Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.383
Filter
1.
Cancer Immunol Immunother ; 73(10): 190, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105882

ABSTRACT

Transforming growth factor ß (TGFß) is present in blood of patients who do not respond to anti-programmed cell death (ligand) 1 [PD-(L)1] treatment, and through synergy with vascular endothelial growth factor (VEGF), it helps to create an environment that promotes tumor immune evasion and immune tolerance. Therefore, simultaneous inhibition of TGFß/VEGF is more effective than targeting TGFß alone. In this study, the dual inhibitory mechanism of TU2218 was identified through in vitro analysis mimicking the tumor microenvironment, and its antitumor effects were analyzed using mouse syngeneic tumor models. TU2218 directly restored the activity of damaged cytotoxic T lymphocytes (CTLs) and natural killer cells inhibited by TGFß and suppressed the activity and viability of regulatory T cells. The inactivation of endothelial cells induced by VEGF stimulation was completely ameliorated by TU2218, an effect not observed with vactosertib, which inhibits only TGFß signaling. The combination of TU2218 and anti-PD1 therapy had a significantly greater antitumor effect than either drug alone in the poorly immunogenic B16F10 syngeneic tumor model. The mechanism of tumor reduction was confirmed by flow cytometry, which showed upregulated VCAM-1 expression in vascular cells and increased influx of CD8 + CTLs into the tumor. As another strategy, combination of anti-CTLA4 therapy and TU2218 resulted in high complete regression (CR) rates in CT26 and WEHI-164 tumor models. In particular, immunological memory generated by the combination of anti-CTLA4 and TU2218 in the CT26 model prevented the development of tumors after additional tumor cell transplantation, suggesting that the TU2218-based combination has therapeutic potential in immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors , Receptor, Transforming Growth Factor-beta Type I , Vascular Endothelial Growth Factor Receptor-2 , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/immunology , Humans , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Female , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Cell Line, Tumor , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Immunotherapy/methods
2.
Nat Commun ; 15(1): 7366, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191730

ABSTRACT

The lysine-specific histone demethylase 1 A (LSD1) is involved in antitumor immunity; however, its role in shaping CD8 + T cell (CTL) differentiation and function remains largely unexplored. Here, we show that pharmacological inhibition of LSD1 (LSD1i) in CTL in the context of adoptive T cell therapy (ACT) elicits phenotypic and functional alterations, resulting in a robust antitumor immunity in preclinical models in female mice. In addition, the combination of anti-PDL1 treatment with LSD1i-based ACT eradicates the tumor and leads to long-lasting tumor-free survival in a melanoma model, complementing the limited efficacy of the immune or epigenetic therapy alone. Collectively, these results demonstrate that LSD1 modulation improves antitumoral responses generated by ACT and anti-PDL1 therapy, providing the foundation for their clinical evaluation.


Subject(s)
CD8-Positive T-Lymphocytes , Histone Demethylases , Immunotherapy, Adoptive , Mice, Inbred C57BL , Animals , Histone Demethylases/metabolism , Histone Demethylases/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Mice , Female , CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Cell Line, Tumor , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Humans , Melanoma/immunology , Melanoma/therapy
3.
Int J Pharm ; 663: 124581, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39137819

ABSTRACT

Cold tumors lack T cells infiltration and have low immunogenicity, resulting insufficient immunotherapy response. Therefore, how to realize the transformation from cold tumor to hot tumor is an urgent problem to be solved. Photodynamic therapy can induce immunogenic death of tumor cells (ICD) and activate T lymphocytes to produce tumor immune response. However, hypoxia in the cold tumor microenvironment limits the effectiveness of photodynamic therapy. So in this article, MET-HMME/CAT-HMME@Nlip as a functional co-delivery nanoliposomes was constructed based on overcoming the above problems. Firstly, the oxygen-deficient state could be improved by the following two ways, one is catalase loaded in CAT-HMME@Nlip can decompose high concentration hydrogen peroxide to produce oxygen, and the other is metformin loaded in MET-HMME@Nlip can decrease oxygen consumption by inhibiting of mitochondrial respiration. And then with the increase of substrate oxygen concentration, the sensitivity of photodynamic therapy can be greatly improved and the anti-tumor immune response by PDT-induced ICD can also be enhanced obviously. In addition, metformin could act as a small molecule immune checkpoint inhibitor to reduce the expression of PD-L1 on the surface of tumor cells, thereby effectively improving the specific killing ability of cytotoxic T cells to tumor cells which could not only erasing the primary tumor, but also inhibiting the growth of simulated distant tumors through the immune memory function. This study provides a new idea for improving the clinical treatment effect of hypoxic cold tumors, especially for tumors that could not benefit from immunotherapy due to low or no expression of PD-L1 protein on the surface of tumor cells.


Subject(s)
B7-H1 Antigen , Immunotherapy , Liposomes , Metformin , Nanoparticles , Photochemotherapy , Tumor Microenvironment , Photochemotherapy/methods , Animals , Metformin/administration & dosage , Metformin/pharmacology , Immunotherapy/methods , Tumor Microenvironment/drug effects , Cell Line, Tumor , Nanoparticles/administration & dosage , Mice , Catalase/administration & dosage , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/immunology , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacology , Female , Photosensitizing Agents/administration & dosage , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Hypoxia/drug effects , Hydrogen Peroxide , Mice, Inbred BALB C
4.
Nat Cardiovasc Res ; 3(8): 970-986, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39196030

ABSTRACT

Doxorubicin, the most prescribed chemotherapeutic drug, causes dose-dependent cardiotoxicity and heart failure. However, our understanding of the immune response elicited by doxorubicin is limited. Here we show that an aberrant CD8+ T cell immune response following doxorubicin-induced cardiac injury drives adverse remodeling and cardiomyopathy. Doxorubicin treatment in non-tumor-bearing mice increased circulating and cardiac IFNγ+CD8+ T cells and activated effector CD8+ T cells in lymphoid tissues. Moreover, doxorubicin promoted cardiac CD8+ T cell infiltration and depletion of CD8+ T cells in doxorubicin-treated mice decreased cardiac fibrosis and improved systolic function. Doxorubicin treatment induced ICAM-1 expression by cardiac fibroblasts resulting in enhanced CD8+ T cell adhesion and transformation, contact-dependent CD8+ degranulation and release of granzyme B. Canine lymphoma patients and human patients with hematopoietic malignancies showed increased circulating CD8+ T cells after doxorubicin treatment. In human cancer patients, T cells expressed IFNγ and CXCR3, and plasma levels of the CXCR3 ligands CXCL9 and CXCL10 correlated with decreased systolic function.


Subject(s)
Disease Models, Animal , Doxorubicin , Fibrosis , Interferon-gamma , T-Lymphocytes, Cytotoxic , Animals , Doxorubicin/adverse effects , Fibrosis/chemically induced , Humans , Dogs , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Interferon-gamma/metabolism , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Mice, Inbred C57BL , Cardiotoxicity/etiology , Receptors, CXCR3/metabolism , Chemokine CXCL10/metabolism , Male , Granzymes/metabolism , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Cardiomyopathies/immunology , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology , Cell Degranulation/drug effects , Chemokine CXCL9/metabolism , Ventricular Function, Left/drug effects , Systole/drug effects , Mice , Female , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Cell Adhesion/drug effects , Lymphocyte Activation/drug effects
5.
Sci Adv ; 10(34): eadp8647, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178257

ABSTRACT

Agonistic antibodies against CD137 have been demonstrated to completely regress established tumors through activating T cell immunity. Unfortunately, current CD137 antibodies failed to benefit patients with cancer. Moreover, their antitumor mechanisms in vivo remain to be determined. Here, we report the development of a small molecular CD137 agonist, JNU-0921. JNU-0921 effectively activates both human and mouse CD137 through direct binding their extracellular domains to induce oligomerization and signaling and effectively shrinks tumors in vivo. Mechanistically, JNU-0921 enhances effector and memory function of cytotoxic CD8+ T cells (CTLs) and alleviates their exhaustion. JNU-0921 also skews polarization of helper T cells toward T helper 1 type and enhances their activity to boost CTL function. Meanwhile, JNU-0921 attenuates the inhibitory function of regulatory T cells on CTLs. Our current work shows that JNU-0921 shrinks tumors by enhancing the cytotoxicity of CTLs in cis and in trans and sheds light on strategy for developing CD137 small molecular agonists.


Subject(s)
CD8-Positive T-Lymphocytes , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Antineoplastic Agents/pharmacology , Signal Transduction/drug effects , Cytotoxicity, Immunologic/drug effects , Xenograft Model Antitumor Assays , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology
6.
Mol Pharm ; 21(8): 3897-3908, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38959154

ABSTRACT

The antitumor strategies based on innate immunity activation have become favored by researchers in recent years. In particular, strategies targeting antiphagocytic signaling blockade to enhance phagocytosis have been widely reported. For example, the addition of prophagocytic signals such as calreticulin could make the strategy significantly more effective. In this study, an antitumor strategy that combines photodynamic therapy (PDT) with CD47 blockade has been reported. This approach promotes the maturation of dendritic cells and the presentation of tumor antigens by PDT-mediated tumor immunogenic cell death, as well as the enhancement of cytotoxic T lymphocyte infiltration in tumor areas and the phagocytic activity of phagocytes. Furthermore, the downregulation and blockage of CD47 protein could further promote phagocytic activity, strengthen the innate immune system, and ultimately elevate the antitumor efficacy and inhibit tumor metastasis.


Subject(s)
CD47 Antigen , Dendritic Cells , Phagocytosis , Photochemotherapy , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , Photochemotherapy/methods , Animals , Mice , Phagocytosis/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Mice, Inbred C57BL , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Humans , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Immunity, Innate/drug effects , Mice, Inbred BALB C , Female
7.
J Control Release ; 372: 715-727, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38955253

ABSTRACT

Chemo-immunotherapy holds the advantage of specific antitumor effects by activating cytotoxic lymphocyte cells (CTLs) immune response. However, multiple barriers have limited the outcomes partly due to tumor-cell-mediated exhaustion of CTLs in the immunosuppressive tumor microenvironment (iTME). Here, we rationally designed a simple-yet-versatile Ca2+ nanogenerator to modulate iTME for enhancing 2-deoxyglucose (2-DG) mediated chemo-immunotherapy. Briefly, after 2-DG chemotherapy, CaO2 nanoparticles coated with EL4 cell membrane (denoted as CaNP@ECM) could preferentially accumulate in tumor tissue via adhesion between LFA-1 on EL4 cell membrane and ICAM-1 on inflamed endothelial cell in tumor tissues and display a series of benefits for CTLs: i) Increasing glucose availability of CTLs while reducing lactic acid secretion through Ca2+ overloading mediated inhibition of tumor cell glycolysis, as well as relieving hypoxia; ii) Reversing CTLs exhaustion via TGF-ß1 scavenging and PD-L1 blockade through PD-1 and TGF-ß1R on EL4 cell membrane; iii) Boosting tumor immunotherapy via immunologic death (ICD) of tumor cells induced by Ca2+ overloading. We demonstrate that the multi-modal Ca2+ nanogenerator rescues T cells from exhaustion and inhibits tumor growth both in vitro and in vivo. More importantly, the study also facilitate the development of glucose metabolism inhibition-based tumor immunotherapy via Ca2+ overloading.


Subject(s)
Calcium , Deoxyglucose , Immunotherapy , Mice, Inbred C57BL , Nanoparticles , Tumor Microenvironment , Animals , Immunotherapy/methods , Deoxyglucose/pharmacology , Deoxyglucose/administration & dosage , Nanoparticles/administration & dosage , Tumor Microenvironment/drug effects , Cell Line, Tumor , Calcium/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Mice , Female , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/drug therapy , Oxides , Humans , Mice, Inbred BALB C , T-Cell Exhaustion , Calcium Compounds
8.
Biomaterials ; 311: 122660, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38865911

ABSTRACT

In "immune-cold" tumors, the upregulation of immunosuppressive cells and insufficient infiltration of lymphocytes contribute to the resistance against immune therapy. Herein, we have developed a dual-enzyme-like photosensitive nanozyme (PBAF) to remodel the tumor immunosuppressive microenvironment (TIME) and induce the tumor infiltration of cytotoxic T lymphocytes (CTLs). Specifically, PBAF exhibits peroxidase (POD)-like activity and glutathione oxidase (GSHOx)-like activity and can be stimulated by 750 nm laser, promoting oxidative stress at the tumor site. Consequently, this process further leads to the reconstruction of TIME in animal experiments, inducing tumor-associated macrophages (TAMs) toward the immunostimulatory M1 phenotype, eliminating myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Simultaneously, PBAF also promotes dendritic cells (DCs) maturation to enhance CTLs infiltration into the tumor. The remodeled TIME and enhanced immune responses by PBAF demonstrate significant post-administration inhibition of recurrence and metastasis in the treatment of malignant tumors.


Subject(s)
Immunotherapy , Tumor Microenvironment , Animals , Tumor Microenvironment/drug effects , Immunotherapy/methods , Mice , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Mice, Inbred C57BL , Dendritic Cells/immunology , Dendritic Cells/metabolism , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Mice, Inbred BALB C , Nanoparticles/chemistry , Female , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/pathology
9.
Acta Biomater ; 184: 383-396, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936753

ABSTRACT

Triple-negative breast cancer (TNBC) is a relatively "cold" tumour with low immunogenicity compared to other tumour types. Especially, the immune checkpoint inhibitors to treat metastatic TNBC only shows the modest immune response rates. Here, we used Chlorella vulgaris as a bioreactor to synthesize an efficient nanobomb (Bio-MnSe) aimed at eliciting systemic anti-tumour immune response. Despite possessing extremely low Mn content, Bio-MnSe effectively produced more ROS and activated stronger cGAS-STING signal pathway compared to pure Se nanoparticles and free Mn2+ ions, promoting the infiltration of natural killer (NK) cells, cytotoxic T lymphocytes (CTLs) in tumour, effectively turning "cold" tumour into "hot" tumour, and achieving strong antitumour immunotherapy. Additionally, the use of αPD-L1 as an immune checkpoint antagonist further increased the anti-tumour immune response of Bio-MnSe, resulting in enhanced anti-tumour effects. Doxorubicin (Dox), an immunogenic cell death (ICD) inducer, was combined with Bio-MnSe to form Bio-MnSe@Dox. This Bio-MnSe@Dox not only directly damaged tumour cells and induced tumour ICD but also promoted dendritic cell maturation, cytotoxic T lymphocyte infiltration, and NK cell recruitment, synergistically intensifying anti-tumour immune responses and suppressing tumour relapse and lung metastasis. Collectively, our findings propose an effective strategy for transforming 'cold' tumours to 'hot' ones, thereby advancing the development of anti-tumour immune drugs. STATEMENT OF SIGNIFICANCE: A biogenic MnSe (Bio-MnSe) nanocomposite was synthesized using Chlorella vulgaris as a bioreactor for enhanced immunotherapy of TNBC. Bio-MnSe demonstrated a stronger ability to activate the cGAS-STING signalling pathway and generate more ROS compared to pure Se nanoparticles and free Mn2+ ions. Apoptotic cells induced by Bio-MnSe released a significant amount of interferon, leading to the activation of T and natural killer (NK) cells, ultimately transforming immunologically 'cold' breast tumours to 'hot' tumours and enhancing the tumour's response to immune checkpoint inhibitors. The combination of Bio-MnSe with Dox or αPD-L1 further enhanced the anti-tumour immune response, fostering dendritic cell maturation, infiltration of cytotoxic T lymphocytes, and recruitment of NK cells, thereby enhancing the anti-tumour immunotherapy of TNBC.


Subject(s)
Immunogenic Cell Death , Manganese , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Animals , Female , Immunogenic Cell Death/drug effects , Mice , Humans , Cell Line, Tumor , Signal Transduction/drug effects , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Manganese/chemistry , Manganese/pharmacology , Doxorubicin/pharmacology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/drug therapy , Selenium/chemistry , Selenium/pharmacology , Immunotherapy/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , B7-H1 Antigen/metabolism , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects
10.
Nat Cancer ; 5(8): 1206-1226, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38844817

ABSTRACT

Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Uridine Diphosphate , Humans , Uridine Diphosphate/metabolism , Immunotherapy/methods , Drug Resistance, Neoplasm/immunology , Animals , Mice , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Receptors, Purinergic P2/metabolism , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Microenvironment/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Nucleotides/metabolism , Immune Tolerance , Programmed Cell Death 1 Receptor
11.
JCI Insight ; 9(12)2024 May 21.
Article in English | MEDLINE | ID: mdl-38912586

ABSTRACT

Immune therapy is the new frontier of cancer treatment. Therapeutic radiation is a known inducer of immune response and can be limited by immunosuppressive mediators including cyclooxygenase-2 (COX2) that is highly expressed in aggressive triple negative breast cancer (TNBC). A clinical cohort of TNBC tumors revealed poor radiation therapeutic efficacy in tumors expressing high COX2. Herein, we show that radiation combined with adjuvant NSAID (indomethacin) treatment provides a powerful combination to reduce both primary tumor growth and lung metastasis in aggressive 4T1 TNBC tumors, which occurs in part through increased antitumor immune response. Spatial immunological changes including augmented lymphoid infiltration into the tumor epithelium and locally increased cGAS/STING1 and type I IFN gene expression were observed in radiation-indomethacin-treated 4T1 tumors. Thus, radiation and adjuvant NSAID treatment shifts "immune desert phenotypes" toward antitumor M1/TH1 immune mediators in these immunologically challenging tumors. Importantly, radiation-indomethacin combination treatment improved local control of the primary lesion, reduced metastatic burden, and increased median survival when compared with radiation treatment alone. These results show that clinically available NSAIDs can improve radiation therapeutic efficacy through increased antitumor immune response and augmented local generation of cGAS/STING1 and type I IFNs.


Subject(s)
Membrane Proteins , Signal Transduction , T-Lymphocytes, Cytotoxic , Animals , Membrane Proteins/metabolism , Mice , Female , Signal Transduction/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/radiotherapy , Indomethacin/pharmacology , Indomethacin/therapeutic use , Cell Line, Tumor , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Nucleotidyltransferases/metabolism , Interferon Type I/metabolism , Cyclooxygenase 2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Mice, Inbred BALB C
12.
Sci Rep ; 14(1): 14248, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902343

ABSTRACT

Treatment of advanced triple-negative breast cancer (TNBC) is a great challenge in clinical practice. The immune checkpoints are a category of immunosuppressive molecules that cancer could hijack and impede anti-tumor immunity. Targeting immune checkpoints, such as anti-programmed cell death 1 (PD-1) therapy, is a promising therapeutic strategy in TNBC. The efficacy and safety of PD-1 monoclonal antibody (mAb) with chemotherapy have been validated in TNBC patients. However, the precise mechanisms underlying the synergistic effect of chemotherapy and anti-PD-1 therapy have not been elucidated, causing the TNBC patients that might benefit from this combination regimen not to be well selected. In the present work, we found that IL-23, an immunological cytokine, is significantly upregulated after chemotherapy in TNBC cells and plays a vital role in enhancing the anti-tumor immune response of cytotoxic T cells (CTLs), especially in combination with PD-1 mAb. In addition, the combination of IL-23 and PD-1 mAb could synergistically inhibit the expression of Phosphoinositide-3-Kinase Regulatory Subunit 1 (PIK3R1), which is a regulatory subunit of PI3K and inhibit p110 activity, and promote phosphorylation of AKT in TNBC-specific CTLs. Our findings might provide a molecular marker that could be used to predict the effects of combination chemotherapy therapy and PD-1 mAb in TNBC.


Subject(s)
Interleukin-23 Subunit p19 , Phosphatidylinositol 3-Kinases , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-akt , Signal Transduction , T-Lymphocytes, Cytotoxic , Triple Negative Breast Neoplasms , Humans , Proto-Oncogene Proteins c-akt/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/immunology , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Receptor/metabolism , Cell Line, Tumor , Female , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Interleukin-23 Subunit p19/metabolism , Animals , Mice , Antibodies, Monoclonal/pharmacology
13.
Int J Pharm ; 660: 124303, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38848801

ABSTRACT

Although the combination of anti-vascular strategy plus immunotherapy has emerged as the optimal first-line treatment of hepatocellular carcinoma, lack of tumor targeting leads to low antitumor efficacy and serious side effect. Here, we report an ultra-pH-sensitive nanoparticle of gambogenic acid (GNA) encapsulated by poly(ethylene glycol)-poly(2-azepane ethyl methacrylate) (PEG-PAEMA) for tumor-targeting combined therapy of anti-vascular strategy plus immunotherapy. PEG-PAEMA-GNA nanoparticle was quite stable at pH 7.4 for 30 d. In contrast, it exerted size shrinkage, charge reversal and the release of GNA at pH 6.7 within 24 h. Moreover, PEG-PAEMA-GNA significantly enhanced the anti-vascular activity, membrane-disruptive capability and pro-apoptosis when pH changed from 7.4 to 6.7. Western blot analysis exhibits that PEG-PAEMA and its GNA nanoparticle facilitated the phosphorylation of STING protein. In vivo assays show that PEG-PAEMA-GNA not only displayed much higher tumor inhibition of 92 % than 37 % of free GNA, but also inhibited tumor vasculature, promoted the maturation of dendritic cells and recruited more cytotoxic t-lymphocytes for sufficient anti-vascular therapy and immunotherapy. All these results demonstrate that PEG-PAEMA-GNA displayed tumor-targeting combined treatment of anti-vascular therapy and immunotherapy. This study offers a simple and novel method for the combination of anti-vascular therapy and immunotherapy with high selectivity towards tumor.


Subject(s)
Immunotherapy , Nanoparticles , Polyethylene Glycols , Xanthenes , Animals , Immunotherapy/methods , Nanoparticles/chemistry , Hydrogen-Ion Concentration , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Xanthenes/chemistry , Xanthenes/administration & dosage , Xanthenes/pharmacology , Cell Line, Tumor , Humans , Mice , Apoptosis/drug effects , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/therapy , Liver Neoplasms/drug therapy , Mice, Inbred C57BL , Dendritic Cells/drug effects , Dendritic Cells/immunology , Mice, Inbred BALB C , Xanthones/chemistry , Xanthones/administration & dosage , Xanthones/pharmacology , Drug Liberation , Human Umbilical Vein Endothelial Cells , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects
14.
Adv Mater ; 36(33): e2401974, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38889229

ABSTRACT

Currently, sonodynamic therapy (SDT) has limited therapeutic outcomes and immune responses, highlighting the urgent need for enhanced strategies that can stimulate robust and long-lasting antitumor effects. Microcystis, a notorious microalga, reveals the possibility of mediating SDT owing to the presence of gas vesicles (GVs) and phycocyanin (PC). Herein, a nontoxic strain of Microcystis elabens (labeled Me) is developed as a novel agent for SDT because it generates O2 under red light (RL) illumination, while GVs and PC act as cavitation nuclei and sonosensitizers, respectively. Moreover, algal debris is released after ultrasound (US) irradiation, which primes the Toll-like receptor pathway to initiate a cascade of immune responses. This sono-immune strategy inhibits CT26 colon tumor growth largely by promoting dendritic cell (DC) maturation and cytotoxic T-cell activation. After combination with the immune checkpoint blockade (ICB), the therapeutic outcome is further amplified, accompanied by satisfactory abscopal and immune memory effects; the similar potency is proven in the "cold" 4T1 triple-negative breast tumor. In addition, Me exhibits good biosafety without significant acute or chronic toxicity. Briefly, this study turns waste into wealth by introducing sono-immunotherapy based on Microcystis that achieved encouraging therapeutic effects on cancer, which is expected to be translated into the clinic.


Subject(s)
Microcystis , Animals , Mice , Cell Line, Tumor , Ultrasonic Therapy/methods , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Phycocyanin/chemistry , Phycocyanin/pharmacology , Immunotherapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Colonic Neoplasms/therapy , Colonic Neoplasms/immunology
15.
Respir Res ; 25(1): 240, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867225

ABSTRACT

BACKGROUND: Despite the success of antiretroviral therapy (ART), people living with HIV (PLWH) suffer from a high burden of pulmonary diseases, even after accounting for their smoking status. Cytotoxic CD8 T-cells are likely implicated in this phenomenon and may act as a double-edged sword. While being essential in viral infection control, their hyperactivation can also contribute to lung mucosal tissue damage. The effects of HIV and smoking on pulmonary mucosal CD8 T-cell dynamics has been a neglected area of research, which we address herein. METHODS: Bronchoalveolar lavage (BAL) fluid were obtained from ART-treated PLWH (median duration of supressed viral load: 9 years; smokers: n = 14; non-smokers: n = 21) and HIV-uninfected controls (smokers: n = 11; non-smokers: n = 20) without any respiratory symptoms or active infection. Lymphocytes were isolated and CD8 T-cell subsets and homing markers were characterized by multiparametric flow cytometry. RESULTS: Both smoking and HIV infection were independently associated with a significant increase in frequencies of total pulmonary mucosal CD8 T-cell. BAL CD8 T-cells were primarily CD69 + expressing CD103 and/or CD49a, at least one of the two granzymes (GzmA/GzmB), and little Perforin. Higher expression levels of CD103, CD69, and GzmB were observed in smokers versus non-smokers. The ex vivo phenotype of GzmA + and GzmB + cells revealed increased expression of CD103 and CXCR6 in smokers, while PLWH displayed elevated levels of CX3CR1 compared to controls. CONCLUSION: Smoking and HIV could promote cytotoxic CD8 T-cell retention in small airways through different mechanisms. Smoking likely increases recruitment and retention of GzmB + CD8 Trm via CXCR6 and CD103. Heightened CX3CR1 expression could be associated with CD8 non-Trm recruitment from the periphery in PLWH.


Subject(s)
HIV Infections , Humans , Male , HIV Infections/drug therapy , HIV Infections/immunology , Female , Middle Aged , Adult , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism , Smoking/adverse effects , Bronchoalveolar Lavage Fluid/immunology , Anti-Retroviral Agents/therapeutic use , Anti-HIV Agents/therapeutic use , Lung/immunology , Lung/drug effects , Lung/metabolism
16.
Int J Biol Macromol ; 269(Pt 2): 132177, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729484

ABSTRACT

Tumor vaccine, which can effectively prevent tumor recurrence and metastasis, is a promising tool in tumor immunotherapy. However, heterogeneity of tumors and the inability to achieve a cascade effect limit the therapeutic effects of most developing tumor vaccine. We have developed a cascading immunoinducible in-situ mannose-functionalized polydopamine loaded with imiquimod phenylboronic hyaluronic acid nanocomposite gel vaccine (M/P-PDA@IQ PHA) through a boronic ester-based reaction. This reaction utilizes mannose-functionalized polydopamine loaded with imiquimod (M/P-PDA@IQ NAs) as a cross-linking agent to react with phenylboronic-grafted hyaluronic acid. Under near-infrared light irradiation, the M/P-PDA@IQ PHA caused local hyperthermia to trigger immunogenic cell death of tumor cells and tumor-associated antigens (TAAs) releasing. Subsequently, the M/P-PDA@IQ NAs which were gradually released by the pH/ROS/GSH-triggered degradation of M/P-PDA@IQ PHA, could capture and deliver these TAAs to lymph nodes. Finally, the M/P-PDA@IQ NAs facilitated maturation and cross-presentation of dendritic cells, as well as activation of cytotoxic T lymphocytes. Overall, the M/P-PDA@IQ PHA could serve as a novel in situ vaccine to stimulate several key nodes including TAAs release and capture, targeting lymph nodes and enhanced dendritic cells uptake and maturation as well as T cells activation. This cascading immune activation strategy can effectively elicit antitumor immune response.


Subject(s)
Cancer Vaccines , Hyaluronic Acid , Hydrogels , Indoles , Nanoparticles , Polymers , Hyaluronic Acid/chemistry , Polymers/chemistry , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Indoles/chemistry , Indoles/pharmacology , Animals , Mice , Hydrogels/chemistry , Nanoparticles/chemistry , Humans , Imiquimod/chemistry , Imiquimod/pharmacology , Dendritic Cells/immunology , Vaccination , Cell Line, Tumor , Immunotherapy/methods , Cross-Linking Reagents/chemistry , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects
17.
Adv Mater ; 36(29): e2400196, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734875

ABSTRACT

The activation of sequential events in the cancer-immunity cycle (CIC) is crucial for achieving effective antitumor immunity. However, formidable challenges, such as innate and adaptive immune resistance, along with the off-target adverse effects of nonselective immunomodulators, persist. In this study, a tumor-selective nano-regulator named PNBJQ has been presented, focusing on targeting two nonredundant immune nodes: inducing immunogenic cancer cell death and abrogating immune resistance to fully activate endogenous tumor immunity. PNBJQ is obtained by encapsulating the immunomodulating agent JQ1 within a self-assembling system formed by linking a Type-I photosensitizer to polyethylene glycol through a hypoxia-sensitive azo bond. Benefiting from the Type-I photosensitive mechanism, PNBJQ triggers the immunogenic cell death of hypoxic tumors under near-infrared (NIR) light irradiation. This process resolves innate immune resistance by stimulating sufficient cytotoxic T-lymphocytes. Simultaneously, PNBJQ smartly responds to the hypoxic tumor microenvironment for precise drug delivery, adeptly addressing adaptive immune resistance by using JQ1 to downregulate programmed death ligand 1 (PD-L1) and sustaining the response of cytotoxic T lymphocytes. The activatable synergic photoimmunotherapy promotes an immune-promoting tumor microenvironment by activating an iterative revolution of the CIC, which remarkably eradicates established hypoxic tumors and suppresses distal lesions under low light dose irradiation.


Subject(s)
Nanoparticles , Photosensitizing Agents , Animals , Mice , Cell Line, Tumor , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Nanoparticles/chemistry , Tumor Microenvironment/drug effects , Triazoles/chemistry , Triazoles/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/pathology , Immunotherapy , Azepines/chemistry , Azepines/pharmacology , Polyethylene Glycols/chemistry , Tumor Hypoxia/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , B7-H1 Antigen/metabolism , Infrared Rays
18.
Neurol Neuroimmunol Neuroinflamm ; 11(4): e200250, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38662990

ABSTRACT

BACKGROUND AND OBJECTIVES: The role of B cells in the pathogenic events leading to relapsing multiple sclerosis (R-MS) has only been recently elucidated. A pivotal step in defining this role has been provided by therapeutic efficacy of anti-CD20 monoclonal antibodies. Indeed, treatment with anti-CD20 can also alter number and function of other immune cells not directly expressing CD20 on their cell surface, whose activities can contribute to unknown aspects influencing therapeutic efficacy. We examined the phenotype and function of cytotoxic lymphocytes and Epstein-Barr virus (EBV)-specific immune responses in people with R-MS before and after ocrelizumab treatment. METHODS: In this prospective study, we collected blood samples from people with R-MS (n = 41) before and 6 and 12 months after initiating ocrelizumab to assess the immune phenotype and the indirect impact on cytotoxic functions of CD8+ T and NK cells. In addition, we evaluated the specific anti-EBV proliferative responses of both CD8+ T and NK lymphocytes as surrogate markers of anti-EBV activity. RESULTS: We observed that while ocrelizumab depleted circulating B cells, it also reduced the expression of activation and migratory markers on both CD8+ T and NK cells as well as their in vitro cytotoxic activity. A comparable pattern in the modulation of immune molecules by ocrelizumab was observed in cytotoxic cells even when patients with R-MS were divided into groups based on their prior disease-modifying treatment. These effects were accompanied by a significant and selective reduction of CD8+ T-cell proliferation in response to EBV antigenic peptides. DISCUSSION: Taken together, our findings suggest that ocrelizumab-while depleting B cells-affects the cytotoxic function of CD8+ and NK cells, whose reduced cross-activity against myelin antigens might also contribute to its therapeutic efficacy during MS.


Subject(s)
Antibodies, Monoclonal, Humanized , CD8-Positive T-Lymphocytes , Herpesvirus 4, Human , Immunologic Factors , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Female , Adult , Male , Herpesvirus 4, Human/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Middle Aged , Immunologic Factors/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/blood , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Prospective Studies , Cell Proliferation/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects
19.
J Control Release ; 369: 556-572, 2024 May.
Article in English | MEDLINE | ID: mdl-38580136

ABSTRACT

Vaccines represent one of the most powerful and cost-effective innovations for controlling a wide range of infectious diseases caused by various viruses and bacteria. Unlike mRNA and DNA-based vaccines, subunit vaccines carry no risk of insertional mutagenesis and can be lyophilized for convenient transportation and long-term storage. However, existing adjuvants are often associated with toxic effect and reactogenicity, necessitating expanding the repertoire of adjuvants with better biocompatibility, for instance, designing self-adjuvating polymeric carriers. We herein report a novel subunit vaccine delivery platform constructed via in situ free radical polymerization of C7A (2-(Hexamethyleneimino) ethyl methacrylate) and acrylamide around the surface of individual protein antigens. Using ovalbumin (OVA) as a model antigen, we observed substantial increases in both diameter (∼70 nm) and surface potential (-1.18 mV) following encapsulation, referred to as n(OVA)C7A. C7A's ultra pH sensitivity with a transition pH around 6.9 allows for rapid protonation in acidic environments. This property facilitates crucial processes such as endosomal escape and major histocompatibility complex (MHC)-I-mediated antigen presentation, culminating in the substantial CD8+ T cell activation. Additionally, compared to OVA nanocapsules without the C7A components and native OVA without modifications, we observed heightened B cell activation within the germinal center, along with remarkable increases in serum antibody and cytokine production. It's important to note that mounting evidence suggests that adjuvant effects, particularly its targeted stimulation of type I interferons (IFNs), can contribute to advantageous adaptive immune responses. Beyond its exceptional potency, the nanovaccine also demonstrated robust formation of immune memory and exhibited a favorable biosafety profile. These findings collectively underscore the promising potential of our nanovaccine in the realm of immunotherapy and vaccine development.


Subject(s)
Mice, Inbred C57BL , Ovalbumin , T-Lymphocytes, Cytotoxic , Animals , Ovalbumin/immunology , Ovalbumin/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Female , Methacrylates/chemistry , Polymers/chemistry , Polymers/administration & dosage , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice , Vaccines/administration & dosage , Vaccines/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Nanovaccines
20.
J Control Release ; 369: 296-308, 2024 May.
Article in English | MEDLINE | ID: mdl-38301925

ABSTRACT

Immunosuppression caused by incomplete radiofrequency ablation (iRFA) is a crucial factor affecting the effectiveness of RFA for solid tumors. However, little is known about the changes iRFA induces in the tumor immune microenvironment (TIME) of hepatocellular carcinoma (HCC), the primary application area for RFA. In this study, we found iRFA promotes a suppressive TIME in residual HCC tumors, characterized by M2 macrophage polarization, inhibited antigen presentation by dendritic cells (DCs), and reduced infiltration of cytotoxic T lymphocytes (CTLs). Interestingly, the STING agonist MSA-2 was able to reorganize M2-like tumor-promoting macrophages into M1-like anti-tumor states and enhance antigen presentation by DCs. To optimize the therapeutic effect of MSA-2, we used a calcium ion (Ca2+) responsive sodium alginate (ALG) as a carrier, forming an injectable hydrogel named ALG@MSA-2. This hydrogel can change from liquid to gel, maintaining continuous drug release in situ. Our results suggested that ALG@MSA-2 effectively activated anti-tumor immunity, as manifested by increased M1-like macrophage polarization, enhanced antigen presentation by DCs, increased CTL infiltration, and inhibited residual tumor growth. ALG@MSA-2 also resulted in a complete regression of contralateral tumors and widespread liver metastases in vivo. In addition, the excellent biosafety of ALG@MSA-2 was also proved by blood biochemical analysis and body weight changes in mice. In summary, this study demonstrated that the immune cascade of ALG@MSA-2 mediated the STING pathway activation and promoted a favorable TIME which might provide novel insights for the RFA treatment of HCC.


Subject(s)
Alginates , Carcinoma, Hepatocellular , Hydrogels , Liver Neoplasms , Membrane Proteins , Mice, Inbred C57BL , Radiofrequency Ablation , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Hydrogels/administration & dosage , Radiofrequency Ablation/methods , Alginates/chemistry , Alginates/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/immunology , Cell Line, Tumor , Macrophages/drug effects , Macrophages/immunology , Tumor Microenvironment/drug effects , Mice , Male , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL