Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 392, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36693842

ABSTRACT

Cancer immunotherapy that deploys the host's immune system to recognize and attack tumors, is a promising strategy for cancer treatment. However, its efficacy is greatly restricted by the immunosuppressive (i.e., immunologically cold) tumor microenvironment (TME). Here, we report an in-situ cryo-immune engineering (ICIE) strategy for turning the TME from immunologically "cold" into "hot". In particular, after the ICIE treatment, the ratio of the CD8+ cytotoxic T cells to the immunosuppressive regulatory T cells is increased by more than 100 times in not only the primary tumors with cryosurgery but also distant tumors without freezing. This is achieved by combining cryosurgery that causes "frostbite" of tumor with cold-responsive nanoparticles that not only target tumor but also rapidly release both anticancer drug and PD-L1 silencing siRNA specifically into the cytosol upon cryosurgery. This ICIE treatment leads to potent immunogenic cell death, which promotes maturation of dendritic cells and activation of CD8+ cytotoxic T cells as well as memory T cells to kill not only primary but also distant/metastatic breast tumors in female mice (i.e., the abscopal effect). Collectively, ICIE may enable an efficient and durable way to leverage the immune system for combating cancer and its metastasis.


Subject(s)
Antineoplastic Agents , Cryotherapy , Immunotherapy , Neoplasms , Tumor Microenvironment , Animals , Female , Mice , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Immunotherapy/methods , Nanotechnology/methods , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Cryotherapy/methods
3.
Nat Biomed Eng ; 5(11): 1320-1335, 2021 11.
Article in English | MEDLINE | ID: mdl-34725507

ABSTRACT

In breast cancer, genetic heterogeneity, the lack of actionable targets and immune evasion all contribute to the limited clinical response rates to immune checkpoint blockade therapy. Here, we report a high-throughput screen based on the functional interaction of mouse- or patient-derived breast tumour organoids and tumour-specific cytotoxic T cells for the identification of epigenetic inhibitors that promote antigen presentation and potentiate T-cell-mediated cytotoxicity. We show that the epigenetic inhibitors GSK-LSD1, CUDC-101 and BML-210, identified by the screen, display antitumour activities in orthotopic mammary tumours in mice, that they upregulate antigen presentation mediated by the major histocompatibility complex class I on breast tumour cells and that treatment with BML-210 substantially sensitized breast tumours to the inhibitor of the checkpoint programmed death-1. Standardized measurements of tumour-cell killing activity facilitated by tumour-organoid-T-cell screens may help with the identification of candidate immunotherapeutics for a range of cancers.


Subject(s)
Antigen Presentation , Breast Neoplasms , Animals , CD8-Positive T-Lymphocytes , Epigenesis, Genetic , Female , Humans , Mice , Organoids
4.
J Clin Invest ; 131(10)2021 05 17.
Article in English | MEDLINE | ID: mdl-33830945

ABSTRACT

One of the primary mechanisms of tumor cell immune evasion is the loss of antigenicity, which arises due to lack of immunogenic tumor antigens as well as dysregulation of the antigen processing machinery. In a screen for small-molecule compounds from herbal medicine that potentiate T cell-mediated cytotoxicity, we identified atractylenolide I (ATT-I), which substantially promotes tumor antigen presentation of both human and mouse colorectal cancer (CRC) cells and thereby enhances the cytotoxic response of CD8+ T cells. Cellular thermal shift assay (CETSA) with multiplexed quantitative mass spectrometry identified the proteasome 26S subunit non-ATPase 4 (PSMD4), an essential component of the immunoproteasome complex, as a primary target protein of ATT-I. Binding of ATT-I with PSMD4 augments the antigen-processing activity of immunoproteasome, leading to enhanced MHC-I-mediated antigen presentation on cancer cells. In syngeneic mouse CRC models and human patient-derived CRC organoid models, ATT-I treatment promotes the cytotoxicity of CD8+ T cells and thus profoundly enhances the efficacy of immune checkpoint blockade therapy. Collectively, we show here that targeting the function of immunoproteasome with ATT-I promotes tumor antigen presentation and empowers T cell cytotoxicity, thus elevating the tumor response to immunotherapy.


Subject(s)
Antigen Presentation/drug effects , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/pharmacology , Immunity, Cellular/drug effects , Immunotherapy , Lactones/pharmacology , Neoplasms, Experimental/therapy , Sesquiterpenes/pharmacology , Animals , Antigens, Neoplasm/genetics , HCT116 Cells , Humans , Immune Checkpoint Inhibitors/pharmacokinetics , Immunity, Cellular/genetics , Lactones/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Sesquiterpenes/pharmacokinetics
5.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32990678

ABSTRACT

Immune evasion is a pivotal event in tumor progression. To eliminate human cancer cells, current immune checkpoint therapy is set to boost CD8+ T cell-mediated cytotoxicity. However, this action is eventually dependent on the efficient recognition of tumor-specific antigens via T cell receptors. One primary mechanism by which tumor cells evade immune surveillance is to downregulate their antigen presentation. Little progress has been made toward harnessing potential therapeutic targets for enhancing antigen presentation on the tumor cell. Here, we identified MAL2 as a key player that determines the turnover of the antigen-loaded MHC-I complex and reduces the antigen presentation on tumor cells. MAL2 promotes the endocytosis of tumor antigens via direct interaction with the MHC-I complex and endosome-associated RAB proteins. In preclinical models, depletion of MAL2 in breast tumor cells profoundly enhanced the cytotoxicity of tumor-infiltrating CD8+ T cells and suppressed breast tumor growth, suggesting that MAL2 is a potential therapeutic target for breast cancer immunotherapy.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Myelin and Lymphocyte-Associated Proteolipid Proteins/immunology , Neoplasm Proteins/immunology , Tumor Escape , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Mice, Nude
6.
Arch Biochem Biophys ; 697: 108659, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33144083

ABSTRACT

Metabolic reprogramming confers cancer cells plasticity and viability under harsh conditions. Such active alterations lead to cell metabolic dependency, which can be exploited as an attractive target in development of effective antitumor therapies. Similar to cancer cells, activated T cells also execute global metabolic reprogramming for their proliferation and effector functions when recruited to the tumor microenvironment (TME). However, the high metabolic activity of rapidly proliferating cancer cells can compete for nutrients with immune cells in the TME, and consequently, suppressing their anti-tumor functions. Thus, therapeutic strategies could aim to restore T cell metabolism and anti-tumor responses in the TME by targeting the metabolic dependence of cancer cells. In this review, we highlight current research progress on metabolic reprogramming and the interplay between cancer cells and immune cells. We also discuss potential therapeutic intervention strategies for targeting metabolic pathways to improve cancer immunotherapy efficacy.


Subject(s)
Immunotherapy/methods , Neoplasms/metabolism , Neoplasms/therapy , Animals , Humans , Immunity , Neoplasms/immunology , Neoplasms/pathology
7.
JCI Insight ; 5(9)2020 05 07.
Article in English | MEDLINE | ID: mdl-32376804

ABSTRACT

Immune checkpoint blockade immunotherapy delivers promising clinical results in colorectal cancer (CRC). However, only a fraction of cancer patients develop durable responses. The tumor microenvironment (TME) negatively impacts tumor immunity and subsequently clinical outcomes. Therefore, there is a need to identify other checkpoint targets associated with the TME. Early-onset factors secreted by stromal cells as well as tumor cells often help recruit immune cells to the TME, among which are alarmins such as IL-33. The only known receptor for IL-33 is stimulation 2 (ST2). Here we demonstrated that high ST2 expression is associated with poor survival and is correlated with low CD8+ T cell cytotoxicity in CRC patients. ST2 is particularly expressed in tumor-associated macrophages (TAMs). In preclinical models of CRC, we demonstrated that ST2-expressing TAMs (ST2+ TAMs) were recruited into the tumor via CXCR3 expression and exacerbated the immunosuppressive TME; and that combination of ST2 depletion using ST2-KO mice with anti-programmed death 1 treatment resulted in profound growth inhibition of CRC. Finally, using the IL-33trap fusion protein, we suppressed CRC tumor growth and decreased tumor-infiltrating ST2+ TAMs. Together, our findings suggest that ST2 could serve as a potential checkpoint target for CRC immunotherapy.


Subject(s)
Colorectal Neoplasms/immunology , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-33/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Animals , Cell Line, Tumor , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Tumor-Associated Macrophages/cytology
8.
Nat Commun ; 9(1): 4718, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413718

ABSTRACT

Chromosome 17q23 amplification occurs in ~11% of human breast cancers. Enriched in HER2+ breast cancers, the 17q23 amplification is significantly correlated with poor clinical outcomes. In addition to the previously identified oncogene WIP1, we uncover an oncogenic microRNA gene, MIR21, in a majority of the WIP1-containing 17q23 amplicons. The 17q23 amplification results in aberrant expression of WIP1 and miR-21, which not only promotes breast tumorigenesis, but also leads to resistance to anti-HER2 therapies. Inhibiting WIP1 and miR-21 selectively inhibits the proliferation, survival and tumorigenic potential of the HER2+ breast cancer cells harboring 17q23 amplification. To overcome the resistance of trastuzumab-based therapies in vivo, we develop pH-sensitive nanoparticles for specific co-delivery of the WIP1 and miR-21 inhibitors into HER2+ breast tumors, leading to a profound reduction of tumor growth. These results demonstrate the great potential of the combined treatment of WIP1 and miR-21 inhibitors for the trastuzumab-resistant HER2+ breast cancers.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 17/genetics , Drug Resistance, Neoplasm/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DEAD-box RNA Helicases/metabolism , Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Female , Gene Amplification/drug effects , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nanoparticles/chemistry , Protein Phosphatase 2C/genetics , Protein Phosphatase 2C/metabolism , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use
9.
Nat Commun ; 9(1): 4394, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30349055

ABSTRACT

Heterozygous deletion of chromosome 17p (17p) is one of the most frequent genomic events in human cancers. Beyond the tumor suppressor TP53, the POLR2A gene encoding the catalytic subunit of RNA polymerase II (RNAP2) is also included in a ~20-megabase deletion region of 17p in 63% of metastatic castration-resistant prostate cancer (CRPC). Using a focused CRISPR-Cas9 screen, we discovered that heterozygous loss of 17p confers a selective dependence of CRPC cells on the ubiquitin E3 ligase Ring-Box 1 (RBX1). RBX1 activates POLR2A by the K63-linked ubiquitination and thus elevates the RNAP2-mediated mRNA synthesis. Combined inhibition of RNAP2 and RBX1 profoundly suppress the growth of CRPC in a synergistic manner, which potentiates the therapeutic effectivity of the RNAP2 inhibitor, α-amanitin-based antibody drug conjugate (ADC). Given the limited therapeutic options for CRPC, our findings identify RBX1 as a potentially therapeutic target for treating human CRPC harboring heterozygous deletion of 17p.


Subject(s)
Chromosomes, Human, Pair 17/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA Polymerase II/metabolism , Apoptosis/genetics , Apoptosis/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Chromosomes, Human, Pair 17/genetics , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Male , Prostatic Neoplasms, Castration-Resistant/genetics , RNA Polymerase II/genetics , Sequence Deletion/genetics , Sequence Deletion/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Ubiquitin-Protein Ligases
10.
J Clin Invest ; 128(7): 2951-2965, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29649003

ABSTRACT

A synthetic lethality-based strategy has been developed to identify therapeutic targets in cancer harboring tumor-suppressor gene mutations, as exemplified by the effectiveness of poly ADP-ribose polymerase (PARP) inhibitors in BRCA1/2-mutated tumors. However, many synthetic lethal interactors are less reliable due to the fact that such genes usually do not perform fundamental or indispensable functions in the cell. Here, we developed an approach to identifying the "essential lethality" arising from these mutated/deleted essential genes, which are largely tolerated in cancer cells due to genetic redundancy. We uncovered the cohesion subunit SA1 as a putative synthetic-essential target in cancers carrying inactivating mutations of its paralog, SA2. In SA2-deficient Ewing sarcoma and bladder cancer, further depletion of SA1 profoundly and specifically suppressed cancer cell proliferation, survival, and tumorigenic potential. Mechanistically, inhibition of SA1 in the SA2-mutated cells led to premature chromatid separation, dramatic extension of mitotic duration, and consequently, lethal failure of cell division. More importantly, depletion of SA1 rendered those SA2-mutated cells more susceptible to DNA damage, especially double-strand breaks (DSBs), due to reduced functionality of DNA repair. Furthermore, inhibition of SA1 sensitized the SA2-deficient cancer cells to PARP inhibitors in vitro and in vivo, providing a potential therapeutic strategy for patients with SA2-deficient tumors.


Subject(s)
Antigens, Nuclear/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Nuclear Proteins/genetics , Animals , Antigens, Nuclear/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , DNA Breaks, Double-Stranded , Female , Gene Knockdown Techniques , Genes, Essential , Humans , Mice , Mice, Nude , Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Xenograft Model Antitumor Assays , Cohesins
11.
FASEB J ; 32(5): 2422-2437, 2018 05.
Article in English | MEDLINE | ID: mdl-29269400

ABSTRACT

The fourth member of the leucine-rich repeat-containing GPCR family (LGR4, frequently referred to as GPR48) and its cognate ligands, R-spondins (RSPOs) play crucial roles in the development of multiple organs as well as the survival of adult stem cells by activation of canonical Wnt signaling. Wnt/ß-catenin signaling acts to regulate breast cancer; however, the molecular mechanisms determining its spatiotemporal regulation are largely unknown. In this study, we identified LGR4 as a master controller of Wnt/ß-catenin signaling-mediated breast cancer tumorigenesis, metastasis, and cancer stem cell (CSC) maintenance. LGR4 expression in breast tumors correlated with poor prognosis. Either Lgr4 haploinsufficiency or mammary-specific deletion inhibited mouse mammary tumor virus (MMTV)- PyMT- and MMTV- Wnt1-driven mammary tumorigenesis and metastasis. Moreover, LGR4 down-regulation decreased in vitro migration and in vivo xenograft tumor growth and lung metastasis. Furthermore, Lgr4 deletion in MMTV- Wnt1 tumor cells or knockdown in human breast cancer cells decreased the number of functional CSCs by ∼90%. Canonical Wnt signaling was impaired in LGR4-deficient breast cancer cells, and LGR4 knockdown resulted in increased E-cadherin and decreased expression of N-cadherin and snail transcription factor -2 ( SNAI2) (also called SLUG), implicating LGR4 in regulation of epithelial-mesenchymal transition. Our findings support a crucial role of the Wnt signaling component LGR4 in breast cancer initiation, metastasis, and breast CSCs.-Yue, Z., Yuan, Z., Zeng, L., Wang, Y., Lai, L., Li, J., Sun, P., Xue, X., Qi, J., Yang, Z., Zheng, Y., Fang, Y., Li, D., Siwko, S., Li, Y., Luo, J., Liu, M. LGR4 modulates breast cancer initiation, metastasis, and cancer stem cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Wnt Signaling Pathway , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Heterografts , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/genetics
12.
Int Rev Cell Mol Biol ; 333: 1-49, 2017.
Article in English | MEDLINE | ID: mdl-28729023

ABSTRACT

In the classical model of tumorigenesis, cancer develops via slowly accumulating mutations that facilitate uncontrolled cell growth and allow cells to escape apoptosis. Oncogenes and tumor suppressor genes regulate the key signaling pathways involved in tumorigenesis and cancer progression. Moreover, studies indicate that MicroRNAs (MiRNAs) are also key parts of these processes. MiRNAs are short, noncoding RNAs that regulate the expression of target genes at the posttranscriptional level. By regulating the expression of numerous cytokines, MiRNAs play crucial roles in cell growth, apoptosis, and stemness maintenance. Abundant evidence demonstrates that MiRNAs can function as both oncogenes and tumor suppressors in accommodating the proliferation and invasion of cancer cells in solid tumors. Targeting these MiRNAs may significantly alter oncogenic signaling pathways and slow or halt progression of many different types of tumors. A better understanding of the functions of MiRNAs in cancer will enable the development of new treatment strategies for chemoresistant malignancies. This review discusses recent findings about the connections between MiRNAs and carcinogenesis and provides insight into the role of MiRNAs in generating chemoresistance.


Subject(s)
Carcinogenesis/metabolism , DNA Damage , Drug Resistance, Neoplasm/genetics , MicroRNAs/metabolism , Animals , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Humans , MicroRNAs/genetics , Signal Transduction
13.
Nat Commun ; 7: 10384, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26785948

ABSTRACT

Bcl-xL suppresses mitochondria-mediated apoptosis and is frequently overexpressed in cancer to promote cancer cell survival. Bcl-xL also promotes metastasis. However, it is unclear whether this metastatic function is dependent on its anti-apoptotic activity in the mitochondria. Here we demonstrate that Bcl-xL promotes metastasis independent of its anti-apoptotic activity. We show that apoptosis-defective Bcl-xL mutants and an engineered Bcl-xL targeted to the nucleus promote epithelial-mesenchymal transition, migration, invasion and stemness in pancreatic neuroendocrine tumour (panNET) and breast cancer cell lines. However, Bcl-xL proteins targeted to the mitochondria or outside of the nucleus do not have these functions. We confirm our findings in spontaneous and xenograft mouse models. Furthermore, Bcl-xL exerts metastatic function through epigenetic modification of the TGFß promoter to increase TGFß signalling. Consistent with these findings, we detect nuclear Bcl-xL in human metastatic panNETs. Taken together, the metastatic function of Bcl-xL is independent of its anti-apoptotic activity and its residence in the mitochondria.


Subject(s)
Apoptosis/physiology , Cell Movement/physiology , bcl-X Protein/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Cell Survival/physiology , Chromatin Immunoprecipitation , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Immunohistochemistry , Immunoprecipitation , In Vitro Techniques , Mice , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , bcl-X Protein/genetics
14.
Oncotarget ; 6(9): 6749-61, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25730907

ABSTRACT

The epidermal growth factor receptor (EGFR) is a therapeutic target (oncotarget) in NSCLC. Using in vitro EGFR kinase activity system, we identified a novel small molecule, WB-308, as an inhibitor of EGFR. WB-308 decreased NSCLC cell proliferation and colony formation, by causing G2/M arrest and apoptosis. Furthermore, WB-308 inhibited the engraft tumor growths in two animal models in vivo (lung orthotopic transplantation model and patient-derived engraft mouse model). WB-308 impaired the phosphorylation of EGFR, AKT, and ERK1/2 protein. WB-308 was less cytotoxic than Gefitinib. Our study suggests that WB-308 is a novel EGFR-TKI and may be considered to substitute for Gefitinib in clinical therapy for NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carbolines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Carbolines/chemistry , Carbolines/toxicity , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gefitinib , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy , Mutation , Phosphorylation , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
J Med Chem ; 57(3): 600-12, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24417479

ABSTRACT

The transforming growth factor beta (TGFß) signaling cascade is considered as one of the pivotal oncogenic pathways in most advanced cancers. Inhibition of the TGFß signaling pathway by specific antagonists, neutralizing antibodies, or small molecules is considered as an effective strategy for the treatment of tumor growth and metastasis. Here we demonstrated the identification of a series of tetrahydro-ß-carboline derivatives from virtual screening which potentially inhibit the TGFß signaling pathway. Optimization of the initial hit compound 2-benzoyl-1,3,4,9-tetrahydro-ß-carboline (8a) through substitution at different positions to define the structure-activity relationship resulted in the discovery of potent inhibitors of the TGFß signaling pathway. Among them, compound 8d, one of the tested compounds, not only showed potent inhibition of lung cancer cell proliferation and migration in vitro but also strongly suppressed growth of lung cancer and breast cancer in vivo.


Subject(s)
Antineoplastic Agents/chemical synthesis , Carbolines/chemical synthesis , Neoplasm Metastasis/prevention & control , Transforming Growth Factor beta/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carbolines/chemistry , Carbolines/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Neoplasm Transplantation , Signal Transduction/drug effects , Structure-Activity Relationship
16.
J Natl Cancer Inst ; 105(1): 47-58, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23178439

ABSTRACT

BACKGROUND: Transforming growth factor beta (TGFß), which is implicated in metastasis to various organs in breast cancer, is a potential target for new antitumor metastasis drugs. METHODS: To identify specific inhibitors of TGFß receptor 1 (TGFßR1) in breast cancer metastasis, a virtual library of more than 400000 different compounds was screened by molecular docking modeling and confirmed with Smad-binding element luciferase and TGFßR1 kinase assays. Affymetrix GeneChip expression analysis of mRNA levels and real-time polymerase chain reaction were performed to determine expression changes of TGFß-mediated, metastasis-associated genes in breast cancer cells after treatment with the small-molecule inhibitor YR-290. YR-290 was also examined for its effects on breast cancer migration, invasion, and metastasis using transwell and epithelial-to-mesenchymal transition (EMT) assays in vitro and three different mouse (BALB/c and NU/NU nude) models (n = 10 per group). Kaplan-Meier analyses were used to assess survival. All statistical tests were two-sided. RESULTS: YR-290 interacted with the kinase domain of TGFßR1, abrogated kinase activity (half maximal inhibitory concentration = 137nM, 95% confidence interval = 126.4 to 147.6nM) and inhibited the TGFß-mediated downstream signaling pathway and metastasis-associated genes in breast cancer cells. YR-290 inhibited TGFß-modulated breast cancer cell migration and invasion. In tumor metastasis mouse models, YR-290 almost completely blocked cancer metastasis. Numbers of lung tumor nodules of mice treated with 1mg/kg and 5mg/kg YR-290 were reduced by 74.93% (95% confidence interval = 61.45% to 88.41%) and 94.93% (95% confidence interval = 82.13% to 100%), respectively, compared with control mice. Treatment with YR-290 also statistically significantly prolonged the survival of tumor-bearing mice. CONCLUSIONS: YR-290 is a novel inhibitor of tumor metastasis that works by blocking TGFß signaling pathways.


Subject(s)
Adenocarcinoma/prevention & control , Adenocarcinoma/secondary , Antineoplastic Agents/pharmacology , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Carbolines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Carbolines/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
PLoS One ; 6(11): e28318, 2011.
Article in English | MEDLINE | ID: mdl-22140575

ABSTRACT

In their expression process, different genes can generate diverse functional products, including various protein-coding or noncoding RNAs. Here, we investigated the protein-coding capacities and the expression levels of their isoforms for human known genes, the conservation and disease association of long noncoding RNAs (ncRNAs) with two transcriptome sequencing datasets from human brain tissues and 10 mixed cell lines. Comparative analysis revealed that about two-thirds of the genes expressed between brain and cell lines are the same, but less than one-third of their isoforms are identical. Besides those genes specially expressed in brain and cell lines, about 66% of genes expressed in common encoded different isoforms. Moreover, most genes dominantly expressed one isoform and some genes only generated protein-coding (or noncoding) RNAs in one sample but not in another. We found 282 human genes could encode both protein-coding and noncoding RNAs through alternative splicing in the two samples. We also identified more than 1,000 long ncRNAs, and most of those long ncRNAs contain conserved elements across either 46 vertebrates or 33 placental mammals or 10 primates. Further analysis showed that some long ncRNAs differentially expressed in human breast cancer or lung cancer, several of those differentially expressed long ncRNAs were validated by RT-PCR. In addition, those validated differentially expressed long ncRNAs were found significantly correlated with certain breast cancer or lung cancer related genes, indicating the important biological relevance between long ncRNAs and human cancers. Our findings reveal that the differences of gene expression profile between samples mainly result from the expressed gene isoforms, and highlight the importance of studying genes at the isoform level for completely illustrating the intricate transcriptome.


Subject(s)
Brain/metabolism , Open Reading Frames/genetics , RNA, Untranslated/genetics , Sequence Analysis, RNA/methods , Adult , Base Sequence , Cell Line , Conserved Sequence , Disease/genetics , Exons/genetics , Female , Gene Expression Regulation , Humans , Male , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
18.
J Pharmacol Exp Ther ; 339(1): 238-47, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21765040

ABSTRACT

Compared with traditional cytotoxic cancer therapy, therapy-induced cancer cell senescence attracts much interest because it is similarly effective, has fewer side effects, and is more efficiently cleared by immune cells. In this study, we demonstrate that unlike caffeic acid phenethyl ester, caffeic acid 3,4-dihydroxy-phenethyl ester (CADPE), which is isolated from the medicinal plants Sarcandra glabra and Teucrium pilosum, inhibits human cancer cell growth and colony formation by inducing cancer cell senescence, not apoptosis. CADPE induces cell senescence and morphology changes by increasing cellular size and cytoplasmic granularity, enhancing senescence-associated ß-galactosidase activity and differentiated embryo-chondrocyte expressed gene 1 expression, and blocking cell-cycle arrest in the G(1) phase. To help understand the underlying mechanisms, we show that CADPE significantly suppressed the expression of Twist1 and led to the up-regulation of rat sarcoma, p53, p21(WAF1/CIP1), and p16(INK4a) proteins in a dose-dependent manner, resulting in the hypophosphorylation of retinoblastoma protein. Furthermore, overexpression of Twist1 prevented CADPE-induced cell senescence in tumor cells. Therefore, our studies provide evidence for a novel role of CADPE in cancer cell senescence by targeting the Twist1-dependent senescence signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Caffeic Acids/pharmacology , Cellular Senescence/drug effects , Twist-Related Protein 1/antagonists & inhibitors , Annexin A5 , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Dose-Response Relationship, Drug , G1 Phase/drug effects , Genes, ras , Humans , Phosphorylation , Retinoblastoma Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Stem Cell Assay , Twist-Related Protein 1/genetics , Twist-Related Protein 1/physiology , Up-Regulation , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...