1
|
Qi M, Yi X, Yue B, Huang M, Zhou S, Xiong J. S100A6 inhibits MDM2 to suppress breast cancer growth and enhance sensitivity to chemotherapy. Breast Cancer Res 2023; 25:55. [PMID: 37217945 DOI: 10.1186/s13058-023-01657-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/10/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND S100A6 and murine double minute 2 (MDM2) are important cancer-related molecules. A previous study identified an interaction between S100A6 and MDM2 by size exclusion chromatography and surface plasmon resonance experiments. The present study investigated whether S100A6 could bind to MDM2 in vivo and further explored its functional implication. METHODS Co-immunoprecipitation, glutathione-S-transferase pull-down assay, and immunofluorescence were performed to determine the in vivo interaction between S100A6 and MDM2. Cycloheximide pulse-chase assay and ubiquitination assay were performed to clarify the mechanism by which S100A6 downregulated MDM2. In addition, clonogenic assay, WST-1 assay, and flow cytometry of apoptosis and the cell cycle were performed and a xenograft model was established to evaluate the effects of the S100A6/MDM2 interaction on growth and paclitaxel-induced chemosensitivity of breast cancer. The expressions of S100A6 and MDM2 in patients with invasive breast cancer were analyzed by immunohistochemistry. In addition, the correlation between the expression of S100A6 and the response to neoadjuvant chemotherapy was statistically analyzed. RESULTS S100A6 promoted the MDM2 translocation from nucleus to cytoplasm, in which the S100A6 bound to the binding site of the herpesvirus-associated ubiquitin-specific protease (HAUSP) in MDM2, disrupted the MDM2-HAUSP-DAXX interactions, and induced the MDM2 self-ubiquitination and degradation. Furthermore, the S100A6-mediated MDM2 degradation suppressed the growth of breast cancer and enhanced its sensitivity to paclitaxel both in vitro and in vivo. For patients with invasive breast cancer who received epirubicin and cyclophosphamide followed by docetaxel (EC-T), expressions of S100A6 and MDM2 were negatively correlated, and high expression of S100A6 suggested a higher rate of pathologic complete response (pCR). Univariate and multivariate analyses showed that the high expression of S100A6 was an independent predictor of pCR. CONCLUSION These results reveal a novel function for S100A6 in downregulating MDM2, which directly enhances sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Mengxin Qi
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xianglan Yi
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Baohui Yue
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mingxiang Huang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Zhou
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jing Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Saadoune C, Nouadi B, Hamdaoui H, Chegdani F, Bennis F. Multiple Myeloma: Bioinformatic Analysis for Identification of Key Genes and Pathways. Bioinform Biol Insights 2022; 16:11779322221115545. [PMID: 35958298 PMCID: PMC9358573 DOI: 10.1177/11779322221115545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/26/2022] [Indexed: 01/02/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy in which monoclonal plasma cells multiply in the bone marrow and monoclonal immunoglobulins are overproduced in older people. Several molecular and cytogenetic advances allow scientists to identify several genetic and chromosomal abnormalities that cause the disease. The comprehension of the pathophysiology of MM requires an understanding of the characteristics of malignant clones and the changes in the bone marrow microenvironment. This study aims to identify the central genes and to determine the key signaling pathways in MM by in silico approaches. A list of 114 differentially expressed genes (DEGs) is important in the prognosis of MM. The DEGs are collected from scientific publications and databases (https://www.ncbi.nlm.nih.gov/). These data are analyzed by Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) software (https://string-db.org/) through the construction of protein-protein interaction (PPI) networks and enrichment analysis of the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, by CytoHubba, AutoAnnotate, Bingo Apps plugins in Cytoscape software (https://cytoscape.org/) and by DAVID database (https://david.ncifcrf.gov/). The analysis of the results shows that there are 7 core genes, including TP53; MYC; CDND1; IL6; UBA52; EZH2, and MDM2. These top genes appear to play a role in the promotion and progression of MM. According to functional enrichment analysis, these genes are mainly involved in the following signaling pathways: Epstein-Barr virus infection, microRNA pathway, PI3K-Akt signaling pathway, and p53 signaling pathway. Several crucial genes, including TP53, MYC, CDND1, IL6, UBA52, EZH2, and MDM2, are significantly correlated with MM, which may exert their role in the onset and evolution of MM.
Collapse
Affiliation(s)
- Chaimaa Saadoune
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Badreddine Nouadi
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Hasna Hamdaoui
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Casablanca, Morocco.,Laboratory of Medical Genetics, University Hospital Center Tangier-Tetouan-Al Hoceima, Tangier, Morocco
| | - Fatima Chegdani
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Faiza Bennis
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
3
|
Discovery of the oncogenic MDM2, a direct binding target of berberine and a potential therapeutic, in multiple myeloma. Funct Integr Genomics 2022; 22:1031-1041. [PMID: 35794284 DOI: 10.1007/s10142-022-00880-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/25/2022] [Accepted: 06/23/2022] [Indexed: 11/04/2022]
Abstract
Recent studies have suggested the potency of berberine (BBR) for multiple cancer treatments, including multiple myeloma (MM). However, the direct target and underlying mechanism of BBR remain largely understood in MM. Here, we demonstrated that BBR inhibited cell proliferation and acted synergistically with bortezomib in MM.1S cells. BBR treatment induced MM cell cycle arrest by downregulating several cell cycle-related proteins. Murine double minute 2 (MDM2) as a BBR-binding protein was identified by surface plasmon resonance image (SPRi) analysis and molecular docking. Overexpression of MDM2 is associated with MM progression and a poor prognosis. Knockdown MDM2 by siRNA transfection can repress MM malignant progression and attenuate the BBR sensitivity to MM.1S cells. BBR treatment induced the degradation of MDM2 through the ubiquitin-proteasome system and reactivated P53/P21 in MM cells. Overall, our data has illustrated that MDM2, as a binding protein of BBR for the first time, may serve as a potential therapeutic option for MM.
Collapse
|
4
|
Trasanidis N, Katsarou A, Ponnusamy K, Shen YA, Kostopoulos IV, Bergonia B, Keren K, Reema P, Xiao X, Szydlo RM, Sabbattini PMR, Roberts IAG, Auner HW, Naresh KN, Chaidos A, Wang TL, Magnani L, Caputo VS, Karadimitris A. Systems medicine dissection of chr1q-amp reveals a novel PBX1-FOXM1 axis for targeted therapy in multiple myeloma. Blood 2022; 139:1939-1953. [PMID: 35015835 DOI: 10.1182/blood.2021014391] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/20/2021] [Indexed: 11/20/2022] Open
Abstract
Understanding the biological and clinical impact of copy number aberrations (CNAs) on the development of precision therapies in cancer remains an unmet challenge. Genetic amplification of chromosome 1q (chr1q-amp) is a major CNA conferring an adverse prognosis in several types of cancer, including in the blood cancer multiple myeloma (MM). Although several genes across chromosome 1 (chr1q) portend high-risk MM disease, the underpinning molecular etiology remains elusive. Here, with reference to the 3-dimensional (3D) chromatin structure, we integrate multi-omics data sets from patients with MM with genetic variables to obtain an associated clinical risk map across chr1q and to identify 103 adverse prognosis genes in chr1q-amp MM. Prominent among these genes, the transcription factor PBX1 is ectopically expressed by genetic amplification and epigenetic activation of its own preserved 3D regulatory domain. By binding to reprogrammed superenhancers, PBX1 directly regulates critical oncogenic pathways and a FOXM1-dependent transcriptional program. Together, PBX1 and FOXM1 activate a proliferative gene signature that predicts adverse prognosis across multiple types of cancer. Notably, pharmacological disruption of the PBX1-FOXM1 axis with existing agents (thiostrepton) and a novel PBX1 small molecule inhibitor (T417) is selectively toxic against chr1q-amp myeloma and solid tumor cells. Overall, our systems medicine approach successfully identifies CNA-driven oncogenic circuitries, links them to clinical phenotypes, and proposes novel CNA-targeted therapy strategies in MM and other types of cancer.
Collapse
Affiliation(s)
- Nikolaos Trasanidis
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Alexia Katsarou
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Foundation Trust, London, United Kingdom
| | - Kanagaraju Ponnusamy
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Yao-An Shen
- Department of Pathology
- Department of Oncology
- Department of Gynecology and Obstetrics, and
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ioannis V Kostopoulos
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Bien Bergonia
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Keren Keren
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Paudel Reema
- Imperial Experimental Cancer Medicine Centre and Cancer Research UK Imperial Centre, London, United Kingdom
| | - Xiaolin Xiao
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Richard M Szydlo
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Pierangela M R Sabbattini
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Irene A G Roberts
- Department of Paediatrics and Medical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford University, Oxford, United Kingdom
- Oxford Biomedical Research Centre Blood Theme, National Institute for Health Research Oxford Biomedical Centre, Oxford, United Kingdom
| | - Holger W Auner
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Foundation Trust, London, United Kingdom
| | - Kikkeri N Naresh
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Foundation Trust, London, United Kingdom
- Imperial Experimental Cancer Medicine Centre and Cancer Research UK Imperial Centre, London, United Kingdom
| | - Aristeidis Chaidos
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Foundation Trust, London, United Kingdom
| | - Tian-Li Wang
- Department of Pathology
- Department of Oncology
- Department of Gynecology and Obstetrics, and
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| | - Valentina S Caputo
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Cancer Biology and Therapy Laboratory, School of Applied Science, London South Bank University, London, United Kingdom
| | - Anastasios Karadimitris
- Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
5
|
Targeting an MDM2/MYC Axis to Overcome Drug Resistance in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14061592. [PMID: 35326742 PMCID: PMC8945937 DOI: 10.3390/cancers14061592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND MDM2 is elevated in multiple myeloma (MM). Although traditionally, MDM2 negatively regulates p53, a growing body of research suggests that MDM2 plays several p53-independent roles in cancer pathogenesis as a regulator of oncogene mRNA stability and translation. Yet, the molecular mechanisms underlying MDM2 overexpression and its role in drug resistance in MM remain undefined. METHODS Both myeloma cell lines and primary MM samples were employed. Cell viability, cell cycle and apoptosis assays, siRNA transfection, quantitative real-time PCR, immunoblotting, co-immunoprecipitation (Co-IP), chromatin immunoprecipitation (ChIP), soft agar colony formation and migration assay, pulse-chase assay, UV cross-linking, gel-shift assay, RNA-protein binding assays, MEME-analysis for discovering c-Myc DNA binding motifs studies, reporter gene constructs procedure, gene transfection and reporter assay, MM xenograft mouse model studies, and statistical analysis were applied in this study. RESULTS We show that MDM2 is associated with poor prognosis. Importantly, its upregulation in primary MM samples and human myeloma cell lines (HMCLs) drives drug resistance. Inhibition of MDM2 by RNAi, or by the MDM2/XIAP dual inhibitor MX69, significantly enhanced the sensitivity of resistant HMCLs and primary MM samples to bortezomib and other anti-myeloma drugs, demonstrating that MDM2 can modulate drug response. MDM2 inhibition resulted in a remarkable suppression of relapsed MM cell growth, colony formation, migration and induction of apoptosis through p53-dependent and -independent pathways. Mechanistically, MDM2 was found to reciprocally regulate c-Myc in MM; MDM2 binds to AREs on c-Myc 3'UTR to increase c-Myc mRNA stability and translation, while MDM2 is a direct transcriptional target of c-Myc. MDM2 inhibition rendered c-Myc mRNA unstable, and reduced c-Myc protein expression in MM cells. Importantly, in vivo delivery of MX69 in combination with bortezomib led to significant regression of tumors and prolonged survival in an MM xenograft model. CONCLUSION Our findings provide a rationale for the therapeutic targeting of MDM2/c-Myc axis to improve clinical outcome of patients with refractory/relapsed MM.
