1
|
Xu Z, Chu M. Advances in Immunosuppressive Agents Based on Signal Pathway. Front Pharmacol 2022; 13:917162. [PMID: 35694243 PMCID: PMC9178660 DOI: 10.3389/fphar.2022.917162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
Immune abnormality involves in various diseases, such as infection, allergic diseases, autoimmune diseases, as well as transplantation. Several signal pathways have been demonstrated to play a central role in the immune response, including JAK/STAT, NF-κB, PI3K/AKT-mTOR, MAPK, and Keap1/Nrf2/ARE pathway, in which multiple targets have been used to develop immunosuppressive agents. In recent years, varieties of immunosuppressive agents have been approved for clinical use, such as the JAK inhibitor tofacitinib and the mTOR inhibitor everolimus, which have shown good therapeutic effects. Additionally, many immunosuppressive agents are still in clinical trials or preclinical studies. In this review, we classified the immunosuppressive agents according to the immunopharmacological mechanisms, and summarized the phase of immunosuppressive agents.
Collapse
Affiliation(s)
- Zhiqing Xu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Pharmacology, Jilin University, Changchun, China
| | - Ming Chu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
2
|
Liu F, Gao Y, Xu B, Xiong S, Yi S, Sun J, Chen Z, Liu X, Li Y, Lin Y, Wen Y, Qin Y, Yang S, Li H, Tejasvi T, Tsoi L, Tu P, Ren X, Wang Y. PEG10 amplification at 7q21.3 potentiates large-cell transformation in cutaneous T-cell lymphoma. Blood 2022; 139:554-571. [PMID: 34582557 PMCID: PMC8893588 DOI: 10.1182/blood.2021012091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/07/2021] [Indexed: 01/29/2023] Open
Abstract
Mycosis fungoides (MF), the most common form of cutaneous T-cell lymphoma, undergo large-cell transformation (LCT) in the late stage, manifesting aggressive behavior, resistance to treatments, and poor prognosis, but the mechanisms involved remain unclear. To identify the molecular driver of LCT, we collected tumor samples from 133 MF patients and performed whole-transcriptome sequencing on 49 advanced-stage MF patients, followed by integrated copy number inference and genomic hybridization. Tumors with LCT showed unique transcriptional programs and enriched expressions of genes at chr7q. Paternally expressed gene 10 (PEG10), an imprinted gene at 7q21.3, was ectopically expressed in malignant T cells from LCT, driven by 7q21.3 amplification. Mechanistically, aberrant PEG10 expression increased cell size, promoted cell proliferation, and conferred treatment resistance by a PEG10/KLF2/NF-κB axis in in vitro and in vivo models. Pharmacologically targeting PEG10 reversed the phenotypes of proliferation and treatment resistance in LCT. Our findings reveal new molecular mechanisms underlying LCT and suggest that PEG10 inhibition may serve as a promising therapeutic approach in late-stage aggressive T-cell lymphoma.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/genetics
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- DNA-Binding Proteins/genetics
- Female
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Genomic Imprinting
- Humans
- Lymphoma, T-Cell, Cutaneous/genetics
- Lymphoma, T-Cell, Cutaneous/pathology
- Mice, Inbred NOD
- Mice, SCID
- Mycosis Fungoides/genetics
- Mycosis Fungoides/pathology
- RNA-Binding Proteins/genetics
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Mice
Collapse
Affiliation(s)
- Fengjie Liu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yumei Gao
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Bufang Xu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shan Xiong
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shengguo Yi
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Jingru Sun
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Zhuojing Chen
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xiangjun Liu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yingyi Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yuchieh Lin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yujie Wen
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Yao Qin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Shuxia Yang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Hang Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Trilokraj Tejasvi
- Department of Dermatology, University of Michigan, Ann Arbor, MI; and
| | - Lam Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI; and
| | - Ping Tu
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100034, China
| | - Yang Wang
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing 100034, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| |
Collapse
|
3
|
Scotto L, Kinahan C, Douglass E, Deng C, Safari M, Casadei B, Marchi E, Lue JK, Montanari F, Falchi L, Qiao C, Renu N, Bates SE, Califano A, O'Connor OA. Targeting the T-Cell Lymphoma Epigenome Induces Cell Death, Cancer Testes Antigens, Immune-Modulatory Signaling Pathways. Mol Cancer Ther 2021; 20:1422-1430. [PMID: 34108263 DOI: 10.1158/1535-7163.mct-20-0377] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/13/2020] [Accepted: 06/07/2021] [Indexed: 11/16/2022]
Abstract
The peripheral T-cell lymphomas (PTCL) could be considered the prototypical epigenetic disease. As a disease, they are uniquely sensitive to histone deacetylase (HDAC) and DNA methyltransferase (DNMT) inhibitors, both alone and in combination, are characterized by a host of mutations in epigenetic genes, and can develop spontaneously in genetically engineered murine models predicated on established recurring mutations in (RHOAG17V) and TET2, an epigenetic gene governing DNA methylation. Given the clinical benefit of HDAC inhibitors (HDACi) and hypomethlyation agents alone and in combination in PTCL, we sought to explore a mechanistic basis for these agents in PTCL. Herein, we reveal profound class synergy between HDAC and DNMT inhibitors in PTCL, and that the combination induces degrees of gene expression that are substantially different and more extensive than that observed for the single agents. A prominent signature of the combination relates to the transcriptional induction of cancer testis antigens and genes involved in the immune response. Interestingly, TBX21 and STAT4, master regulators of TH1 differentiation, were among the genes upregulated by the combination, suggesting the induction of a TH1-like phenotype. Moreover, suppression of genes involved in cholesterol metabolism and the matrisome were also identified. We believe that these data provide a strong rationale for clinical studies, and future combinations leveraging an immunoepigenetic platform.
Collapse
Affiliation(s)
- Luigi Scotto
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Cristina Kinahan
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Eugene Douglass
- Department of Systems Biology, Columbia University, New York, New York
| | - Changchun Deng
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Maryam Safari
- Division of Hematology and Oncology, Columbia University, Medical Center, New York, New York
| | - Beatrice Casadei
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Enrica Marchi
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Jennifer K Lue
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Francesca Montanari
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Lorenzo Falchi
- Center for Lymphoid Malignancies, Columbia University, Medical Center, New York, New York.,Division of Experimental Therapeutics, Columbia University, Medical Center, New York, New York
| | - Changhong Qiao
- Department of Medicine, Biomarkers Core Laboratory, Columbia University, Medical Center, New York, New York
| | - Nandakumar Renu
- Department of Medicine, Biomarkers Core Laboratory, Columbia University, Medical Center, New York, New York
| | - Susan E Bates
- Division of Hematology and Oncology, Columbia University, Medical Center, New York, New York
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York.,Department of Biomedical Informatics, Columbia University, New York, New York.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York.,Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York.,J.P. Sulzberger Columbia Genome Center, New York, New York
| | - Owen A O'Connor
- Department of Medicine, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
4
|
Kalac M, Mangone M, Rinderspacher A, Deng SX, Scotto L, Markson M, Bansal M, Califano A, Landry DW, O'Connor OA. N-quinoline-benzenesulfonamide derivatives exert potent anti-lymphoma effect by targeting NF-κB. iScience 2020; 23:101884. [PMID: 33354662 PMCID: PMC7744703 DOI: 10.1016/j.isci.2020.101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/28/2020] [Accepted: 11/25/2020] [Indexed: 11/18/2022] Open
Abstract
We previously identified the N-quinoline-benzenesulfonamide (NQBS) scaffold as a potent inhibitor of nuclear factor-κB (NF-κB) translocation. Now, we report the structure-activity relationship of compounds with the NQBS scaffold in models of diffuse large B-cell lymphoma (DLBCL). We identified CU-O42, CU-O47, and CU-O75 as NQBS analogs with the most potent cytotoxic activity in DLBCL lines. Their anti-lymphoma effect was mediated by NF-κB sequestration to the cytoplasm of DLBCL cells. Internal Coordinates Mechanics analysis suggested direct binding between CU-O75 and IκBα/p50/p65 which leads to the stabilization of the NF-κB trimer. A whole cellular thermal shift assay confirmed direct binding of the NQBS to IκBα, an inhibitory component of the IκBα/p50/p65 trimer. Lymphoma cell line sequencing revealed CU-O75 induced downregulation of NF-κB-dependent genes and DeMAND analysis identified IκBα as one of the top protein targets for CU-O75. CU-O42 was potent in inhibiting tumor growth in two mouse models of aggressive lymphomas.
Collapse
Affiliation(s)
- Matko Kalac
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Center for Lymphoid Malignancies, Columbia University Irving Medical Center, New York, NY, USA
- Department of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Michael Mangone
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Center for Lymphoid Malignancies, Columbia University Irving Medical Center, New York, NY, USA
| | - Alison Rinderspacher
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Nephrology, Columbia University Irving Medical Center, New York, NY, USA
| | - Shi-Xian Deng
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Nephrology, Columbia University Irving Medical Center, New York, NY, USA
| | - Luigi Scotto
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Center for Lymphoid Malignancies, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Markson
- Department of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Mukesh Bansal
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Psychogenics Inc., Paramus, NJ, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrea Califano
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- J.P. Sulzberger Columbia Genome Center, New York, NY, USA
| | - Donald W. Landry
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Nephrology, Columbia University Irving Medical Center, New York, NY, USA
| | - Owen A. O'Connor
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Center for Lymphoid Malignancies, Columbia University Irving Medical Center, New York, NY, USA
- Division of Hematology and Oncology, University of Virginia Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
5
|
Single Crystal X-Ray Structure for the Disordered Two Independent Molecules of Novel Isoflavone: Synthesis, Hirshfeld Surface Analysis, Inhibition and Docking Studies on IKKβ of 3-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxy-4H-chromen-4-one. CRYSTALS 2020. [DOI: 10.3390/cryst10100911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The structure of the isoflavone compound, 3-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxy-4H-chromen-4-one (5), was elucidated by 2D-NMR spectra, mass spectrum and single crystal X-ray crystallography. Compound 5, C19H16O6, was crystallized in the monoclinic space group P21/c with the cell parameters; a = 12.0654(5) Å, b =11.0666(5) Å, c = 23.9550(11) Å, β = 101.3757(16)°, V = 3135.7(2) Å3, and Z = 8. The asymmetric unit of compound 5 consists of two independent molecules 5I and 5II. Both molecules exhibit the disorder of each methylene group present in their 1,4-dioxane rings with relative occupancies of 0.599(10) (5I) and 0.812(9) (5II) for the major component A, and 0.401(10) (5I) and 0.188(9) (5II) for the minor component B, respectively. Each independent molecule revealed remarkable discrepancies in bond lengths, bond angles and dihedral angles in the disordered regions of 1,4-dioxane rings. The common feature of the molecules 5I and 5II are a chromone ring and a benzodioxin ring, which are more tilted towards each other in 5I than in 5II. An additional difference between the molecules is seen in the relative disposition of two methoxy substituents. In the crystal, the molecule 5II forms inversion dimers which are linked into chains along an a-axis direction by intermolecular C–H⋯O interactions. Additional C–H⋯O hydrogen bonds connected the molecules 5I and 5II each other to form a three-dimensional network. Hirshfeld surface analysis evaluated the relative intermolecular interactions which contribute to each crystal structure 5I and 5II. Western blot analysis demonstrated that compound 5 inhibited the TNFα-induced phosphorylation of IKKα/β, resulting in attenuating further downstream NF-κB signaling. A molecular docking study predicted the possible binding of compound 5 to the active site of IKKβ. Compound 5 showed an inhibitory effect on the clonogenicity of HCT116 human colon cancer cells. These results suggest that compound 5 can be used as a platform for the development of an anti-cancer agent targeting IKKα/β.
Collapse
|
6
|
Anti-Metastasis Fascin Inhibitors Decrease the Growth of Specific Subtypes of Cancers. Cancers (Basel) 2020; 12:cancers12082287. [PMID: 32824026 PMCID: PMC7466159 DOI: 10.3390/cancers12082287] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/04/2020] [Accepted: 08/12/2020] [Indexed: 01/15/2023] Open
Abstract
Fascin is an actin-bundling protein that is critical for filopodial formation and other cellular cytoskeletal structures. An elevated expression of fascin has been observed in tumor cells and is correlated with a shorter survival of cancer patients. Given its roles in tumor cell migration and invasion, we have developed small-molecule fascin inhibitors to prevent and delay tumor metastasis. Here we report the characterization of a new fascin inhibitor in mice. In addition to its inhibitory effects on tumor metastasis, we also report that fascin inhibitors can decrease the growth of specific subtypes of cancers, including epidermal growth factor receptor (EGFR)-high triple-negative breast cancer, and activated B-cell subtypes of diffuse large B-cell lymphoma. Hence, fascin inhibitors can be used to not only inhibit tumor metastasis, but also decrease the tumor growth of specific cancer types.
Collapse
|
7
|
Hypoxic colorectal cancer cells promote metastasis of normoxic cancer cells depending on IL-8/p65 signaling pathway. Cell Death Dis 2020; 11:610. [PMID: 32737283 PMCID: PMC7395770 DOI: 10.1038/s41419-020-02797-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/14/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
Tumor heterogeneity is an important feature of malignant tumors, and cell subpopulations may positively interact to facilitate tumor progression. Studies have shown that hypoxic cancer cells possess enhanced metastatic capacity. However, it is still unclear whether hypoxic cancer cells may promote the metastasis of normoxic cells, which have greater access to the blood circulation. When cocultured with hypoxic CRC cells or treated with hypoxic CRC cell-derived CM, normoxic CRC cells possessed increased metastatic capacity. Furthermore, hypoxic CRC cell-derived CM was enriched in interleukin 8. Hypoxic CRC cell-derived CM and recombinant human IL-8 both enhanced the metastatic capacity of normoxic cells by increasing the phosphorylation of p65 and then by inducing epithelial-mesenchymal transition. Knockdown of IL-8 in hypoxic CRC cells or the use of an anti-IL-8 antibody attenuated the CM- or rhIL-8-induced prometastatic capacity of normoxic CRC cells. Inhibition or knockdown of p65 abrogated IL-8-induced prometastatic effects. Most importantly, hypoxia-treated xenograft tumors enhanced the metastasis of normoxic CRC cells. Hypoxic CRC cell-derived IL-8 promotes the metastatic capacity of normoxic cells, and novel therapies targeting the positive interactions between hypoxic and normoxic cells should be developed.
Collapse
|
8
|
Chen J, Liu A, Lin Z, Wang B, Chai X, Chen S, Lu W, Zheng M, Cao T, Zhong M, Li R, Wu M, Lu Z, Pang W, Huang W, Xiao L, Lin D, Wang Z, Lei F, Chen X, Long W, Zheng Y, Chen Q, Zeng J, Ren D, Li J, Zhang X, Huang Y. Downregulation of the circadian rhythm regulator HLF promotes multiple-organ distant metastases in non-small cell lung cancer through PPAR/NF-κb signaling. Cancer Lett 2020; 482:56-71. [PMID: 32289442 DOI: 10.1016/j.canlet.2020.04.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death due to its early recurrence and widespread metastatic potential. Accumulating studies have reported that dysregulation of circadian rhythms-associated regulators is implicated in the recurrence and metastasis of NSCLC. Therefore, identification of metastasis-associated circadian rhythm genes is clinically necessary. Here we report that the circadian gene hepatic leukemia factor (HLF), which was dramatically reduced in early-relapsed NSCLC tissues, was significantly correlated with early progression and distant metastasis in NSCLC patients. Upregulating HLF inhibited, while silencing HLF promoted lung colonization, as well as metastasis of NSCLC cells to bone, liver and brain in vivo. Importantly, downexpression of HLF promoted anaerobic metabolism to support anchorage-independent growth of NSCLC cells under low nutritional condition by activating NF-κB/p65 signaling through disrupting translocation of PPARα and PPARγ. Further investigations revealed that both genetic deletion and methylation contribute to downexpression of HLF in NSCLC tissues. In conclusion, our results shed light on a plausible mechanism by which HLF inhibits distant metastasis in NSCLC, suggesting that HLF may serve as a novel target for clinical intervention in NSCLC.
Collapse
Affiliation(s)
- Jiarong Chen
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China; Department of Oncology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Aibin Liu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhichao Lin
- Department of Thoracic Surgery, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Bin Wang
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China; Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China; Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, 524023, China
| | - Xingxing Chai
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China; Laboratory Animal Center, Guangdong Medical University, Zhanjiang, 524023, China
| | - Shasha Chen
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China; Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China; Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, 524023, China
| | - Wenjie Lu
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Mingzhu Zheng
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Ting Cao
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Meigong Zhong
- Department of Pharmacy, Jiangmen Maternity and Child Health Care Hospital, Jiangmen, 529030, China
| | - Ronggang Li
- Department of Pathology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Minyan Wu
- Department of Basic Medicine, Guangdong Jiangmen Chinese Medical College, Jiangmen, 529030, China
| | - Zhuming Lu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Wenguang Pang
- Department of Thoracic Surgery, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Wenhai Huang
- Department of Thoracic Surgery, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Lin Xiao
- Department of Radiotherapy Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Daren Lin
- Department of Oncology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Zhihui Wang
- Department of Oncology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Fangyong Lei
- Department of Oncology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Xiangmeng Chen
- Department of Radiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Wansheng Long
- Department of Radiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Yan Zheng
- Department of Research and Development, Research and Development Center for Molecular Diagnosis Engineering Technology of Human Papillomavirus (HPV) Related Diseases of Guangdong Province, Hybribio Limited, Changzhou, 521021, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China; Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, 524023, China
| | - Dong Ren
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China; Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China
| | - Jun Li
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China; Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China; Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Yanming Huang
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, 529030, China.
| |
Collapse
|
9
|
Wang M, Fang X, Wang X. Emerging role of histone deacetylase inhibitors in the treatment of diffuse large B-cell lymphoma. Leuk Lymphoma 2019; 61:763-775. [PMID: 31766900 DOI: 10.1080/10428194.2019.1691194] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Although current immunochemotherapy has increased the therapeutic efficacy in diffuse large B-cell lymphoma (DLBCL), there are still some patients who present unfavorable outcomes. Novel effective treatment strategies are needed to improve the prognosis of DLBCL. In this review, we discussed the functional mechanisms and therapeutic applications of histone deacetylases inhibitors (HDIs) in DLBCL from preclinical and clinical studies. The mechanistic rationale of HDIs involved a wide range of effects including the regulation of transcription factors, tumor suppressors, and cell surface molecules. Histone deacetylases inhibitors as monotherapy performed limited activity in the treatment of DLBCL in present clinical trials, but its combination with other regimens has emerged as potential treatment candidates with generally acceptable and manageable adverse effects. Further investigation on the anti-tumor mechanisms of HDIs and ongoing clinical trials will hopefully facilitate the application of HDIs in patients with DLBCL.
Collapse
Affiliation(s)
- Mingyang Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, People's Republic of China
| |
Collapse
|
10
|
Shen Y, Song Z, Lu X, Ma Z, Lu C, Zhang B, Chen Y, Duan M, Apetoh L, Li X, Guo J, Miao Y, Zhang G, Yang D, Cai Z, Wang J. Fas signaling-mediated T H9 cell differentiation favors bowel inflammation and antitumor functions. Nat Commun 2019; 10:2924. [PMID: 31266950 PMCID: PMC6606754 DOI: 10.1038/s41467-019-10889-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 06/07/2019] [Indexed: 11/17/2022] Open
Abstract
Fas induces apoptosis in activated T cell to maintain immune homeostasis, but the effects of non-apoptotic Fas signaling on T cells remain unclear. Here we show that Fas promotes TH9 cell differentiation by activating NF-κB via Ca2+-dependent PKC-β activation. In addition, PKC-β also phosphorylates p38 to inactivate NFAT1 and reduce NFAT1-NF-κB synergy to promote the Fas-induced TH9 transcription program. Fas ligation exacerbates inflammatory bowel disease by increasing TH9 cell differentiation, and promotes antitumor activity in p38 inhibitor-treated TH9 cells. Furthermore, low-dose p38 inhibitor suppresses tumor growth without inducing systemic adverse effects. In patients with tumor, relatively high TH9 cell numbers are associated with good prognosis. Our study thus implicates Fas in CD4+ T cells as a target for inflammatory bowel disease therapy. Furthermore, simultaneous Fas ligation and low-dose p38 inhibition may be an effective approach for TH9 cell induction and cancer therapy. Fas signalling induces apoptosis of activated T cells to maintain immune homeostasis. Here the authors show that Fas also induces PKC-β activation to promote NF-κB-mediated TH9 cell differentiation, while p38 activation by PKC-β antagonizes this effect, thereby supporting a synergy between p38 inhibitor and Fas for TH9 differentiation.
Collapse
Affiliation(s)
- Yingying Shen
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.,Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, 310003, Hangzhou, China.,Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zhengbo Song
- Department of Medical Oncology, Zhejiang Cancer Hospital, 310022, Hangzhou, China
| | - Xinliang Lu
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zeyu Ma
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Chaojie Lu
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Bei Zhang
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Yinghu Chen
- Division of Infection Disease, Zhejiang Key Laboratory for Neonatal Diseases, Children's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China
| | - Meng Duan
- Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Lionel Apetoh
- INSERM, U866, Dijon, France.,Faculté de Médecine, Université de Bourgogne, Dijon, 21000, France
| | - Xu Li
- School of Life Science, Westlake University, 310024, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China
| | - Ying Miao
- Clinical Trial Center, Qingdao Municipal Hospital, 266011, Qingdao, China
| | - Gensheng Zhang
- Department of Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
| | - Diya Yang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Jianli Wang
- Institute of Immunology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China. .,Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, 310003, Hangzhou, China.
| |
Collapse
|
11
|
Li W, Li S, Yang J, Cui C, Yu M, Zhang Y. ITGBL1 promotes EMT, invasion and migration by activating NF-κB signaling pathway in prostate cancer. Onco Targets Ther 2019; 12:3753-3763. [PMID: 31190876 PMCID: PMC6529605 DOI: 10.2147/ott.s200082] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Integrin beta-like 1 (ITGBL1) was extensively demonstrated to contribute the metastasis and progression in a variety of cancers. However, its role of ITGBL1 in prostate cancer (PCa) is still not reported. Methods: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot were performed to detect ITGBL1 expression in PCa tissues and cell lines. Immunohistochemical (IHC) staining of ITGBL1 in 174 PCa tissues was performed. The influence of ITGL1 expression in PCa cells epithelial-mesenchymal transition (EMT), migration and invasion was investigated. Notably, the possible mechanisms underlying the action of ITGBL1 in vivo and vitro assays were explored. Results: We analyzed PCa dataset from The Cancer Genome Atlas (TCGA) and found that ITGBL1 was upregulated in PCa tissues. Overexpression of ITGBL1 is positively associated with the progression and lymph node metastasis in PCa patients. Furthermore, upregulating ITGBL1 enhanced the invasion, migration abilities and EMT in PCa cells. Conversely, downregulating ITGBL1 exhibited an opposite effect. Our findings further demonstrated that ITGBL1 promoted invasion and migration via activating NF-κB signaling in PCa cells. Conclusion: Therefore, our results identify a novel metastasis-related gene in PCa, which will help to develop a novel therapeutic strategy in metastatic PCa.
Collapse
Affiliation(s)
- Wenze Li
- Department of Urinary Surgery, The First hospital of Xiangtan city, Xiangtan 411101, People's Republic of China
| | - Shuren Li
- Department of Urinary Surgery, The First hospital of Xiangtan city, Xiangtan 411101, People's Republic of China
| | - Jie Yang
- Department of Urinary Surgery, The First hospital of Xiangtan city, Xiangtan 411101, People's Republic of China
| | - Chunyan Cui
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, People's Republic of China
| | - Miao Yu
- Center for Private Medical Service and Healthcare, The First Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Yadong Zhang
- Department of Andrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| |
Collapse
|
12
|
Zhang Q, Wang S, Chen J, Yu Z. Histone Deacetylases (HDACs) Guided Novel Therapies for T-cell lymphomas. Int J Med Sci 2019; 16:424-442. [PMID: 30911277 PMCID: PMC6428980 DOI: 10.7150/ijms.30154] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022] Open
Abstract
T-cell lymphomas are a heterogeneous group of cancers with different pathogenesis and poor prognosis. Histone deacetylases (HDACs) are epigenetic modifiers that modulate many key biological processes. In recent years, HDACs have been fully investigated for their roles and potential as drug targets in T-cell lymphomas. In this review, we have deciphered the modes of action of HDACs, HDAC inhibitors as single agents, and HDACs guided combination therapies in T-cell lymphomas. The overview of HDACs on the stage of T-cell lymphomas, and HDACs guided therapies both as single agents and combination regimens endow great opportunities for the cure of T-cell lymphomas.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Shaobin Wang
- Health Management Center of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Junhui Chen
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Zhendong Yu
- China Central Laboratory of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| |
Collapse
|
13
|
Prescott JA, Cook SJ. Targeting IKKβ in Cancer: Challenges and Opportunities for the Therapeutic Utilisation of IKKβ Inhibitors. Cells 2018; 7:cells7090115. [PMID: 30142927 PMCID: PMC6162708 DOI: 10.3390/cells7090115] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/15/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023] Open
Abstract
Deregulated NF-κB signalling is implicated in the pathogenesis of numerous human inflammatory disorders and malignancies. Consequently, the NF-κB pathway has attracted attention as an attractive therapeutic target for drug discovery. As the primary, druggable mediator of canonical NF-κB signalling the IKKβ protein kinase has been the historical focus of drug development pipelines. Thousands of compounds with activity against IKKβ have been characterised, with many demonstrating promising efficacy in pre-clinical models of cancer and inflammatory disease. However, severe on-target toxicities and other safety concerns associated with systemic IKKβ inhibition have thus far prevented the clinical approval of any IKKβ inhibitors. This review will discuss the potential reasons for the lack of clinical success of IKKβ inhibitors to date, the challenges associated with their therapeutic use, realistic opportunities for their future utilisation, and the alternative strategies to inhibit NF-κB signalling that may overcome some of the limitations associated with IKKβ inhibition.
Collapse
Affiliation(s)
- Jack A Prescott
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
14
|
Zhang X, Liu C, Hu F, Zhang Y, Wang J, Gao Y, Jiang Y, Zhang Y, Lan X. PET Imaging of VCAM-1 Expression and Monitoring Therapy Response in Tumor with a 68Ga-Labeled Single Chain Variable Fragment. Mol Pharm 2018; 15:609-618. [PMID: 29308904 DOI: 10.1021/acs.molpharmaceut.7b00961] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) is a transmembrane glycoprotein closely related to tumorigenicity as well as tumor metastasis. It is also a well-known candidate for detecting tumors. LY2409881, an IKKβ inhibitor, could induce apoptosis of VCAM-1 positive cells. Our purpose is to prepare a novel tracer to evaluate its feasibility of detecting VCAM-1 expression and monitoring LY2409881 tumor curative effect. The tracer was prepared by conjugating the single chain variable fragment (scFv) of VCAM-1 and NOTA-NHS-ester and then labeled with 68Ga. 68Ga-NOTA-VCAM-1scFv was successfully prepared with high radiochemical yield. VCAM-1 overexpression and underexpression melanoma cell lines, B16F10 and A375m, were used in this study. The results of microPET/CT imaging in small animals indicated that the uptake of 68Ga-NOTA-VCAM-1scFv in B16F10 tumor was much higher than that of A375m, which was also confirmed by the biodistribution and autoradiography results. LY2409881 inhibits the growth of B16F10 melanoma in vivo by inducing dose- and time-dependent growth inhibition and apoptosis of the cells. The LY2409881 treated group and DMSO control group were established and imaged by microPET/CT. In the LY2409881 group, uptake of the tracer in tumor was decreased at the first week, and then gradually recovered to the initial level. In DMSO control, the uptake of the tracer remained at the same level during the whole time. The results suggested that LY2409881 inhibits the expression of VCAM-1 and suppresses tumor growth. 68Ga-NOTA-VCAM-1scFv, an easily synthesized probe, has a potential clinical application in the visual monitoring of IKKβ inhibitor intervention on VCAM-1 positive tumors.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Chunbao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Fan Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yingying Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, 710032, China
| | - Yongheng Gao
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, 710032, China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China.,Hubei Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430022, China
| |
Collapse
|
15
|
Vancurova I, Uddin MM, Zou Y, Vancura A. Combination Therapies Targeting HDAC and IKK in Solid Tumors. Trends Pharmacol Sci 2017; 39:295-306. [PMID: 29233541 DOI: 10.1016/j.tips.2017.11.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022]
Abstract
The rationale for developing histone deacetylase (HDAC) inhibitors (HDACi) as anticancer agents was based on their ability to induce apoptosis and cell cycle arrest in cancer cells. However, while HDACi have been remarkably effective in the treatment of hematological malignancies, clinical studies with HDACi as single agents in solid cancers have been disappointing. Recent studies have shown that, in addition to inducing apoptosis in cancer cells, class I HDACi induce IκB kinase (IKK)-dependent expression of proinflammatory chemokines, such as interleukin-8 (IL8; CXCL8), resulting in the increased proliferation of tumor cells, and limiting the effectiveness of HDACi in solid tumors. Here, we discuss the mechanisms responsible for HDACi-induced CXCL8 expression, and opportunities for combination therapies targeting HDACs and IKK in solid tumors.
Collapse
Affiliation(s)
- Ivana Vancurova
- Department of Biological Sciences, St John's University, New York, NY 11439, USA.
| | - Mohammad M Uddin
- Department of Biological Sciences, St John's University, New York, NY 11439, USA
| | - Yue Zou
- Department of Biological Sciences, St John's University, New York, NY 11439, USA
| | - Ales Vancura
- Department of Biological Sciences, St John's University, New York, NY 11439, USA
| |
Collapse
|
16
|
Quagliano A, Gopalakrishnapillai A, Barwe SP. Epigenetic drug combination overcomes osteoblast-induced chemoprotection in pediatric acute lymphoid leukemia. Leuk Res 2017; 56:36-43. [PMID: 28171800 PMCID: PMC5366080 DOI: 10.1016/j.leukres.2017.01.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/06/2023]
Abstract
Although there has been much progress in the treatment of acute lymphoblastic leukemia (ALL), decreased sensitivity to chemotherapy remains a significant issue. Recent studies have shown how interactions with the bone marrow microenvironment can protect ALL cells from chemotherapy and allow for the persistence of the disease. Epigenetic drugs have been used for the treatment of ALL, but there are no reports on whether these drugs can overcome bone marrow-induced chemoprotection. Our study investigates the ability of the DNA methyltransferase inhibitor azacitidine and the histone deacetylase inhibitor panobinostat to overcome chemoprotective effects mediated by osteoblasts. We show that the combination of azacitidine and panobinostat has a synergistic killing effect and that this combination is more effective than cytarabine in inducing ALL cell death in co-culture with osteoblasts. We also show that this combination can be used to sensitize ALL cells to chemotherapeutics in the presence of osteoblasts. Finally, we demonstrate that these effects can be replicated ex vivo in a number of mouse passaged xenograft lines from both B-ALL and T-ALL patients with varying cytogenetics. Thus, our data provides evidence that azacitidine and panobinostat can successfully overcome osteoblast-induced chemoprotection in vitro and ex vivo in both B-ALL and T-ALL cells.
Collapse
Affiliation(s)
- Anthony Quagliano
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, United States; Department of Biological Sciences, University of Delaware, Wilmington, DE 19716, United States
| | - Anilkumar Gopalakrishnapillai
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, United States.
| | - Sonali P Barwe
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, United States; Department of Biological Sciences, University of Delaware, Wilmington, DE 19716, United States.
| |
Collapse
|
17
|
Silencing c-Myc translation as a therapeutic strategy through targeting PI3Kδ and CK1ε in hematological malignancies. Blood 2016; 129:88-99. [PMID: 27784673 DOI: 10.1182/blood-2016-08-731240] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/12/2016] [Indexed: 12/20/2022] Open
Abstract
Phosphoinositide 3-kinase (PI3K) and the proteasome pathway are both involved in activating the mechanistic target of rapamycin (mTOR). Because mTOR signaling is required for initiation of messenger RNA translation, we hypothesized that cotargeting the PI3K and proteasome pathways might synergistically inhibit translation of c-Myc. We found that a novel PI3K δ isoform inhibitor TGR-1202, but not the approved PI3Kδ inhibitor idelalisib, was highly synergistic with the proteasome inhibitor carfilzomib in lymphoma, leukemia, and myeloma cell lines and primary lymphoma and leukemia cells. TGR-1202 and carfilzomib (TC) synergistically inhibited phosphorylation of the eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1), leading to suppression of c-Myc translation and silencing of c-Myc-dependent transcription. The synergistic cytotoxicity of TC was rescued by overexpression of eIF4E or c-Myc. TGR-1202, but not other PI3Kδ inhibitors, inhibited casein kinase-1 ε (CK1ε). Targeting CK1ε using a selective chemical inhibitor or short hairpin RNA complements the effects of idelalisib, as a single agent or in combination with carfilzomib, in repressing phosphorylation of 4E-BP1 and the protein level of c-Myc. These results suggest that TGR-1202 is a dual PI3Kδ/CK1ε inhibitor, which may in part explain the clinical activity of TGR-1202 in aggressive lymphoma not found with idelalisib. Targeting CK1ε should become an integral part of therapeutic strategies targeting translation of oncogenes such as c-Myc.
Collapse
|
18
|
Zullo KM, Guo Y, Cooke L, Jirau-Serrano X, Mangone M, Scotto L, Amengual JE, Mao Y, Nandakumar R, Cremers S, Duong J, Mahadevan D, O'Connor OA. Aurora A Kinase Inhibition Selectively Synergizes with Histone Deacetylase Inhibitor through Cytokinesis Failure in T-cell Lymphoma. Clin Cancer Res 2015; 21:4097-109. [PMID: 25878331 PMCID: PMC4581881 DOI: 10.1158/1078-0432.ccr-15-0033] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/24/2015] [Indexed: 01/23/2023]
Abstract
PURPOSE Aurora A kinase (AAK) is expressed exclusively during mitosis, and plays a critical role in centrosome duplication and spindle formation. Alisertib is a highly selective AAK inhibitor that has demonstrated marked clinical activity of alisertib across a spectrum of lymphomas, though particularly in patients with T-cell lymphoma (TCL). We sought to compare and contrast the activity of alisertib in preclinical models of B-cell lymphoma (BCL) and TCL, and identify combinations worthy of clinical study. High-throughput screening of pralatrexate, the proteasome inhibitor (ixazomib), and the histone deacetylase (HDAC) inhibitor (romidepsin) revealed that only romidepsin synergized with alisertib, and only in models of TCL. We discovered that the mechanism of synergy between AAK inhibitors and HDAC inhibitors appears to be mediated through cytokinesis failure. EXPERIMENTAL DESIGN A high-throughput screening approach was used to identify drugs that were potentially synergistic in combination with alisertib. Live-cell imaging was used to explore the mechanistic basis for the drug: drug interaction between alisertib and romidepsin. An in vivo xenograft TCL model was used to confirm in vitro results. RESULTS In vitro, alisertib exhibited concentration-dependent cytotoxicity in BCL and TCL cell lines. Alisertib was synergistic with romidepsin in a T-cell-specific fashion that was confirmed in vivo. Live-cell imaging demonstrated that the combination treatment resulted in profound cytokinesis failure. CONCLUSIONS These data strongly suggest that the combination of alisertib and romidepsin is highly synergistic in TCL through modulation of cytokinesis and merits clinical development.
Collapse
Affiliation(s)
- Kelly M Zullo
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Yige Guo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Laurence Cooke
- University of Tennessee Health Science Center, West Cancer Center, Memphis, Tennessee
| | - Xavier Jirau-Serrano
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Michael Mangone
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Luigi Scotto
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Jennifer E Amengual
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York
| | - Yinghui Mao
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Renu Nandakumar
- Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, New York
| | - Serge Cremers
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York. Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, New York
| | - Jimmy Duong
- Mailman School of Public Health, Columbia University, New York, New York
| | - Daruka Mahadevan
- University of Tennessee Health Science Center, West Cancer Center, Memphis, Tennessee
| | - Owen A O'Connor
- Department of Medicine, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, New York.
| |
Collapse
|
19
|
Abstract
INTRODUCTION Patients with relapsed or refractory lymphoma remain a population with unmet medical needs. Histone deacetylase inhibitors (HDACIs) represent a novel class of anticancer drugs currently in development in several malignancies. Inhibition of HDACs leads to acetylation of histone and non-histone proteins, which in turn results in epigenetic modification of gene expression that leads to a plethora of effects, such as cell cycle arrest, apoptosis and inhibition of angiogenesis. Romidepsin is a novel HDACI that has demonstrated preclinical and clinical activity. AREAS COVERED This review discusses the different HDACs and epigenetic regulation with a particular focus on the preclinical and clinical development of romidepsin in lymphoma. The review of romidepsin includes: the mechanism of action, its synergistic interaction with novel agents, pivotal clinical trials that lead to its US FDA approval in cutaneous T-cell lymphoma and peripheral T-cell lymphoma as well as active combinations currently in clinical trials. EXPERT OPINION Romidepsin is a potent HDACI with clinical activity in T-cell lymphoma where novel agents and combinations are desperately needed. A deeper understanding of the molecular characteristics of this class of agents will allow the design of more potent drugs with improved toxicity profiles and future rational combinations that will expand the indication and benefit from these novel agents.
Collapse
Affiliation(s)
- Victor Y Yazbeck
- Virginia Commonwealth University, Massey Cancer Center , Richmond, VA , USA
| | | |
Collapse
|
20
|
la Rosa AHD, Acker M, Swain S, Manoharan M. The role of epigenetics in kidney malignancies. Cent European J Urol 2015; 68:157-64. [PMID: 26251734 PMCID: PMC4526599 DOI: 10.5173/ceju.2015.453] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 02/03/2015] [Accepted: 02/20/2015] [Indexed: 01/18/2023] Open
Abstract
Introduction Renal cell carcinomas (RCC) are collectively the third most common type of genitourinary neoplasms, surpassed only by prostate and bladder cancer. Cure rates for renal cell carcinoma are related to tumor grade and stage; therefore, diagnostic methods for early detection and new therapeutic modalities are of paramount importance. Epigenetics can be defined as inherited modifications in gene expression that are not encoded in the DNA sequence itself. Epigenetics may play an important role in the pursuit of early diagnosis, accurate prognostication and identification of new therapeutic targets. Material and methods We used PubMed to conduct a comprehensive search of the English medical literature using search terms including epigenetics, DNA methylation, histone modification, microRNA regulation (miRNA) and RCC. In this review, we discuss the potential application of epigenetics in the diagnosis, prognosis and treatment of kidney cancer. Results During the last decade, many different types of epigenetic alterations of DNA have been found to be associated with malignant renal tumors. This has led to the research of the diagnostic and prognostic implications of these changes in renal malignancies as well as to the development of novel drugs to target these changes, with the aim of achieving a survival benefit. Conclusions Epigenetics has become a promising field in cancer research. The potential to achieve early detection and accurate prognostication in kidney cancer might be feasible through the application of epigenetics. The possibility to reverse these epigenetic changes with new therapeutic agents motivates researchers to continue pursuing better treatment options for kidney cancer and other malignancies.
Collapse
Affiliation(s)
| | - Matthew Acker
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sanjaya Swain
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Murugesan Manoharan
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|