF-7 Cells , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Peptides/genetics , Peptides/metabolism , Phenylalanine/genetics , Phenylalanine/immunology , Phenylalanine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/metabolism
16.
Amino Acids ; 44(2): 499-510, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22802137

ABSTRACT

CyclinD1/CDK4 and cyclinD3/CDK4 complexes are key regulators of the cell progression and therefore constitute promising targets for the design of anticancer agents. In the present study, the key peptide motifs were selected from these two complexes. Chimeric peptides with these peptides conjugated to the protein transduction domain 4 (PTD4) were designed and synthesized. The chimeric peptides, PTD4-D1, PTD4-D3, PTD4-K4 exhibited significant anti-proliferation effects on cancer cell lines. These peptides could compete with the cyclinD/CDK4 complex and induce the G1/S phase arrest and apoptosis of cancer cells. In the tumor challenge experiment, these peptides showed potent antitumor effects with no significant side effects. Our results suggested that these peptides could be served as novel leading compounds with potent antitumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin D1/genetics , Cyclin D3/genetics , Cyclin-Dependent Kinase 4/genetics , Neoplasms/drug therapy , Peptide Fragments/genetics , Peptides/pharmacology , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Cycle/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/physiopathology , Peptides/chemical synthesis , Peptides/genetics
17.
Acta Pharm Sin B ; 13(11): 4511-4522, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37969728

ABSTRACT

Developing new therapeutic agents for cancer immunotherapy is highly demanding due to the low response ratio of PD-1/PD-L1 blockade in cancer patients. Here, we discovered that the novel immune checkpoint VISTA is highly expressed on a variety of tumor-infiltrating immune cells, especially myeloid derived suppressor cells (MDSCs) and CD8+ T cells. Then, peptide C1 with binding affinity to VISTA was developed by phage displayed bio-panning technique, and its mutant peptide VS3 was obtained by molecular docking based mutation. Peptide VS3 could bind VISTA with high affinity and block its interaction with ligand PSGL-1 under acidic condition, and elicit anti-tumor activity in vivo. The peptide DVS3-Pal was further designed by d-amino acid substitution and fatty acid modification, which exhibited strong proteolytic stability and significant anti-tumor activity through enhancing CD8+ T cell function and decreasing MDSCs infiltration. This is the first study to develop peptides to block VISTA/PSGL-1 interaction, which could act as promising candidates for cancer immunotherapy.

18.
Biochem Pharmacol ; 212: 115583, 2023 06.
Article in English | MEDLINE | ID: mdl-37148978

ABSTRACT

PD-1/PD-L1 blockade has achieved substantial clinical results in cancer treatment. However, the expression of other immune checkpoints leads to resistance and hinders the efficacy of PD-1/PD-L1 blockade. T cell immunoglobulin and mucin domain 3 (TIM-3), a non-redundant immune checkpoint, synergizes with PD-1 to mediate T cell dysfunction in tumor microenvironment. Development of small molecules targeting TIM-3 is a promising strategy for cancer immunotherapy. Here, to identify small molecule inhibitors targeting TIM-3, the docking pocket in TIM-3 was analyzed by Molecular Operating Environment (MOE) and the Chemdiv compound database was screened. The small molecule SMI402 could bind to TIM-3 with high affinity and prevent the ligation of PtdSer, HMGB1, and CEACAM1. SMI402 reinvigorated T cell function in vitro. In the MC38-bearing mouse model, SMI402 inhibited tumor growth by increasing CD8+ T and natural killing (NK) cells infiltration at the tumor site, as well as restoring the function of CD8+ T and NK cells. In conclusions, the small molecule SMI402 shows promise as a leading compound which targets TIM-3 for cancer immunotherapy.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Neoplasms , Animals , Mice , Hepatitis A Virus Cellular Receptor 2/metabolism , Programmed Cell Death 1 Receptor , B7-H1 Antigen , Neoplasms/drug therapy , Immunotherapy/methods , Tumor Microenvironment
19.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37344099

ABSTRACT

BACKGROUND: Aside from immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1), intervention of CD47/Sirpα mediated 'don't eat me' signal between macrophage and tumor cell is considered as a promising therapeutic approach for cancer immunotherapy. Compared with CD47, the novel immune checkpoint CD24/Siglec-10 can also deliver 'don't eat me' signal and CD24 shows much lower expression level in normal tissue which might avoid unwanted side effects. METHODS: Cell-based phage display biopanning and D-amino acid modification strategy were used to identify the CD24/Siglec-10 blocking peptide. Cell-based blocking assay and microscale thermophoresis assay were used to validate the blocking and binding activities of the peptide. Phagocytosis and co-culture assays were used to explore the in vitro function of the peptide. Flow cytometry was performed to assess the immune microenvironment after the peptide treatment in vivo. RESULTS: A CD24/Siglec-10 blocking peptide (CSBP) with hydrolysis-resistant property was identified. Surprisingly, we found that CSBP could not only block the interaction of CD24/Siglec-10 but also PD-1/PD-L1. CSBP could induce the phagocytosis of tumor cell by both the macrophages and monocytic myeloid-derived suppressor cells (M-MDSCs), which can further activate CD8+ T cells. Besides, combination of radiotherapy and CSBP synergistically reduced tumor growth and altered the tumor microenvironment in both anti-PD-1-responsive MC38 and anti-PD-1-resistant 4T1 tumor models. CONCLUSIONS: In summary, this is the first CD24/Siglec-10 blocking peptide which blocked PD-1/PD-L1 interaction as well, functioned via enhancing the phagocytosis of tumor cells by macrophages and M-MDSCs, and elevating the activity of CD8+ T cells for cancer immunotherapy.


Subject(s)
CD47 Antigen , Neoplasms , Humans , B7-H1 Antigen , CD24 Antigen/metabolism , CD47 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Immunotherapy , Neoplasms/radiotherapy , Neoplasms/drug therapy , Peptides/pharmacology , Peptides/therapeutic use , Sialic Acid Binding Immunoglobulin-like Lectins/therapeutic use , Tumor Microenvironment
20.
Microbiol Immunol ; 56(8): 548-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22537173

ABSTRACT

The identification of novel cytotoxic T lymphocyte (CTL) epitopes is important to analysis of the involvement of CD8(+) T cells in Mycobacterium tuberculosis infection as well as to the development of peptide vaccines. In this study, a novel CTL epitope from region of difference 11 encoded antigen Rv3425 was identified. Epitopes were predicted by the reversal immunology approach. Rv3425-p118 (LIASNVAGV) was identified as having relatively strong binding affinity and stability towards the HLA-A*0201 molecule. Peripheral blood mononuclear cells pulsed by this peptide were able to release interferon-γ in healthy donors (HLA-A*02(+) purified protein derivative(+)). In cytotoxicity assays in vitro and in vivo, Rv3425-p118 induced CTLs to specifically lyse the target cells. Therefore, this epitope could provide a subunit component for designing vaccines against Mycobacterium tuberculosis.


Subject(s)
Bacterial Proteins/immunology , Epitopes, T-Lymphocyte/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Computational Biology , Cytotoxicity Tests, Immunologic , Epitope Mapping , HLA-A2 Antigen/metabolism , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice , Mice, Transgenic , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL