Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nature ; 624(7990): 154-163, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37968405

ABSTRACT

CD8+ cytotoxic T cells (CTLs) orchestrate antitumour immunity and exhibit inherent heterogeneity1,2, with precursor exhausted T (Tpex) cells but not terminally exhausted T (Tex) cells capable of responding to existing immunotherapies3-7. The gene regulatory network that underlies CTL differentiation and whether Tex cell responses can be functionally reinvigorated are incompletely understood. Here we systematically mapped causal gene regulatory networks using single-cell CRISPR screens in vivo and discovered checkpoints for CTL differentiation. First, the exit from quiescence of Tpex cells initiated successive differentiation into intermediate Tex cells. This process is differentially regulated by IKAROS and ETS1, the deficiencies of which dampened and increased mTORC1-associated metabolic activities, respectively. IKAROS-deficient cells accumulated as a metabolically quiescent Tpex cell population with limited differentiation potential following immune checkpoint blockade (ICB). Conversely, targeting ETS1 improved antitumour immunity and ICB efficacy by boosting differentiation of Tpex to intermediate Tex cells and metabolic rewiring. Mechanistically, TCF-1 and BATF are the targets for IKAROS and ETS1, respectively. Second, the RBPJ-IRF1 axis promoted differentiation of intermediate Tex to terminal Tex cells. Accordingly, targeting RBPJ enhanced functional and epigenetic reprogramming of Tex cells towards the proliferative state and improved therapeutic effects and ICB efficacy. Collectively, our study reveals that promoting the exit from quiescence of Tpex cells and enriching the proliferative Tex cell state act as key modalities for antitumour effects and provides a systemic framework to integrate cell fate regulomes and reprogrammable functional determinants for cancer immunity.


Subject(s)
Cell Differentiation , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Mutagenesis , Neoplasms , Single-Cell Analysis , T-Lymphocytes, Cytotoxic , Humans , Cell Differentiation/drug effects , Cell Differentiation/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/genetics , Neoplasms/immunology , Single-Cell Analysis/methods , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
2.
Nature ; 620(7972): 200-208, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37407815

ABSTRACT

Cancer cells evade T cell-mediated killing through tumour-immune interactions whose mechanisms are not well understood1,2. Dendritic cells (DCs), especially type-1 conventional DCs (cDC1s), mediate T cell priming and therapeutic efficacy against tumours3. DC functions are orchestrated by pattern recognition receptors3-5, although other signals involved remain incompletely defined. Nutrients are emerging mediators of adaptive immunity6-8, but whether nutrients affect DC function or communication between innate and adaptive immune cells is largely unresolved. Here we establish glutamine as an intercellular metabolic checkpoint that dictates tumour-cDC1 crosstalk and licenses cDC1 function in activating cytotoxic T cells. Intratumoral glutamine supplementation inhibits tumour growth by augmenting cDC1-mediated CD8+ T cell immunity, and overcomes therapeutic resistance to checkpoint blockade and T cell-mediated immunotherapies. Mechanistically, tumour cells and cDC1s compete for glutamine uptake via the transporter SLC38A2 to tune anti-tumour immunity. Nutrient screening and integrative analyses show that glutamine is the dominant amino acid in promoting cDC1 function. Further, glutamine signalling via FLCN impinges on TFEB function. Loss of FLCN in DCs selectively impairs cDC1 function in vivo in a TFEB-dependent manner and phenocopies SLC38A2 deficiency by eliminating the anti-tumour therapeutic effect of glutamine supplementation. Our findings establish glutamine-mediated intercellular metabolic crosstalk between tumour cells and cDC1s that underpins tumour immune evasion, and reveal glutamine acquisition and signalling in cDC1s as limiting events for DC activation and putative targets for cancer treatment.


Subject(s)
Amino Acid Transport System A , Dendritic Cells , Glutamine , Neoplasms , Signal Transduction , Amino Acid Transport System A/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glutamine/metabolism , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism
3.
Cell Metab ; 35(1): 3-5, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36599301

ABSTRACT

Metabolic communication in the tumor microenvironment underscores tumor-immune interactions and affects anti-tumor immunity, yet cell-extrinsic signals driving tumor metabolic remodeling are incompletely understood. In this issue, Tsai et al. show that during initial tumorigenesis, T cell-derived IFNγ triggers STAT3 activation and c-Myc-dependent alterations of tumor cell metabolism, which potentiates immune evasion.


Subject(s)
Neoplasms , Humans , Neoplasms/metabolism , Carcinogenesis , Tumor Microenvironment
4.
EBioMedicine ; 86: 104333, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36335665

ABSTRACT

BACKGROUND: Targeting helper T cells, especially Th17 cells, has become a plausible therapy for many autoimmune diseases. METHODS: Using an in vitro culture system, we screened an epigenetics compound library for inhibitors of IFN-γ and IL-17 expression in murine Th1 and Th17 cultures. FINDINGS: This identified IOX1 as an effective suppressor of IL-17 expression in both murine and human CD4+ T cells. Furthermore, we found that IOX1 suppresses Il17a expression directly by targeting TET2 activity on its promoter in Th17 cells. Using established pre-clinical models of intraocular inflammation, treatment with IOX1 in vivo reduced the migration/infiltration of Th17 cells into the site of inflammation and tissue damage. INTERPRETATION: These results provide evidence of the strong potential for IOX1 as a viable therapy for inflammatory diseases, in particular of the eye. FUNDING: This study was supported by the National Key Research and Development Program of China 2021YFA1101200 (2021YFA1101204) to LW and XW; the National Natural Science Foundation of China 81900844 to XH and 82171041 to LW; the China Postdoctoral Science Foundation 2021M700776 and the Scientific Research Project of Guangdong Provincial Bureau of Traditional Chinese Medicine 20221373 to YZ; and the National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS (National Health Service) Foundation Trust and University College London Institute of Ophthalmology, UK (DAC, LPS, PJPL, MS, ADD and RWJL). The views expressed are those of the authors and not necessarily those of the NIHR or the UK's Department of Health and Social Care.


Subject(s)
Dioxygenases , Th17 Cells , Animals , Humans , Mice , Cell Differentiation , Dioxygenases/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Inflammation/drug therapy , Inflammation/genetics , Interleukin-17/metabolism , State Medicine , Th1 Cells
5.
Nature ; 607(7917): 135-141, 2022 07.
Article in English | MEDLINE | ID: mdl-35732731

ABSTRACT

The identification of mechanisms to promote memory T (Tmem) cells has important implications for vaccination and anti-cancer immunotherapy1-4. Using a CRISPR-based screen for negative regulators of Tmem cell generation in vivo5, here we identify multiple components of the mammalian canonical BRG1/BRM-associated factor (cBAF)6,7. Several components of the cBAF complex are essential for the differentiation of activated CD8+ T cells into T effector (Teff) cells, and their loss promotes Tmem cell formation in vivo. During the first division of activated CD8+ T cells, cBAF and MYC8 frequently co-assort asymmetrically to the two daughter cells. Daughter cells with high MYC and high cBAF display a cell fate trajectory towards Teff cells, whereas those with low MYC and low cBAF preferentially differentiate towards Tmem cells. The cBAF complex and MYC physically interact to establish the chromatin landscape in activated CD8+ T cells. Treatment of naive CD8+ T cells with a putative cBAF inhibitor during the first 48 h of activation, before the generation of chimeric antigen receptor T (CAR-T) cells, markedly improves efficacy in a mouse solid tumour model. Our results establish cBAF as a negative determinant of Tmem cell fate and suggest that manipulation of cBAF early in T cell differentiation can improve cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Differentiation , DNA Helicases , Multiprotein Complexes , Nuclear Proteins , Proto-Oncogene Proteins c-myc , Transcription Factors , Animals , CD8-Positive T-Lymphocytes/cytology , DNA Helicases/metabolism , Disease Models, Animal , Immunologic Memory , Immunotherapy , Memory T Cells/cytology , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Neoplasms , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Chimeric Antigen , Transcription Factors/metabolism
6.
EMBO Mol Med ; 14(1): e14502, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34898004

ABSTRACT

Impressive clinical benefit is seen in clinic with PD-1 inhibitors on portion of cancer patients. Yet, there remains an urgent need to develop effective synergizers to expand their clinical application. Tumor-associated macrophage (TAM), a type of M2-polarized macrophage, eliminates or suppresses T-cell-mediated anti-tumor responses. Transforming TAMs into M1 macrophages is an attractive strategy of anti-tumor therapy. Here, we conducted a high-throughput screening and found that Carfilzomib potently drove M2 macrophages to express M1 cytokines, phagocytose tumor cells, and present antigens to T cells. Mechanistically, Carfilzomib elicited unfolded protein response (UPR), activated IRE1α to recruit TRAF2, and activated NF-κB to transcribe genes encoding M1 markers in M2 macrophages. In vivo, Carfilzomib effectively rewired tumor microenvironment through reprogramming TAMs into M1-like macrophages and shrank autochthonous lung cancers in transgenic mouse model. More importantly, Carfilzomib synergized with PD-1 antibody to almost completely regress autochthonous lung cancers. Given the safety profiles of Carfilzomib in clinic, our work suggested a potentially immediate application of combinational treatment with Carfilzomib and PD-1 inhibitors for patients with solid tumors.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms , Oligopeptides/pharmacology , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Animals , Cellular Reprogramming , Endoribonucleases , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases
7.
Cancer Immunol Res ; 9(4): 371-385, 2021 04.
Article in English | MEDLINE | ID: mdl-33608256

ABSTRACT

Immunologic checkpoint blockade has been proven effective in a variety of malignancies. However, high rates of resistance have substantially hindered its clinical use. Understanding the underlying mechanisms may lead to new strategies for improving therapeutic efficacy. Although a number of signaling pathways have been shown to be associated with tumor cell-mediated resistance to immunotherapy, T cell-intrinsic resistant mechanisms remain elusive. Here, we demonstrated that diacylglycerol kinase alpha (Dgka) mediated T-cell dysfunction during anti-PD-1 therapy by exacerbating the exhaustion of reinvigorated tumor-specific T cells. Pharmacologic ablation of Dgka postponed T-cell exhaustion and delayed development of resistance to PD-1 blockade. Dgka inhibition also enhanced the efficacy of anti-PD-1 therapy. We further found that the expression of DGKA in cancer cells promoted tumor growth via the AKT signaling pathway, suggesting that DGKA might be a target in tumor cells as well. Together, these findings unveiled a molecular pathway mediating resistance to PD-1 blockade and provide a potential therapeutic strategy with combination immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Diacylglycerol Kinase/metabolism , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Diacylglycerol Kinase/antagonists & inhibitors , Drug Resistance, Neoplasm , Humans , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/immunology
8.
Food Chem ; 278: 77-83, 2019 Apr 25.
Article in English | MEDLINE | ID: mdl-30583441

ABSTRACT

Phenolic acids perform biological effects which are largely influenced by their binding to serum albumin. Therefore, investigating structure-affinity relationship of binding between phenolic acids and serum albumin is important. In this study, 114 phenolic acids and their derivatives, sharing the benzoic acid core with different substituents groups, were selected to investigate structure-affinity relationships with bovine serum albumin. The binding constants were obtained through fluorescence quenching, and a comprehensive mathematical model with inner-filter effect correction was applied. The results showed that the hydroxy group at the 2-position led to stronger binding affinity, while it had a negative influence at the 4-position. Substituting hydroxy groups with methoxy groups at 4-position and with methyl groups at 3-position both strengthened the binding affinity, respectively. Hydrogen bonding was one of the key binding forces for this binding interaction. Our findings provide a fundamental insight on the binding mechanism of phenolic acids to bovine serum albumin.


Subject(s)
Hydroxybenzoates/chemistry , Serum Albumin, Bovine/chemistry , Animals , Benzoic Acid/chemistry , Cattle , Hydrogen Bonding , Hydroxybenzoates/metabolism , Methylation , Protein Binding , Serum Albumin, Bovine/metabolism
9.
Cell Death Dis ; 8(6): e2835, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28569774

ABSTRACT

The M2 isoform of pyruvate kinase (PKM2) is a key driver of glycolysis in cancer cells and has critical 'non-metabolic' functions in some cancers; however, the role of PKM2 in pancreatic cancer remains unclear. The aim of the current study was to elucidate the role of PKM2 in pancreatic cancer progression and the potential of PKM2 as a therapeutic target. In this study, we observed that PKM2 is highly expressed in patients with pancreatic cancer and is correlated to survival. Elevated PKM2 expression promoted cell proliferation, migration and tumor formation. The inhibition of cell growth by silencing PKM2 is caused by impairment of the autophagy process. To test the potential effects of downregulating PKM2 as a clinical therapy, we constructed an RGD-modified oncolytic adenovirus containing shPKM2 (OAd.R.shPKM2) to knock down PKM2 in pancreatic cancer cells. Cells transduced with OAd.R.shPKM2 exhibited decreased cell viability, and, in a PANC-1 xenograft model, intratumoral injection of OAd.R.shPKM2 resulted in reduced tumor growth. Furthermore, OAd.R.shPKM2 induced apoptosis and impaired autophagy in PANC-1 cells. Our results suggested that targeting PKM2 with an oncolytic adenovirus produced a strong antitumor effect, and that this strategy could broaden the therapeutic options for treating pancreatic cancer.


Subject(s)
Adenoviridae/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , RNA, Small Interfering/genetics , Thyroid Hormones/genetics , Adenoviridae/metabolism , Aged , Animals , Autophagy , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , HEK293 Cells , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice, Nude , Middle Aged , Molecular Targeted Therapy/methods , Neoplasm Staging , Oligopeptides/genetics , Oligopeptides/metabolism , Oncolytic Viruses/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Thyroid Hormones/metabolism , Xenograft Model Antitumor Assays , Thyroid Hormone-Binding Proteins
10.
Hum Gene Ther ; 28(8): 690-700, 2017 08.
Article in English | MEDLINE | ID: mdl-28530127

ABSTRACT

SNORD44 is a C/D box small nucleolar RNA, and exhibits low expression in breast cancer and head and neck squamous cell carcinoma tissues. Its host gene is growth arrest specific transcript 5 (GAS5), which is a long noncoding RNA. GAS5 is downregulated in colorectal cancer (CRC), and overexpression of GAS5 suppresses cell proliferation. However, the function of SNORD44 in CRC remains largely unknown, and the application of SNORD44 combined with GAS5 in CRC treatment has not been reported. In this study, the expression levels of SNORD44 and GAS5 were measured in CRC tissues by quantitative RT-PCR. The correlation between SNORD44 and GAS5 was evaluated by Pearson correlation analysis. An oncolytic adenovirus expressing SNORD44 and GAS5 (SPDD-UG) was constructed. The biological effects of SPDD-UG were investigated in CRC cell line SW620 and LS174T in vitro and in xenografts. The synergistic effect of rapamycin and SPDD-UG was explored in SW620 and LS174T cells and tumors. We demonstrated that SNORD44 expression level was markedly decreased in CRC tissues and positively correlated with GAS5 expression. SPDD-UG significantly inhibited SW620 and LS174T cell growth and induced cell apoptosis. Intratumoral injection of SPDD-UG significantly suppressed xenografts growth in nude mice. Moreover, the mechanistic target of rapamycin (mTOR) inhibitor, rapamycin, enhanced the antitumor effect through antagonizing the PI3K/Akt pathway activated by SPDD-UG. These results suggest that overexpression of SNORD44 and GAS5 by oncolytic adenovirus provides a promising method for CRC therapy.


Subject(s)
Adenoviridae/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genetic Vectors/genetics , Oncolytic Viruses/genetics , RNA, Long Noncoding/genetics , RNA, Small Nucleolar/genetics , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Disease Models, Animal , Gene Expression , Gene Order , Gene Transfer Techniques , Genetic Therapy , Humans , Lentivirus/genetics , Mice , Signal Transduction/drug effects , Sirolimus/pharmacology , Tumor Burden , Xenograft Model Antitumor Assays
11.
J Mol Cell Biol ; 9(3): 243-255, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27913571

ABSTRACT

Small nucleolar RNA (snoRNA) dysfunctions have been associated with cancer development. SNORD126 is an orphan C/D box snoRNA that is encoded within introns 5-6 of its host gene, cyclin B1-interacting protein 1 (CCNB1IP1). The cancer-associated molecular mechanisms triggered by SNORD126 are not fully understood. Here, we demonstrate that SNORD126 is highly expressed in hepatocellular carcinoma (HCC) and colorectal cancer (CRC) patient samples. SNORD126 increased Huh-7 and SW480 cell growth and tumorigenicity in nude mice. Knockdown of SNORD126 inhibited HepG2 and LS174T cell growth. We verified that SNORD126 was not processed into small RNAs with miRNA activity. Moreover, SNORD126 did not show a significant expression correlation with CCNB1IP1 in HCC samples or regulate CCNB1IP1 expression. Our gene expression profile analysis indicated that SNORD126-upregulated genes frequently mapped to the PI3K-AKT pathway. SNORD126 overexpression increased the levels of phosphorylated AKT, GSK-3ß, and p70S6K and elevated fibroblast growth factor receptor 2 (FGFR2) expression. siRNA-mediated knockdown or AZD4547-mediated inactivation of FGFR2 in SNORD126-overexpressing Huh-7 cells inhibited AKT phosphorylation and suppressed cell growth. These findings indicate an oncogenic role for SNORD126 in cancer and suggest its potential as a therapeutic target.


Subject(s)
Carcinoma, Hepatocellular/pathology , Colorectal Neoplasms/pathology , Liver Neoplasms/pathology , RNA, Small Nucleolar/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice, Nude , Middle Aged , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Nucleolar/genetics , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 23(8): 2038-2049, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27683179

ABSTRACT

Purpose: Antiproliferative, antiviral, and immunomodulatory activities of endogenous type I IFNs (IFN1) prompt the design of recombinant IFN1 for therapeutic purposes. However, most of the designed IFNs exhibited suboptimal therapeutic efficacies against solid tumors. Here, we report evaluation of the in vitro and in vivo antitumorigenic activities of a novel recombinant IFN termed sIFN-I.Experimental Design: We compared primary and tertiary structures of sIFN-I with its parental human IFNα-2b, as well as affinities of these ligands for IFN1 receptor chains and pharmacokinetics. These IFN1 species were also compared for their ability to induce JAK-STAT signaling and expression of the IFN1-stimulated genes and to elicit antitumorigenic effects. Effects of sIFN-I on tumor angiogenesis and immune infiltration were also tested in transplanted and genetically engineered immunocompetent mouse models.Results: sIFN-I displayed greater affinity for IFNAR1 (over IFNAR2) chain of the IFN1 receptor and elicited a greater extent of IFN1 signaling and expression of IFN-inducible genes in human cells. Unlike IFNα-2b, sIFN-I induced JAK-STAT signaling in mouse cells and exhibited an extended half-life in mice. Treatment with sIFN-I inhibited intratumoral angiogenesis, increased CD8+ T-cell infiltration, and robustly suppressed growth of transplantable and genetically engineered tumors in immunodeficient and immunocompetent mice.Conclusions: These findings define sIFN-I as a novel recombinant IFN1 with potent preclinical antitumorigenic effects against solid tumor, thereby prompting the assessment of sIFN-I clinical efficacy in humans. Clin Cancer Res; 23(8); 2038-49. ©2016 AACR.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Interferon-alpha/chemistry , Interferon-alpha/pharmacology , Animals , Female , Flow Cytometry , Humans , Immunoblotting , Interferon alpha-2 , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/drug therapy , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , Xenograft Model Antitumor Assays
13.
ACS Nano ; 10(12): 11548-11560, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27977128

ABSTRACT

Oncolytic adenovirus (OncoAd) is a promising therapeutic agent for treating cancer. However, the therapeutic potential of OncoAd is hindered by hepatic sequestration and the host immune response in vivo. Here, we constructed a PEG/Lipids/calcium phosphate (CaP)-OncoAd (PLC-OncoAd) delivery system for ZD55-IL-24, an oncolytic adenovirus that carries the IL-24 gene. The negatively charged PLC-ZD55-IL-24 were disperse and resisted serum-induced aggregation. Compared to naked ZD55-IL-24, the systemic administration of PLC-ZD55-IL-24 in BALB/c mice resulted in reduced liver sequestration and systemic toxicity and evaded the innate immune response. In addition, masking the surface of OncoAd protected it from neutralization by pre-existing neutralizing antibody. PLC-OncoAd achieved efficient targeted delivery in Huh-7-bearing nude mice, and intravenous administration of a high dose of PLC-ZD55-IL-24 increased therapeutic efficacy without inducing toxicity. The developed PLC-OncoAd delivery system represents a promising improvement for oncolytic adenovirus-based cancer gene therapy in vivo.


Subject(s)
Calcium Phosphates , Genetic Therapy , Genetic Vectors , Lipids , Oncolytic Virotherapy , Adenoviridae , Animals , Cell Line, Tumor , Mice , Mice, Nude , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(37): 60491-60502, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27528029

ABSTRACT

Hepatocellular carcinoma (HCC) is characterized by alterations in multiple genes. High expression of CD147 on the surface of HCC cells promotes proliferation. The monoclonal antibody HAb18 recognizes CD147. We constructed an oncolytic adenoviral vector to express HAb18 (ZD55-HAb18) in HCC cells. Interleukin 24 (IL24) was co-expressed through the use of an F2A linker. ZD55-HAb18-IL24 decreased HCC cell viability to a greater degree than either ZD55-HAb18 or ZD55-IL24 alone. ZD55-HAb18-IL24 also induced apoptosis and autophagy in PLC/PRF/5 HCC cells. Intratumoral injection of ZD55-HAb18-IL24 repressed tumor growth in a PLC/PRF/5 xenograft model. Our results suggest that antibody-antitumor gene conjugation elicited a stronger antitumor effect than the antibody alone, and that this strategy could broaden the applications of antibody-based therapies in HCC.


Subject(s)
Antibodies, Monoclonal/metabolism , Carcinoma, Hepatocellular/therapy , Immunotherapy/methods , Interleukins/metabolism , Liver Neoplasms/therapy , Oncolytic Virotherapy , Adenoviridae/genetics , Antibodies, Monoclonal/genetics , Apoptosis , Autophagy , Basigin/immunology , Genetic Vectors/genetics , Humans , Interleukins/genetics , Tumor Cells, Cultured , Vaccines, DNA
16.
Biochem Biophys Res Commun ; 476(1): 21-8, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27208781

ABSTRACT

Lung cancer has an especially high incidence rate worldwide, and its resistance to cell death and chemotherapeutic drugs increases its intractability. The vaccinia virus has been shown to destroy neoplasm within a short time and disseminate rapidly and extensively as an enveloped virion throughout the circulatory system, and this virus has also demonstrated a strong ability to overexpress exogenous genes. Interleukin-24 (IL-24/mda-7) is an important cytokine that belongs to the activating caspase family and facilitates the inhibition of STAT3 when a cell enters the apoptosis pathway. In this study, we constructed a cancer-targeted vaccinia virus carrying the IL-24 gene knocked in the region of the viral thymidine kinase (TK) gene (VV-IL-24). Our results showed that VV-IL-24 efficiently infected and destroyed lung cancer cells via caspase-dependent apoptosis and decreased the expression of STAT3. In vivo, VV-IL-24 expressed IL-24 at a high level in the transplanted tumour, reduced STAT3 activity, and eventually led to apoptosis. In conclusion, we demonstrated that vv-IL-24 has the potential for use as a new human lung cancer treatment.


Subject(s)
Cancer Vaccines/genetics , Interleukins/genetics , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung/pathology , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Animals , Apoptosis , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lung/metabolism , Lung Neoplasms/genetics , Mice, Inbred C57BL , Mice, Nude , Oncolytic Virotherapy , STAT3 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...