Collapse
|
6
|
Pan D, Richter J. Where We Stand With Precision Therapeutics in Myeloma: Prosperity, Promises, and Pipedreams. Front Oncol 2022; 11:819127. [PMID: 35127532 PMCID: PMC8811139 DOI: 10.3389/fonc.2021.819127] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma remains an incurable disease despite numerous novel agents being approved in the last decade. Furthermore, disease behavior and susceptibility to current treatments often vary drastically from patient to patient. To date there are no approved therapies in myeloma that are targeted to specific patient populations based on genomic or immunologic findings. Precision medicine, using biomarkers descriptive of a specific tumor's biology and predictive of response to appropriate agents, may continue to push the field forward by expanding our treatment arsenal while refining our ability to expose patients to only those treatments likely to be efficacious. Extensive research efforts have been carried out in this endeavor including the use of agents targeting Bcl2 and the RAS/MAPK and PI3K/AKT/mTOR pathways. Thus far, clinical trials have yielded occasional successes intermixed with disappointments, reflecting significant hurdles which still remain including the complex crosstalk between oncogenic pathways and the nonlinear genetic development of myeloma, prone to cultivating sub-clones with distinctive mutations. In this review, we explore the landscape of precision therapeutics in multiple myeloma and underscore the degree to which research efforts have produced tangible clinical results.
Collapse
Affiliation(s)
- Darren Pan
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | |
Collapse
|
7
|
Liu J, Hideshima T, Xing L, Wang S, Zhou W, Samur MK, Sewastianik T, Ogiya D, An G, Gao S, Yang L, Ji T, Bianchi G, Wen K, Tai YT, Munshi N, Richardson P, Carrasco R, Cang Y, Anderson KC. ERK signaling mediates resistance to immunomodulatory drugs in the bone marrow microenvironment. SCIENCE ADVANCES 2021; 7:7/23/eabg2697. [PMID: 34088671 PMCID: PMC8177702 DOI: 10.1126/sciadv.abg2697] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/19/2021] [Indexed: 05/05/2023]
Abstract
Immunomodulatory drugs (IMiDs) have markedly improved patient outcome in multiple myeloma (MM); however, resistance to IMiDs commonly underlies relapse of disease. Here, we identify that tumor necrosis factor (TNF) receptor-associated factor 2 (TRAF2) knockdown (KD)/knockout (KO) in MM cells mediates IMiD resistance via activation of noncanonical nuclear factor κB (NF-κB) and extracellular signal-regulated kinase (ERK) signaling. Within MM bone marrow (BM) stromal cell supernatants, TNF-α induces proteasomal degradation of TRAF2, noncanonical NF-κB, and downstream ERK signaling in MM cells, whereas interleukin-6 directly triggers ERK activation. RNA sequencing of MM patient samples shows nearly universal ERK pathway activation at relapse on lenalidomide maintenance therapy, confirming its clinical relevance. Combination MEK inhibitor treatment restores IMiD sensitivity of TRAF2 KO cells both in vitro and in vivo. Our studies provide the framework for clinical trials of MEK inhibitors to overcome IMiD resistance in the BM microenvironment and improve patient outcome in MM.
Collapse
Affiliation(s)
- Jiye Liu
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Lijie Xing
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Su Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Wenrong Zhou
- Oncology and Immunology Unit, Research Service Division, WuXi AppTec (Shanghai) Co., Ltd., Shanghai 200131, China
| | - Mehmet K Samur
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Department of Biostatistics and Computational Biology, Harvard T.H. Chan School of Public Health, Boston, 02115 MA, USA
| | - Tomasz Sewastianik
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw 02776, Poland
| | - Daisuke Ogiya
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Gang An
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300020, China
| | - Shaobing Gao
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Li Yang
- Multiple Myeloma Treatment Center and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tong Ji
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China
| | - Giada Bianchi
- Division of Hematology, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Paul Richardson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Ruben Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yong Cang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
8
|
Hassan H, Szalat R. Genetic Predictors of Mortality in Patients with Multiple Myeloma. APPLICATION OF CLINICAL GENETICS 2021; 14:241-254. [PMID: 33953598 PMCID: PMC8092627 DOI: 10.2147/tacg.s262866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/31/2021] [Indexed: 12/19/2022]
Abstract
Multiple myeloma (MM) is a heterogeneous disease featured by clonal plasma cell proliferation and genomic instability. The advent of next-generation sequencing allowed unraveling the complex genomic landscape of the disease. Several recurrent genomic aberrations including immunoglobulin genes translocations, copy number abnormalities, complex chromosomal events, transcriptomic and epigenomic deregulation, and mutations define various molecular subgroups with distinct outcomes. In this review, we describe the recurrent genomic events identified in MM impacting patients’ outcome and survival. These genomic aberrations constitute new markers that could be incorporated into a prognostication model to eventually guide therapy at every stage of the disease.
Collapse
Affiliation(s)
- Hamza Hassan
- Department of Hematology and Medical Oncology, Boston University Medical Center, Boston, MA, USA
| | - Raphael Szalat
- Department of Hematology and Medical Oncology, Boston University Medical Center, Boston, MA, USA.,Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Wu Z, Gu L, Zhang S, Liu T, Lukka PB, Meibohm B, Bollinger JC, Zhou M, Li W. Discovery of N-(3,4-Dimethylphenyl)-4-(4-isobutyrylphenyl)-2,3,3a,4,5,9b-hexahydrofuro[3,2- c]quinoline-8-sulfonamide as a Potent Dual MDM2/XIAP Inhibitor. J Med Chem 2021; 64:1930-1950. [PMID: 33556244 PMCID: PMC9128806 DOI: 10.1021/acs.jmedchem.0c00932] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Murine double minute 2 (MDM2) and X-linked inhibitor of apoptosis protein (XIAP) are important cell survival proteins in tumor cells. As a dual MDM2/XIAP inhibitor reported previously, compound MX69 has low potency with an IC50 value of 7.5 μM against an acute lymphoblastic leukemia cell line EU-1. Herein, we report the structural optimization based on the MX69 scaffold, leading to the discovery of a 25-fold more potent analogue 14 (IC50 = 0.3 μM against EU-1). We demonstrate that 14 maintains its mode of action by dual targeting of MDM2 and XIAP through inducing MDM2 protein degradation and inhibiting XIAP mRNA translation, respectively, which resulted in cancer cell growth inhibition and cell death. The results strongly suggest that the scaffold based on 14 is promising for further optimization to develop a new therapeutic agent for leukemia and possibly other cancers where MDM2 and XIAP are dysregulated.
Collapse
Affiliation(s)
- Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Lubing Gu
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Sicheng Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Tao Liu
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Pradeep B Lukka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - John C Bollinger
- Department of Structural Biology, Biomolecular X-Ray Crystallography Center, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Muxiang Zhou
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
10
|
Conlon IL, Drennen B, Lanning ME, Hughes S, Rothhaas R, Wilder PT, MacKerell AD, Fletcher S. Rationally Designed Polypharmacology: α-Helix Mimetics as Dual Inhibitors of the Oncoproteins Mcl-1 and HDM2. ChemMedChem 2020; 15:1691-1698. [PMID: 32583936 PMCID: PMC8477420 DOI: 10.1002/cmdc.202000278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Protein-protein interactions (PPIs), many of which are dominated by α-helical recognition domains, play key roles in many essential cellular processes, and the dysregulation of these interactions can cause detrimental effects. For instance, aberrant PPIs involving the Bcl-2 protein family can lead to several diseases including cancer, neurodegenerative diseases, and diabetes. Interactions between Bcl-2 pro-life proteins, such as Mcl-1, and pro-death proteins, such as Bim, regulate the intrinsic pathway of apoptosis. p53, a tumor-suppressor protein, also has a pivotal role in apoptosis and is negatively regulated by its E3 ubiquitin ligase HDM2. Both Mcl-1 and HDM2 are upregulated in numerous cancers, and, interestingly, there is crosstalk between both protein pathways. Recently, synergy has been observed between Mcl-1 and HDM2 inhibitors. Towards the development of new anticancer drugs, we herein describe a polypharmacology approach for the dual inhibition of Mcl-1 and HDM2 by employing three densely functionalized isoxazoles, pyrazoles, and thiazoles as mimetics of key α-helical domains of their partner proteins.
Collapse
Affiliation(s)
- Ivie L Conlon
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Brandon Drennen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Maryanna E Lanning
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Samuel Hughes
- School of Chemistry, Cardiff University, Cardiff, CF10 3AT, UK
| | - Rebecca Rothhaas
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Paul T Wilder
- Department of Biochemistry and Molecular Biology Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| |
Collapse
|
11
|
He Y, Khan S, Huo Z, Lv D, Zhang X, Liu X, Yuan Y, Hromas R, Xu M, Zheng G, Zhou D. Proteolysis targeting chimeras (PROTACs) are emerging therapeutics for hematologic malignancies. J Hematol Oncol 2020; 13:103. [PMID: 32718354 PMCID: PMC7384229 DOI: 10.1186/s13045-020-00924-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional small molecules that utilize the ubiquitin proteasome system (UPS) to degrade proteins of interest (POI). PROTACs are potentially superior to conventional small molecule inhibitors (SMIs) because of their unique mechanism of action (MOA, i.e., degrading POI in a sub-stoichiometric manner), ability to target “undruggable” and mutant proteins, and improved target selectivity. Therefore, PROTACs have become an emerging technology for the development of novel targeted anticancer therapeutics. In fact, some of these reported PROTACs exhibit unprecedented efficacy and specificity in degrading various oncogenic proteins and have advanced to various stages of preclinical and clinical development for the treatment of cancer and hematologic malignancy. In this review, we systematically summarize the known PROTACs that have the potential to be used to treat various hematologic malignancies and discuss strategies to improve the safety of PROTACs for clinical application. Particularly, we propose to use the latest human pan-tissue single-cell RNA sequencing data to identify hematopoietic cell type-specific/selective E3 ligases to generate tumor-specific/selective PROTACs. These PROTACs have the potential to become safer therapeutics for hematologic malignancies because they can overcome some of the on-target toxicities of SMIs and PROTACs.
Collapse
Affiliation(s)
- Yonghan He
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Sajid Khan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Dongwen Lv
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Xuan Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Xingui Liu
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Yaxia Yuan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Robert Hromas
- Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mingjiang Xu
- Department of Molecular Medicine, College of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Prognosis, Biology, and Targeting of TP53 Dysregulation in Multiple Myeloma. Cells 2020; 9:cells9020287. [PMID: 31991614 PMCID: PMC7072230 DOI: 10.3390/cells9020287] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/10/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological cancer and is characterized by genetic features including translocations, chromosomal copy number aberrations, and mutations in key oncogene and tumor suppressor genes. Dysregulation of the tumor suppressor TP53 is important in the pathogenesis of many cancers, including MM. In newly-diagnosed MM patients, TP53 dysregulation occurs in three subsets: monoallelic deletion as part of deletion of chromosome 17p (del17p) (~8%), monoallelic mutations (~6%), and biallelic inactivation (~4%). Del17p is an established high-risk feature in MM and is included in current disease staging criteria. Biallelic inactivation and mutation have also been reported in MM patients but are not yet included in disease staging criteria for high-risk disease. Emerging clinical and genomics data suggest that the biology of high-risk disease is complex, and so far, traditional drug development efforts to target dysregulated TP53 have not been successful. Here we review the TP53 dysregulation literature in cancer and in MM, including the three segments of TP53 dysregulation observed in MM patients. We propose a reverse translational approach to identify novel targets and disease drivers from TP53 dysregulated patients to address the unmet medical need in this setting.
Collapse
|
13
|
Anwer F, Gee KM, Iftikhar A, Baig M, Russ AD, Saeed S, Zar MA, Razzaq F, Carew J, Nawrocki S, Al-Kateb H, Cavalcante Parr NN, McBride A, Valent J, Samaras C. Future of Personalized Therapy Targeting Aberrant Signaling Pathways in Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2019; 19:397-405. [PMID: 31036508 PMCID: PMC6626550 DOI: 10.1016/j.clml.2019.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/19/2019] [Accepted: 03/17/2019] [Indexed: 12/19/2022]
Abstract
Multiple myeloma (MM) is a genetically complex disease. Identification of mutations and aberrant signaling pathways that contribute to the progression of MM and drug resistance has potential to lead to specific targets and personalized treatment. Aberrant signal pathways include RAS pathway activation due to RAS or BRAF mutations (targeted by vemurafenib alone or combined with cobimetinib), BCL-2 overexpression in t(11:14) (targeted by venetoclax), JAK2 pathway activation (targeted by ruxolitinib), NF-κB pathway activation (treated with DANFIN combined with bortezomib), MDM2 overexpression, and PI3K/mTOR pathway activation (targeted by BEZ235). Cyclin D1 (CCND1) and MYC are also emerging as key potential targets. In addition, histone deacetylase inhibitors are already in use for the treatment of MM in combination therapy, and targeted inhibition of FGFR3 (AZD4547) is effective in myeloma cells with t(4;14) translocation. Bromodomain and extra terminal (BET) protein antagonists decrease the expression of MYC and have displayed promising antimyeloma activity. A better understanding of the alterations in signaling pathways that promote MM progression will further inform the development of precision therapy for patients.
Collapse
Affiliation(s)
- Faiz Anwer
- Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, OH.
| | - Kevin Mathew Gee
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ
| | - Ahmad Iftikhar
- Department of Medicine, The University of Arizona, Tucson, AZ
| | - Mirza Baig
- Department of Medicine, Summit Medical Group, Summit, NJ
| | | | - Sabina Saeed
- College of Public Health, The University of Arizona, Tucson, AZ
| | - Muhammad Abu Zar
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Faryal Razzaq
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Jennifer Carew
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Steffan Nawrocki
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Hussam Al-Kateb
- Division of Human Genetics, Children's Hospital, Cincinnati, OH
| | | | - Ali McBride
- College of Pharmacy, The University of Arizona, Tucson, AZ
| | - Jason Valent
- Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Christy Samaras
- Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
14
|
Li S, Vallet S, Sacco A, Roccaro A, Lentzsch S, Podar K. Targeting transcription factors in multiple myeloma: evolving therapeutic strategies. Expert Opin Investig Drugs 2019; 28:445-462. [DOI: 10.1080/13543784.2019.1605354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Shirong Li
- Division of Hematology/Oncology, Columbia University, New York, NY, USA
| | - Sonia Vallet
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Antonio Sacco
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Aldo Roccaro
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Suzanne Lentzsch
- Division of Hematology/Oncology, Columbia University, New York, NY, USA
| | - Klaus Podar
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| |
Collapse
|
15
|
Jovanović KK, Escure G, Demonchy J, Willaume A, Van de Wyngaert Z, Farhat M, Chauvet P, Facon T, Quesnel B, Manier S. Deregulation and Targeting of TP53 Pathway in Multiple Myeloma. Front Oncol 2019; 8:665. [PMID: 30687640 PMCID: PMC6333636 DOI: 10.3389/fonc.2018.00665] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple Myeloma (MM) is an incurable disease characterized by a clonal evolution across the course of the diseases and multiple lines of treatment. Among genomic drivers of the disease, alterations of the tumor suppressor TP53 are associated with poor outcomes. In physiological situation, once activated by oncogenic stress or DNA damage, p53 induces either cell-cycle arrest or apoptosis depending on the cellular context. Its inactivation participates to drug resistance in MM. The frequency of TP53 alterations increases along with the progression of the disease, from 5 at diagnosis to 75% at late relapses. Multiple mechanisms of regulation lead to decreased expression of p53, such as deletion 17p, TP53 mutations, specific microRNAs overexpression, TP53 promoter methylations, and MDM2 overexpression. Several therapeutic approaches aim to target the p53 pathway, either by blocking its interaction with MDM2 or by restoring the function of the altered protein. In this review, we describe the mechanism of deregulation of TP53 in MM, its role in MM progression, and the therapeutic options to interact with the TP53 pathway.
Collapse
Affiliation(s)
| | - Guillaume Escure
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| | - Jordane Demonchy
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| | | | | | - Meryem Farhat
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| | - Paul Chauvet
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| | - Thierry Facon
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| | - Bruno Quesnel
- IRCL, INSERM UMR-S1172, University of Lille, Lille, France
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| | - Salomon Manier
- IRCL, INSERM UMR-S1172, University of Lille, Lille, France
- Department of Hematology, CHU Lille, University of Lille, Lille, France
| |
Collapse
|
16
|
Cross Talk Networks of Mammalian Target of Rapamycin Signaling With the Ubiquitin Proteasome System and Their Clinical Implications in Multiple Myeloma. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 343:219-297. [PMID: 30712673 DOI: 10.1016/bs.ircmb.2018.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy and results from the clonal amplification of plasma cells. Despite recent advances in treatment, MM remains incurable with a median survival time of only 5-6years, thus necessitating further insights into MM biology and exploitation of novel therapeutic approaches. Both the ubiquitin proteasome system (UPS) and the PI3K/Akt/mTOR signaling pathways have been implicated in the pathogenesis, and treatment of MM and different lines of evidence suggest a close cross talk between these central cell-regulatory signaling networks. In this review, we outline the interplay between the UPS and mTOR pathways and discuss their implications for the pathophysiology and therapy of MM.
Collapse
|
17
|
Polymorphism inANRILis associated with relapse in patients with multiple myeloma after autologous stem cell transplant. Mol Carcinog 2017; 56:1722-1732. [DOI: 10.1002/mc.22626] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 12/18/2016] [Accepted: 01/30/2017] [Indexed: 12/12/2022]
|
18
|
Trino S, De Luca L, Laurenzana I, Caivano A, Del Vecchio L, Martinelli G, Musto P. P53-MDM2 Pathway: Evidences for A New Targeted Therapeutic Approach in B-Acute Lymphoblastic Leukemia. Front Pharmacol 2016; 7:491. [PMID: 28018226 PMCID: PMC5159974 DOI: 10.3389/fphar.2016.00491] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/30/2016] [Indexed: 11/13/2022] Open
Abstract
The tumor suppressor p53 is a canonical regulator of different biological functions, like apoptosis, cell cycle arrest, DNA repair, and genomic stability. This gene is frequently altered in human tumors generally by point mutations or deletions. Conversely, in acute lymphoblastic leukemia (ALL) genomic alterations of TP53 are rather uncommon, and prevalently occur in patients at relapse or with poor prognosis. On the other hand, p53 pathway is often compromised by the inactivation of its regulatory proteins, as MDM2 and ARF. MDM2 inhibitor molecules are able to antagonize p53-MDM2 interaction allowing p53 to exert tumor suppressor transcriptional regulation and to induce apoptotic pathways. Recent preclinical and clinical studies propose that MDM2 targeted therapy represents a promising anticancer strategy restoring p53 dependent mechanisms in ALL disease. Here, we discussed the use of new small molecule targeting p53 pathways as a promising drug target therapy in ALL.
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Luciana De Luca
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Antonella Caivano
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Luigi Del Vecchio
- CEINGE - Biotecnologie Avanzate S.C.a R.L.Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, Universita' degli Studi di Napoli Federico IINaples, Italy
| | - Giovanni Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. and A. Seràgnoli," University of Bologna Bologna, Italy
| | - Pellegrino Musto
- Scientific Direction, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| |
Collapse
|
19
|
Molecular Mechanisms of p53 Deregulation in Cancer: An Overview in Multiple Myeloma. Int J Mol Sci 2016; 17:ijms17122003. [PMID: 27916892 PMCID: PMC5187803 DOI: 10.3390/ijms17122003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/14/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022] Open
Abstract
The p53 pathway is inactivated in the majority of human cancers. Although this perturbation frequently occurs through the mutation or deletion of p53 itself, there are other mechanisms that can attenuate the pathway and contribute to tumorigenesis. For example, overexpression of important p53 negative regulators, such as murine double minute 2 (MDM2) or murine double minute 4 (MDM4), epigenetic deregulation, or even alterations in TP53 mRNA splicing. In this work, we will review the different mechanisms of p53 pathway inhibition in cancer with special focus on multiple myeloma (MM), the second most common hematological malignancy, with low incidence of p53 mutations/deletions but growing evidence of indirect p53 pathway deregulation. Translational implications for MM and cancer prognosis and treatment are also reviewed.
Collapse
|
20
|
Gu L, Zhang H, Liu T, Zhou S, Du Y, Xiong J, Yi S, Qu CK, Fu H, Zhou M. Discovery of Dual Inhibitors of MDM2 and XIAP for Cancer Treatment. Cancer Cell 2016; 30:623-636. [PMID: 27666947 PMCID: PMC5079537 DOI: 10.1016/j.ccell.2016.08.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/16/2015] [Accepted: 08/26/2016] [Indexed: 12/17/2022]
Abstract
MDM2 and XIAP are mutually regulated. Binding of MDM2 RING protein to the IRES region on XIAP mRNA results in MDM2 protein stabilization and enhanced XIAP translation. In this study, we developed a protein-RNA fluorescence polarization (FP) assay for high-throughput screening (HTS) of chemical libraries. Our FP-HTS identified eight inhibitors that blocked the MDM2 protein-XIAP RNA interaction, leading to MDM2 degradation. The compound-induced MDM2 downregulation resulted not only in inhibition of XIAP expression, but also in activation of p53, which contributed to cancer cell apoptosis in vitro and inhibition of cancer cell proliferation in vivo. Importantly, one of the MDM2/XIAP inhibitors, MX69, showed minimal inhibitory effect on normal human hematopoiesis in vitro and was very well tolerated in animal models.
Collapse
Affiliation(s)
- Lubing Gu
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Hailong Zhang
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Tao Liu
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Sheng Zhou
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuhong Du
- Department of Pharmacology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sha Yi
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Cheng-Kui Qu
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Haian Fu
- Department of Pharmacology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Muxiang Zhou
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
21
|
Anreddy N, Hazlehurst LA. Targeting Intrinsic and Extrinsic Vulnerabilities for the Treatment of Multiple Myeloma. J Cell Biochem 2016; 118:15-25. [PMID: 27261328 DOI: 10.1002/jcb.25617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder, clinically characterized by osteolytic lesions, immunodeficiency, and renal disease. Over the past decade, MM therapy is significantly improved by the introduction of novel therapeutics such as immunomodulatory agents (thalidomide, lenalidomide, and pomalidomide), proteasome inhibitors (bortezomib, carfilzomib, and ixazomib), monoclonal antibodies (daratumumab and elotuzumab), histone deacetylase (HDAC) inhibitors (Panobinostat). The clinical success of these agents has clearly identified vulnerabilities intrinsic to the MM cell, as well as targets that emanate from the tumor microenvironment. Despite these significant improvements, MM remains incurable due to the development of drug resistance. This perspective will discuss more recent strategies which take advantage of multiple targets within the proteome recycling pathway, chromatin remodeling, and disruption of nuclear export. In addition, we will review the development of strategies designed to block opportunistic survival signaling that occurs between the MM cell and the tumor microenvironment including strategies for inhibiting myeloma-induced immune suppression. It has become clear that MM tumors continue to evolve on therapy leading to drug resistance. It will be important to understand the emerging drug resistant mechanisms and additional vulnerabilities that occur due to the development of clinical resistance. J. Cell. Biochem. 118: 15-25, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nagaraju Anreddy
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506
| | - Lori A Hazlehurst
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506
| |
Collapse
|
22
|
Maneix L, Catic A. Touch and go: nuclear proteolysis in the regulation of metabolic genes and cancer. FEBS Lett 2016; 590:908-23. [PMID: 26832397 PMCID: PMC4833644 DOI: 10.1002/1873-3468.12087] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/15/2016] [Accepted: 01/26/2016] [Indexed: 01/07/2023]
Abstract
The recruitment of transcription factors to promoters and enhancers is a critical step in gene regulation. Many of these proteins are quickly removed from DNA after they completed their function. Metabolic genes in particular are dynamically regulated and continuously adjusted to cellular requirements. Transcription factors controlling metabolism are therefore under constant surveillance by the ubiquitin–proteasome system, which can degrade DNA‐bound proteins in a site‐specific manner. Several of these metabolic transcription factors are critical to cancer cells, as they promote uncontrolled growth and proliferation. This review highlights recent findings in the emerging field of nuclear proteolysis and outlines novel paradigms for cancer treatment, with an emphasis on multiple myeloma.
Collapse
Affiliation(s)
- Laure Maneix
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - André Catic
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
23
|
Lub S, Maes K, Menu E, De Bruyne E, Vanderkerken K, Van Valckenborgh E. Novel strategies to target the ubiquitin proteasome system in multiple myeloma. Oncotarget 2016; 7:6521-37. [PMID: 26695547 PMCID: PMC4872730 DOI: 10.18632/oncotarget.6658] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/23/2015] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the accumulation of plasma cells in the bone marrow (BM). The success of the proteasome inhibitor bortezomib in the treatment of MM highlights the importance of the ubiquitin proteasome system (UPS) in this particular cancer. Despite the prolonged survival of MM patients, a significant amount of patients relapse or become resistant to therapy. This underlines the importance of the development and investigation of novel targets to improve MM therapy. The UPS plays an important role in different cellular processes by targeted destruction of proteins. The ubiquitination process consists of enzymes that transfer ubiquitin to proteins targeting them for proteasomal degradation. An emerging and promising approach is to target more disease specific components of the UPS to reduce side effects and overcome resistance. In this review, we will focus on different components of the UPS such as the ubiquitin activating enzyme E1, the ubiquitin conjugating enzyme E2, the E3 ubiquitin ligases, the deubiquitinating enzymes (DUBs) and the proteasome. We will discuss their role in MM and the implications in drug discovery for the treatment of MM.
Collapse
Affiliation(s)
- Susanne Lub
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Van Valckenborgh
- Laboratory of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
24
|
Liu T, Zhang H, Xiong J, Yi S, Gu L, Zhou M. Inhibition of MDM2 homodimerization by XIAP IRES stabilizes MDM2, influencing cancer cell survival. Mol Cancer 2015; 14:65. [PMID: 25888903 PMCID: PMC4379586 DOI: 10.1186/s12943-015-0334-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 03/05/2015] [Indexed: 04/13/2023] Open
Abstract
Background It is known that the MDM2 protein is stabilized when it forms a heterodimer with its partner MDM4, but MDM2 protein stability in its homodimer form is not known. The MDM2 protein contains a C-terminal RING domain that not only functions as an E3 ligase to regulate ubiquitination of p53 and MDM2 itself, but also is characterized to be able to bind several specific cellular mRNAs to regulate gene expression. In this study, we evaluate whether the MDM2 protein stability is regulated by the binding of a specific small RNA (XIAP IRES mRNA). Methods We performed chemical cross-linking and bimolecular fluorescence complementation (BiFC) assay to measure the human MDM2 protein stability in its homodimer form and the effect of XIAP IRES on MDM2 homodimerization and protein stabilization. Ubiquitination and pulse-chase assays were used to detect MDM2 self-ubiquitination and protein turn-over. Fluorescent titration and ITC were used to examine the binding between MDM2 RING protein and XIAP IRES. Western blot assay was used for determining protein expression. Clonogenic assay, WST and flow cytometry were used to test the effects of XIAP IRES, siXIAP and IR on cancer cell growth and apoptosis. Results We found that self-association (homodimerization) of MDM2 occurs through the C-terminal RING domain of MDM2 and that the MDM2 protein becomes unstable when it is homodimerized. MDM2 homodimerization resulted in an increased function of the RING domain for MDM2 self-ubiquitination. Binding of XIAP IRES to the RING domain inhibited MDM2 homodimerization and self-ubiquitination, which resulted in stabilization of MDM2, as well as increased XIAP expression. Upregulation of XIAP and MDM2 that led to inhibition of p53 by the XIAP IRES resulted in cell growth and survival in both p53-normal and -deficient cancer cells. Conclusions Our study identified a new IRES RNA that interacts with MDM2 protein and regulates its stabilization, which suggested that targeting of MDM2 through disruption of MDM2 protein-RNA interaction might be a useful strategy for developing novel anti-cancer therapeutics.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA, 30322, USA.
| | - Hailong Zhang
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA, 30322, USA.
| | - Jing Xiong
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA, 30322, USA.
| | - Sha Yi
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA, 30322, USA.
| | - Lubing Gu
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA, 30322, USA.
| | - Muxiang Zhou
- Department of Pediatrics and Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA, 30322, USA.
| |
Collapse
|
25
|
Gu D, Wang S, Kuiatse I, Wang H, He J, Dai Y, Jones RJ, Bjorklund CC, Yang J, Grant S, Orlowski RZ. Inhibition of the MDM2 E3 Ligase induces apoptosis and autophagy in wild-type and mutant p53 models of multiple myeloma, and acts synergistically with ABT-737. PLoS One 2014; 9:e103015. [PMID: 25181509 PMCID: PMC4151993 DOI: 10.1371/journal.pone.0103015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 06/25/2014] [Indexed: 02/05/2023] Open
Abstract
Intracellular proteolytic pathways have been validated as rational targets in multiple myeloma with the approval of two proteasome inhibitors in this disease, and with the finding that immunomodulatory agents work through an E3 ubiquitin ligase containing Cereblon. Another E3 ligase that could be a rational target is the murine double minute (MDM) 2 protein, which plays a role in p53 turnover. A novel inhibitor of this complex, MI-63, was found to induce apoptosis in p53 wild-type myeloma models in association with activation of a p53-mediated cell death program. MI-63 overcame adhesion-mediated drug resistance, showed anti-tumor activity in vivo, enhanced the activity of bortezomib and lenalidomide, and also overcame lenalidomide resistance. In mutant p53 models, inhibition of MDM2 with MI-63 also activated apoptosis, albeit at higher concentrations, and this was associated with activation of autophagy. When MI-63 was combined with the BH3 mimetic ABT-737, enhanced activity was seen in both wild-type and mutant p53 models. Finally, this regimen showed efficacy against primary plasma cells from patients with newly diagnosed and relapsed/refractory myeloma. These findings support the translation of novel MDM2 inhibitors both alone, and in combination with other novel agents, to the clinic for patients with multiple myeloma.
Collapse
Affiliation(s)
- Dongmin Gu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Shuhong Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Isere Kuiatse
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hua Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jin He
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yun Dai
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Richard J. Jones
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Chad C. Bjorklund
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jing Yang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Steven Grant
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert Z. Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
26
|
Rocci A, Hofmeister CC, Pichiorri F. The potential of miRNAs as biomarkers for multiple myeloma. Expert Rev Mol Diagn 2014; 14:947-59. [DOI: 10.1586/14737159.2014.946906] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
27
|
p53 abnormalities and potential therapeutic targeting in multiple myeloma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:717919. [PMID: 25028664 PMCID: PMC4083709 DOI: 10.1155/2014/717919] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/20/2014] [Indexed: 01/02/2023]
Abstract
p53 abnormalities are regarded as an independent prognostic marker in multiple myeloma. Patients harbouring this genetic anomaly are commonly resistant to standard therapy. Thus, various p53 reactivating agents have been developed in order to restore its tumour suppressive abilities. Small molecular compounds, especially, have gained popularity in its efficacy against myeloma cells. For instance, promising preclinical results have steered both nutlin-3 and PRIMA-1 into phase I/II clinical trials. This review summarizes different modes of p53 inactivation in myeloma and highlights the current p53-based therapies that are being utilized in the clinic. Finally, we discuss the potential and promise that the novel small molecules possess for clinical application in improving the treatment outcome of myeloma.
Collapse
|
28
|
Cao MN, Zhou YB, Gao AH, Cao JY, Gao LX, Sheng L, Xu L, Su MB, Cao XC, Han MM, Wang MK, Li J. Curcusone D, a novel ubiquitin–proteasome pathway inhibitor via ROS-induced DUB inhibition, is synergistic with bortezomib against multiple myeloma cell growth. Biochim Biophys Acta Gen Subj 2014; 1840:2004-13. [DOI: 10.1016/j.bbagen.2014.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 01/24/2014] [Accepted: 02/04/2014] [Indexed: 12/13/2022]
|
29
|
Teoh PJ, Chung TH, Sebastian S, Choo SN, Yan J, Ng SB, Fonseca R, Chng WJ. p53 haploinsufficiency and functional abnormalities in multiple myeloma. Leukemia 2014; 28:2066-74. [DOI: 10.1038/leu.2014.102] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/20/2014] [Accepted: 03/06/2014] [Indexed: 12/28/2022]
|
30
|
Targeting the ubiquitin proteasome system in haematological malignancies. Blood Rev 2013; 27:297-304. [PMID: 24183816 DOI: 10.1016/j.blre.2013.10.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 02/07/2023]
Abstract
The ubiquitin proteasome system (UPS) plays a central role in cellular protein homeostasis through the targeted destruction of damaged/misfolded proteins and regulatory proteins that control critical cellular functions. The UPS comprises a sequential series of enzymatic activities to covalently attach ubiquitin to proteins to target them for degradation through the proteasome. Aberrancies within this system have been associated with transformation and tumourigenesis and thus, the UPS represents an attractive target for the development of anti-cancer therapies. The use of the first-in-class proteasome inhibitor, bortezomib, in the treatment of Plasma Cell Myeloma and Mantle Cell Lymphoma has validated the UPS as a therapeutic target. Following on its success, efforts are focused on the development of second-generation proteasome inhibitors and small molecule inhibitors of other components of the UPS. This review will provide an overview of the UPS and discuss current and novel therapies targeting the UPS.
Collapse
|
31
|
Natoni A, Coyne MRE, Jacobsen A, Rainey MD, O’Brien G, Healy S, Montagnoli A, Moll J, O’Dwyer M, Santocanale C. Characterization of a Dual CDC7/CDK9 Inhibitor in Multiple Myeloma Cellular Models. Cancers (Basel) 2013; 5:901-18. [PMID: 24202326 PMCID: PMC3795371 DOI: 10.3390/cancers5030901] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/04/2013] [Accepted: 07/04/2013] [Indexed: 12/14/2022] Open
Abstract
Two key features of myeloma cells are the deregulation of the cell cycle and the dependency on the expression of the BCL2 family of anti-apoptotic proteins. The cell division cycle 7 (CDC7) is an essential S-phase kinase and emerging CDC7 inhibitors are effective in a variety of preclinical cancer models. These compounds also inhibit CDK9 which is relevant for MCL-1 expression. The activity and mechanism of action of the dual CDC7/CDK9 inhibitor PHA-767491 was assessed in a panel of multiple myeloma cell lines, in primary samples from patients, in the presence of stromal cells and in combination with drugs used in current chemotherapeutic regimens. We report that in all conditions myeloma cells undergo cell death upon PHA-767491 treatment and we report an overall additive effect with melphalan, bortezomib and doxorubicin, thus supporting further assessment of targeting CDC7 and CDK9 in multiple myeloma.
Collapse
Affiliation(s)
- Alessandro Natoni
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
| | - Mark R. E. Coyne
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
- Department of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Haematology, Galway University Hospital, Galway, Ireland
| | - Alan Jacobsen
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
| | - Michael D. Rainey
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
| | - Gemma O’Brien
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
| | - Sandra Healy
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
| | - Alessia Montagnoli
- Nerviano Medical Sciences S.r.l., Via Pasteur 10, Nerviano 20014, Italy; E-Mail:
| | - Jürgen Moll
- Nerviano Medical Sciences S.r.l., Via Pasteur 10, Nerviano 20014, Italy; E-Mail:
| | - Michael O’Dwyer
- Department of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Haematology, Galway University Hospital, Galway, Ireland
- Authors to whom correspondence should be addressed; E-Mails: (M.O.); (C.S.); Tel.: +353-91-544-281 (M.O.); Fax: +353-91-542-469 (M.O.); Tel.: +353-91-495-174 (C.S.); Fax: +353-91-495-547 (C.S.)
| | - Corrado Santocanale
- Centre for Chromosome Biology, School of Natural Sciences National University of Ireland Galway, Galway, Ireland; E-Mails: (A.N.); (M.R.E.C.); (A.J.); (M.D.R.); (G.O.); (S.H.)
- Authors to whom correspondence should be addressed; E-Mails: (M.O.); (C.S.); Tel.: +353-91-544-281 (M.O.); Fax: +353-91-542-469 (M.O.); Tel.: +353-91-495-174 (C.S.); Fax: +353-91-495-547 (C.S.)
| |
Collapse
|
32
|
Pichiorri F, Suh SS, Rocci A, De Luca L, Taccioli C, Santhanam R, Wenchao Z, Benson DM, Hofmainster C, Alder H, Garofalo M, Di Leva G, Volinia S, Lin HJ, Perrotti D, Kuehl M, Aqeilan RI, Palumbo A, Croce CM. RETRACTED: Downregulation of p53-inducible microRNAs 192, 194, and 215 impairs the p53/MDM2 autoregulatory loop in multiple myeloma development. Cancer Cell 2010; 18:367-81. [PMID: 20951946 PMCID: PMC3561766 DOI: 10.1016/j.ccr.2010.09.005] [Citation(s) in RCA: 347] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 06/16/2010] [Accepted: 08/09/2010] [Indexed: 01/24/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the editors. This article was published on October 19, 2010, and Figures 4A and 7A were later corrected on August 8, 2016. In January 2021, The Ohio State University notified the Cancer Cell editors that an internal investigation concluded that Figures 1E, 4A, 4D, 5A, and 7A were falsified and that part of Figure 1E of the article is the same as part of Figure 1F in the correction of another article (Pichiorri et al., 2017, J. Exp. Med., 214, 1557, https://doi.org/10.1084/jem.2012095001172017c) and recommended retraction of the article. The editors no longer have confidence in the validity of the data and are retracting the article. S.-S. S. agrees with the retraction, and F.P., C.H., A.P., and C.M.C. disagree with the retraction; all other authors couldn't be reached or didn't respond.
Collapse
Affiliation(s)
- Flavia Pichiorri
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
- IMRIC-The Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel 91120
| | - Sung-Suk Suh
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
- IMRIC-The Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel 91120
| | - Alberto Rocci
- Division of Hematology, University of Turin, Turin, Italy, 10149
| | - Luciana De Luca
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
- Molecular Oncology Unit, IRCCS, Referral Cancer Center of Basilicata –Crob, Rionero in Vulture (PZ), Italy, 85028
| | - Cristian Taccioli
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
- Cancer Institute - University College London, London WC1E
| | - Ramasamy Santhanam
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
| | - Zhou Wenchao
- Medical Technology Division, School of Allied Medical Professions, The Ohio state University, Columbus, OH 43210
| | - Don M. Benson
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, The Ohio state University, Columbus, OH 43210
| | - Craig Hofmainster
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, The Ohio state University, Columbus, OH 43210
| | - Hansjuerg Alder
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
| | - Michela Garofalo
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
| | - Gianpiero Di Leva
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
| | - Stefano Volinia
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
- Telethon Facility-Data Mining for Analysis of DNA Microarrays, Department of Morphology and Embryology, University of Ferrara, Ferrara, Italy
| | - Huey-Jen Lin
- Medical Technology Division, School of Allied Medical Professions, The Ohio state University, Columbus, OH 43210
| | - Danilo Perrotti
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
| | - Michael Kuehl
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20889-5105
| | - Rami I. Aqeilan
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
- IMRIC-The Lautenberg Center for Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel 91120
| | - Antonio Palumbo
- Division of Hematology, University of Turin, Turin, Italy, 10149
| | - Carlo M. Croce
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, The Ohio State; University, Columbus, OH 43210
| |
Collapse
|
33
|
Genolet R, Rahim G, Gubler-Jaquier P, Curran J. The translational response of the human mdm2 gene in HEK293T cells exposed to rapamycin: a role for the 5'-UTRs. Nucleic Acids Res 2010; 39:989-1003. [PMID: 20876686 PMCID: PMC3035446 DOI: 10.1093/nar/gkq805] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Polysomal messenger RNA (mRNA) populations change rapidly in response to alterations in the physiological status of the cell. For this reason, translational regulation, mediated principally at the level of initiation, plays a key role in the maintenance of cellular homeostasis. In an earlier translational profiling study, we followed the impact of rapamycin on polysome re-seeding. Despite the overall negative effect on transcript recruitment, we nonetheless observed that some mRNAs were significantly less affected. Consequently, their relative polysomal occupancy increased in the rapamycin-treated cells. The behaviour of one of these genes, mdm2, has been further analysed. Despite the absence of internal ribosome entry site activity we demonstrate, using a dual reporter assay, that both the reported mdm2 5′-UTRs confer resistance to rapamycin relative to the 5′-UTR of β-actin. This relative resistance is responsive to the downstream targets mTORC1 but did not respond to changes in the La protein, a reported factor acting positively on MDM2 translational expression. Furthermore, extended exposure to rapamycin in the presence of serum increased the steady-state level of the endogenous MDM2 protein. However, this response was effectively reversed when serum levels were reduced. Taken globally, these studies suggest that experimental conditions can dramatically modulate the expressional output during rapamycin exposure.
Collapse
Affiliation(s)
- Raphael Genolet
- Department of Microbiology and Molecular Medicine, University of Geneva Medical School (CMU) 1, rue Michel Servet, CH-1205 Geneva, Switzerland
| | | | | | | |
Collapse
|
34
|
Wong WWL, Clendening JW, Martirosyan A, Boutros PC, Bros C, Khosravi F, Jurisica I, Stewart AK, Bergsagel PL, Penn LZ. Determinants of sensitivity to lovastatin-induced apoptosis in multiple myeloma. Mol Cancer Ther 2007; 6:1886-97. [PMID: 17575117 DOI: 10.1158/1535-7163.mct-06-0745] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Statins, commonly used to treat hypercholesterolemia, have been shown to trigger tumor-specific apoptosis in certain cancers, including multiple myeloma (MM), a plasma cell malignancy with poor prognosis. In this article, we show that of a panel of 17 genetically distinct MM cell lines, half were sensitive to statin-induced apoptosis and, despite pharmacodynamic evidence of drug uptake and activity, the remainder were insensitive. Sensitive cells were rescued from lovastatin-induced apoptosis by mevalonate, geranylgeranyl PPi, and partially by farnesyl PPi, highlighting the importance of isoprenylation. Expression profiling revealed that Rho GTPase mRNAs were differentially expressed upon lovastatin exposure in sensitive cells, yet ectopic expression of constitutively active Rho or Ras proteins was insufficient to alter sensitivity to lovastatin-induced apoptosis. This suggests that sensitivity involves more than one isoprenylated protein and that statins trigger apoptosis by blocking many signaling cascades, directly or indirectly deregulated by the oncogenic lesions of the tumor cell. Indeed, clustering on the basis of genetic abnormalities was shown to be significantly associated with sensitivity (P = 0.003). These results suggest that statins may be a useful molecular targeted therapy in the treatment of a subset of MM.
Collapse
Affiliation(s)
- W Wei-Lynn Wong
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sauthoff H, Pipiya T, Chen S, Heitner S, Cheng J, Huang YQ, Rom WN, Hay JG. Modification of the p53 transgene of a replication-competent adenovirus prevents mdm2- and E1b-55kD-mediated degradation of p53. Cancer Gene Ther 2006; 13:686-95. [PMID: 16470212 DOI: 10.1038/sj.cgt.7700936] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Clinical efficacy of adenovirus-mediated cancer gene therapy has been limited thus far. To improve its oncolytic effect, a replication-competent adenoviral vector was previously constructed to express high levels of p53 at a late time point in the viral life cycle. p53 expression from this vector improved tumor cell killing and viral spread in vitro. However, p53 function is antagonized by cellular mdm2 and adenoviral E1b-55kD, both of which are known to bind to and inactivate p53. Therefore, a new vector (Adp53W23S) that expresses a modified p53 transgene, which does not bind to E1b-55kd and mdm2, was constructed. The modified p53 protein was demonstrated to have a substantially prolonged half-life, and its localization was predominantly nuclear. Viral replication was unaffected by expression of the modified p53 and cancer cell killing was improved in vitro. However, in a xenograft model, efficacy was not significantly different from control virus. In conclusion, expression of a degradation-resistant p53 transgene late in the life cycle of a replication-competent adenovirus improves p53 stability and cancer cell killing in vitro. However, other factors, such as the adenoviral E1b-19kD and E1a proteins, which oppose p53 function, and limitations to viral spread need to be addressed to further improve in vivo efficacy.
Collapse
Affiliation(s)
- H Sauthoff
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, New York University School of Medicine, New York 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Stühmer T, Chatterjee M, Hildebrandt M, Herrmann P, Gollasch H, Gerecke C, Theurich S, Cigliano L, Manz RA, Daniel PT, Bommert K, Vassilev LT, Bargou RC. Nongenotoxic activation of the p53 pathway as a therapeutic strategy for multiple myeloma. Blood 2005; 106:3609-17. [PMID: 16081689 DOI: 10.1182/blood-2005-04-1489] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Mutation of p53 is a rare event in multiple myeloma, but it is unknown if p53 signaling is functional in myeloma cells, and if targeted nongenotoxic activation of the p53 pathway is sufficient to kill tumor cells. Here, we demonstrate that treatment of primary tumor samples with a small-molecule inhibitor of the p53-murine double minute 2 (MDM2) interaction increases the level of p53 and induces p53 targets and apoptotic cell death. Significantly, given the importance of the bone marrow microenvironment for the support and drug resistance of myeloma cells, tumor cells undergo effective apoptosis also in the presence of stromal cells, which themselves appear to tolerate exposure to nutlin-3. The in vitro toxicity of nutlin-3 was similar to that of the genotoxic drug melphalan. Because nutlin-mediated p53 activation is not dependent on DNA damage, MDM2 antagonists may help to avoid or reduce the severe genotoxic side effects of chemotherapeutic agents currently used to treat multiple myeloma. Therefore, MDM2 antagonists may offer a new treatment option for this disease.
Collapse
Affiliation(s)
- Thorsten Stühmer
- Department of Internal Medicine II, Division of Hematology and Oncology, University Clinics Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wu HC, Lu TY, Lee JJ, Hwang JK, Lin YJ, Wang CK, Lin CT. MDM2 expression in EBV-infected nasopharyngeal carcinoma cells. J Transl Med 2004; 84:1547-56. [PMID: 15448710 DOI: 10.1038/labinvest.3700183] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
To understand whether the p53-regulated mdm2 gene expression was altered by the Epstein-Barr virus (EBV) in nasopharyngeal carcinoma (NPC), the NPC-TW01 cell line was infected by EBV through IgA receptor-mediated endocytosis. The mdm2 gene was expressed only in a small fraction of the NPC cell population and could be enhanced in the EBV-infected (EBV+) cells. In the animals bearing EBV+ and EBV- NPC xenografts, the MDM2+ cells only appeared in clusters in both EBV+ and EBV- tumors with stronger expression in EBV+ cells. Cotransfection of pmdm2-Luc plus pSV40-p53 plus pCMV-LMP1 in the NPC-TW06 line that had p53 heterozygous point mutation showed stronger mdm2 promoter activity than cells cotransfected with pmdm2-Luc plus pSV40-p53, but no mdm2 promoter activity was seen in cells cotransfected with pmdm2-Luc plus pCMV-LMP1. Only the EBV-LMP1 but not the EBV-LMP2A gene could enhance p53 to upregulated mdm2 expression. Tumor cells in NPC biopsy specimens revealed similar mdm2 expression as in the animal model. It is concluded that although EBV can indirectly enhance mdm2 gene expression in tumor cells that express this gene, it cannot turn on or directly regulate mdm2 expression in cells that do not express this gene. In other words, EBV plays a role as an enhancer in NPC tumorigenesis.
Collapse
Affiliation(s)
- Han-Chung Wu
- Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
38
|
Zhang Z, Wang H, Prasad G, Li M, Yu D, Bonner JA, Agrawal S, Zhang R. Radiosensitization by antisense anti-MDM2 mixed-backbone oligonucleotide in in vitro and in vivo human cancer models. Clin Cancer Res 2004; 10:1263-73. [PMID: 14977824 DOI: 10.1158/1078-0432.ccr-0245-03] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE The MDM2 oncogene, amplified or overexpressed in many human cancers, has been suggested to be a novel target for cancer therapy. We have demonstrated a second-generation antisense antihuman-MDM2 oligonucleotide to have antitumor activity when administered alone or in combination with cancer chemotherapeutic agents. In the present study, we investigated the effect of the antisense oligonucleotide on radiation therapy. EXPERIMENTAL DESIGN The in vitro radiosensitization activity was determined in cell lines of human cancers of prostate (LNCaP and PC3), breast (MCF-7 and MDA-MB-468), pancreas (PANC-1), and glioma (U87-MG and A172) and its in vivo radiosensitization activity in xenograft models of LNCaP, PC3, MCF-7, MDA-MB-468, and PANC-1. RESULTS In cells containing at least one functional p53 allele (LNCaP, U87-MG, and A172), after specific inhibition of MDM2 expression, p53 and p21 levels were elevated. In LNCaP cells, the Bax level was increased, and Bcl-2 and E2F1 levels were decreased. In PC3 cells that are p53 null, after inhibition of MDM2 expression, Bax and p21 levels were elevated, and E2F1 levels were decreased. On the basis of in vitro clonogenic assay, the antisense oligonucleotide, in a sequence-specific manner, significantly increased radiation-induced antiproliferation effects. It also increased radiation-induced inhibitory effects on tumor growth in SCID or nude mice bearing LNCaP, PC3, MCF-7, MDA-MB-468, and PANC-1 xenografts. CONCLUSIONS These results suggest that MDM2 has a role in radiation therapy of human cancers, regardless of p53 status, providing a basis for future development of MDM2 inhibitors, such as antisense oligonucleotides, as radiosensitizers.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, Chesi M, Davies FE, Drach J, Greipp PR, Kirsch IR, Kuehl WM, Hernandez JM, Minvielle S, Pilarski LM, Shaughnessy JD, Stewart AK, Avet-Loiseau H. Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 2004; 64:1546-58. [PMID: 14989251 DOI: 10.1158/0008-5472.can-03-2876] [Citation(s) in RCA: 479] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Much has been learned regarding the biology and clinical implications of genetic abnormalities in multiple myeloma. Because of recent advances in the field, an International Workshop was held in Paris in february of 2003. This summary describes the consensus recommendations arising from that meeting with special emphasis on novel genetic observations. For instance, it is increasingly clear that translocations involving the immunoglobin heavy-chain locus are important for the pathogenesis of one-half of patients. As a corollary, it also clear that the remaining patients, lacking IgH translocations, have hyperdiploidy as the hallmark of their disease. Several important genetic markers are associated with a shortened survival such as chromosome 13 monosomy, hypodiploidy, and others. The events leading the transformation of the monoclonal gammopathy of undetermined significance (MGUS) to myeloma are still unclear. One of the few differential genetic lesions between myeloma and MGUS is the presence of ras mutations in the latter. Gene expression platforms are capable of detecting many of the genetic aberrations found in the clonal cells of myeloma. Areas in need of further study were identified. The study of the genetic aberrations will likely form the platform for targeted therapy for the disease.
Collapse
Affiliation(s)
- Rafael Fonseca
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang H, Oliver P, Zhang Z, Agrawal S, Zhang R. Chemosensitization and radiosensitization of human cancer by antisense anti-MDM2 oligonucleotides: in vitro and in vivo activities and mechanisms. Ann N Y Acad Sci 2004; 1002:217-35. [PMID: 14751837 DOI: 10.1196/annals.1281.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
MDM2 oncogene is overexpressed in many human cancers including breast, colon, and prostate cancer, and MDM2 levels are associated with poor prognosis in patients with cancer. Here, we summarize the investigation of the functions of MDM2 oncogene in human cancer growth and the value of MDM2 as a drug target for prostate cancer therapy by using antisense to inhibit MDM2 expression. Antisense anti-human-MDM2 oligonucleotides and mismatch controls were tested in in vitro and in vivo human cancer models for antitumor activity. Targeted gene products and related proteins were analyzed and the antitumor activity was determined when the oligonucleotides were used alone or in combination with cancer chemotherapeutics and radiation therapy. The antisense oligonucleotide specifically inhibited MDM2 expression in a dose- and time-dependent manner, resulting in significant antitumor activity in vitro and in vivo. The antisense oligonucleotides also potentiated the effects of p53 activation and p21 induction by chemotherapeutic agents 10-hydroxycamptothecin, adriamycin, 5-fluorouracil, and paclitaxel. In a dose-dependent manner, the antisense oligonucleotide showed antitumor activity in nude mice bearing human cancer xenografts and increased therapeutic effectiveness of the chemotherapeutic agents irinotecan, paclitaxel, and Rituxan and radiation therapy. These results indicate that MDM2 has a role in various tumor growth through both p53-dependent and p53-independent mechanisms, indicating that MDM2 inhibitors have a broad spectrum of antitumor activities in human cancers regardless of p53 status. These results provide a basis for clinical evaluation of antisense anti-MDM2 oligonucleotides as chemosensitizer and radiosensitizer.
Collapse
Affiliation(s)
- Hui Wang
- Departments of Pharmacology and Toxicology, Division of Clinical Pharmacology, Comprehensive Cancer Center, and Gene Therapy Center, University of Alabama at Birmingham, Alabama 35294-0019, USA
| | | | | | | | | |
Collapse
|
41
|
Zhu L, Tu Y, Huang C, Liu H, Feng A. Expression of MDM2 protein and mRNA in condyloma acuminata. Curr Med Sci 2004; 24:493-6. [PMID: 15641702 DOI: 10.1007/bf02831118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2003] [Indexed: 11/24/2022]
Abstract
In order to investigate the role of murine double minute 2 (MDM2) in cell proliferation and carcinogenesis in condyloma acuminatum (CA), immunohistochemistry and in situ hybridization were used to detect the expression of MDM2 protein and mRNA in normal skin and skin lesions of CA of vulva. PCR was also used to detect HPV types. The results showed that in 32 observed CA specimens, the expression of MDM2 protein and mRNA was detected in 18 (56.25%) and 22 (68.75%) respectively, while the co-expression of MDM2 protein and mRNA was found in 14. PCR results revealed that HPV6/11 and HPV16/18 subtypes were shown in 28(87.5%) and 4 (12.5%) respectively out of 32 CA specimens. Out of the 18 positive specimens expressing MDM2 protein, HPV6/11 subtypes were shown in 15 and HPV16/18 subtypes in 3. In 22 positive specimens expressing MDM2 mRNA, HPV6/11 subtypes were shown in 18 and HPV16/18 subtypes in 4. No expression of MDM2 protein and MDM2 mRNA was observed in normal skin. Our study indicated that the overexpression of MDM2 might be involved in malignant proliferation and carcinogenesis of CA.
Collapse
Affiliation(s)
- Li Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | | | | | | | | |
Collapse
|
42
|
Catley L, Weisberg E, Tai YT, Atadja P, Remiszewski S, Hideshima T, Mitsiades N, Shringarpure R, LeBlanc R, Chauhan D, Munshi NC, Schlossman R, Richardson P, Griffin J, Anderson KC. NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood 2003; 102:2615-22. [PMID: 12816865 DOI: 10.1182/blood-2003-01-0233] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are emerging as a promising new treatment strategy in hematologic malignancies. Here we show that NVP-LAQ824, a novel hydroxamic acid derivative, induces apoptosis at physiologically achievable concentrations (median inhibitory concentration [IC50] of 100 nM at 24 hours) in multiple myeloma (MM) cell lines resistant to conventional therapies. MM.1S myeloma cell proliferation was also inhibited when cocultured with bone marrow stromal cells, demonstrating ability to overcome the stimulatory effects of the bone marrow microenvironment. Importantly, NVP-LAQ824 also inhibited patient MM cell growth in a dose- and time-dependent manner. NVP-LAQ824-induced apoptotic signaling includes up-regulation of p21, caspase cascade activation, and poly (adenosine diphosphate [ADP]) ribose (PARP) cleavage. Apoptosis was confirmed with cell cycle analysis and annexin-propidium iodide staining. Interestingly, treatment of MM cells with NVPLAQ824 also led to proteasome inhibition, as determined by reduced proteasome chymotrypsin-like activity and increased levels of cellular polyubiquitin conjugates. Finally, a study using NVP-LAQ824 in a preclinical murine myeloma model provides in vivo relevance to our in vitro studies. Taken together, these findings provide the framework for NVP-LAQ824 as a novel therapeutic in MM.
Collapse
Affiliation(s)
- Laurence Catley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pruneri G, Carboni N, Baldini L, Intini D, Colombi M, Bertolini F, Valentini S, Maisonneuve P, Viale G, Neri A. Cell cycle regulators in multiple myeloma: prognostic implications of p53 nuclear accumulation. Hum Pathol 2003; 34:41-7. [PMID: 12605365 DOI: 10.1053/hupa.2003.6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Multiple myeloma (MM) is characterized by a multistep process of tumorigenesis involving genes that control cell cycle progression. The prevalence and clinical implications of p53, p21, HDM-2, p27, and cyclin E immunoreactivity in MM patients, however, have not been fully elucidated. We evaluated the immunoreactivity (IR) for p53, p21, HDM-2, p27, cyclin E, and Ki-67 in bone marrow biopsies from 48 patients. In 34 (70.8%) cases, TP53 gene mutations and HDM-2 gene amplification were analyzed by polymerase chain reaction-single-strand conformation polymorphism (PCR-SSCP) and Southern blot densitometric analyses in the corresponding bone marrow aspirates. Nineteen (39.6%) biopsy specimens exhibited > or =10% neoplastic cells immunoreactive for p53, 23 (47.9%) for p21, 28 (58.3%) for HDM-2, 29 (60.4%) for cyclin E, and 16 (33.3%) for Ki-67; 23 (47.9%) tumors had > or =50% neoplastic cells immunoreactive for p27. TP53 gene mutations in exons 5 through 8 were detected in 3 (8.8%) cases, whereas none exhibited HDM-2 gene amplification. In the cases bearing a wild-type TP53 gene, no association was found between p53 accumulation and HDM-2 or p21 IR. The same cases had been previously investigated for the presence of the t(11;14) translocation and cyclin D1 IR; interestingly, a significant inverse correlation between cyclin D1 and p27 or cyclin E IR was noted. In addition to clinical stage and Bartl's histologic stage and grade, p53 accumulation was significantly associated with survival, and it maintained its prognostic significance in a multivariate analysis adjusted for age, clinical stage, and relapse. Our data suggest that the immunohistochemical evaluation of p53 IR in bone marrow biopsies may represent an adjunct in MM patient prognostication.
Collapse
Affiliation(s)
- Giancarlo Pruneri
- Division of Pathology and Laboratory Medicine, European Institute of Oncology and University of Milan, School of Medicine, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhao X, Demary K, Wong L, Vaziri C, McKenzie AB, Eberlein TJ, Spanjaard RA. Retinoic acid receptor-independent mechanism of apoptosis of melanoma cells by the retinoid CD437 (AHPN). Cell Death Differ 2001; 8:878-86. [PMID: 11526443 DOI: 10.1038/sj.cdd.4400894] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2001] [Revised: 03/20/2001] [Accepted: 04/09/2001] [Indexed: 11/09/2022] Open
Abstract
Retinoic acid (RA) induces differentiation of S91 melanoma cells through activation of RA receptor (RAR)gamma without affecting cell viability. The novel RARgamma-agonist CD437 (AHPN), however, also induces concomitant apoptosis through an unknown mechanism which was investigated here. By utilizing DNA microarray analysis, five apoptosis-associated, CD437-induced transcripts (CITs) were identified. Interestingly, all CITs are also regulated by p53 in a DNA damage response, and consistent with this interpretation, CD437 was found to cause DNA adduct-formation. However, p53 is not required for CD437-dependent regulation of CITs. Among this set of genes, induction of p21(WAF1/CIP1) is likely to be responsible for early S-phase growth-arrest of CD437-treated cells, whereas ei24 is a critical mediator of CD437-induced apoptosis in S91 cells. These data suggest an RAR-independent mechanism in which CD437 causes DNA adduct-formation, resulting in induction of a p53-independent DNA damage response, and subsequent growth-arrest and apoptosis. CD437-mediated DNA adduct-formation may also explain its apoptotic effects in other cell types.
Collapse
Affiliation(s)
- X Zhao
- Department of Otolaryngology, Cancer Research Center, Boston University School of Medicine, 75 Albany Street R903, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Vincent T, Jourdan M, Sy MS, Klein B, Mechti N. Hyaluronic acid induces survival and proliferation of human myeloma cells through an interleukin-6-mediated pathway involving the phosphorylation of retinoblastoma protein. J Biol Chem 2001; 276:14728-36. [PMID: 11278272 DOI: 10.1074/jbc.m003965200] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Originating from a post-switch memory B cell or plasma cell compartment in peripheral lymphoid tissues, malignant myeloma cells accumulate in the bone marrow of patients with multiple myeloma. In this favorable microenvironment their growth and survival are dependent upon both soluble factors and physical cell-to-cell and cell-to-extracellular matrix contacts. In this report we show that hyaluronan (HA), a major nonprotein glycosaminoglycan component of the extracellular matrix in mammalian bone marrow, is a survival and proliferation factor for human myeloma cells. The effect of HA is mainly mediated through a gp 80-interleukin 6 (IL-6) receptor pathway by a CD44-independent mechanism, suggesting that HA retains and concentrates IL-6 close to its site of secretion, thus favoring its autocrine activity. In addition, we show that HA-mediated survival and proliferation of myeloma cells is associated with a down-regulation in the expression of p27(kip1) cyclin-dependent kinase inhibitor and a hyperphosphorylation of the retinoblastoma protein (pRb). These data suggest that HA could be an important component in the myeloma cell physiopathology in vivo by potentiating autocrine and/or paracrine IL-6 activities.
Collapse
Affiliation(s)
- T Vincent
- INSERM Unité U475, 99 rue Puech Villa, 34197 Montpellier cedex 5, France
| | | | | | | | | |
Collapse
|
46
|
Capoulade C, Mir LM, Carlier K, Lécluse Y, Tétaud C, Mishal Z, Wiels J. Apoptosis of tumoral and nontumoral lymphoid cells is induced by both mdm2 and p53 antisense oligodeoxynucleotides. Blood 2001; 97:1043-9. [PMID: 11159535 DOI: 10.1182/blood.v97.4.1043] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Following stress signals, the p53 tumor suppressor protein plays a critical role in regulation of cell proliferation, mainly through induction of growth arrest or apoptosis. Therefore, this protein needs to be strictly regulated and numerous studies have shown that the MDM2 protein is an essential element for p53 regulation in normal cells and, most importantly, that overexpression of MDM2 is responsible for p53 inactivation in various types of tumors. A previous study showed that this is the case in some Burkitt lymphoma (BL) cell lines, where enhanced translation of mdm2 messenger RNA results in overexpression of the protein that complexes and inactivates wild-type p53. To further investigate the role of the p53/MDM2 complex in these BL cells, as well as in other lymphoid cells that do not overexpress MDM2, this study used antisense oligodeoxynucleotides directed either against mdm2 or against p53. Results show that the mdm2 antisense oligodeoxynucleotide induces apoptosis of cells that express a high or low level of MDM2 protein, only if they contain wild-type p53. Moreover, apoptosis is independent of the accumulation of p53 following mdm2 antisense treatment. Finally, the p53 antisense oligodeoxynucleotide, which inhibits the expression of wild-type p53, also induces a decrease of the MDM2 level in cells, whether or not they overexpress this protein, and causes apoptosis of these cells. These results indicate that decreasing the MDM2 protein level by directly or indirectly targeting its biosynthesis is a potent tool for the induction of apoptosis.
Collapse
Affiliation(s)
- C Capoulade
- CNRS UMR 1598, Interactions moléculaires et cancer, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
47
|
Trinh E, Boutillier AL, Loeffler JP. Regulation of the retinoblastoma-dependent Mdm2 and E2F-1 signaling pathways during neuronal apoptosis. Mol Cell Neurosci 2001; 17:342-53. [PMID: 11178871 DOI: 10.1006/mcne.2000.0928] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously demonstrated that the apoptotic signaling pathway in K(+)-deprived cerebellar granule neurons involves a caspase-dependent cleavage of the retinoblastoma protein (Rb). Here, we have further investigated the functional consequences of this cleavage on two Rb-binding partners: the oncoprotein Mdm2 and the transcription factor E2F-1. A K(+) deprivation time course leads to a caspase inhibitor-sensitive degradation of Mdm2. Experimental blockade of Mdm2 expression with antisense oligodeoxynucleotides (ODN) results in neuronal death, suggesting an active role of Mdm2 in neuroprotection. By contrast, the E2F-1 protein accumulates in a caspase-independent manner following K(+) withdrawal, a consequence of increased gene transcription. This is likely to result from the rapid cyclin-dependent kinase 4 activation observed in LK, that correlates with a transient Rb phosphorylation. Counteracting E2F-1 upregulation with antisense ODNs prevents neuronal loss. Taken together, these data demonstrate that Rb is a central player in regulating both caspase-dependent and -independent events leading to apoptosis.
Collapse
Affiliation(s)
- E Trinh
- UMR 7519 CNRS-21, rue René Descartes, Strasbourg Cedex, 67 084, France
| | | | | |
Collapse
|
48
|
Abstract
This review discusses laboratory and clinical studies of antisense oligodeoxynucleotides as potential treatments for haematological malignancies and solid tumours. Mechanisms of action, pharmacokinetics, toxicities and potential clinical applications of these agents are described.
Collapse
Affiliation(s)
- W Pawlak
- Department of Oncology, Central Clinical Hospital of Military Medical Academy, Warsaw, Poland
| | | | | | | |
Collapse
|
49
|
Kastrinakis NG, Gorgoulis VG, Foukas PG, Dimopoulos MA, Kittas C. Molecular aspects of multiple myeloma. Ann Oncol 2000; 11:1217-28. [PMID: 11106109 DOI: 10.1023/a:1008331714186] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell neoplasm characterized by bone marrow infiltration with malignant plasma cells, which synthesize and secrete monoclonal immunoglobulin (Ig) fragments. Despite the considerable progress in the understanding of MM biology, the molecular basis of the disease remains elusive. The initial transformation is thought to occur in a postgerminal center B-lineage cell, carrying a somatically hypermutated Ig heavy chain (IGH) gene. This plasmablastic precursor cell colonizes the bone marrow, propagates clonally and differentiates into a slowly proliferating myeloma cell population, all under the influence of specific cell adhesion molecules and cytokines. Production of interleukin-6 by stromal cells, osteoblasts and, in some cases, neoplastic cells is an essential element of myeloma cell growth, with the cytokine stimulus being delivered intracellularly via the Jack-STAT and ras signaling pathways. While karyotypic changes have been identified in up to 50% of MM patients, recent molecular cytogenetic techniques have revealed chromosomal abnormalities in the vast majority of examined cases. Translocations mostly involve illegal switch rearrangements of the IGH locus with various partner genes (CCND1, FGFR3, c-maf). Such events have been assigned a critical role in MM development. Mutations in coding and regulatory regions, as well as aberrant expression patterns of several oncogenes (c-myc, ras) and tumor suppressor genes (p16, p15) have been reported. Key regulators of programmed cell death (BCL-2, Fas), tumor expansion (metalloproteinases) and drug responsiveness (topoisomerase II alpha) have also been implicated in the pathogenesis of this hematologic malignancy. A tumorigenic role for human herpesvirus 8 (HHV8) was postulated recently, following the detection of viral sequences in bone marrow dendritic cells of MM patients. However, since several research groups were unable to confirm this observation, the role of HHV8 remains unclear. Translation of the advances in MM molecular biology into novel therapeutic strategies is essential in order to improve disease prognosis.
Collapse
Affiliation(s)
- N G Kastrinakis
- Department of Histology, School of Medicine, University of Athens, Greece
| | | | | | | | | |
Collapse
|
50
|
Flores ER, Allen-Hoffmann BL, Lee D, Lambert PF. The human papillomavirus type 16 E7 oncogene is required for the productive stage of the viral life cycle. J Virol 2000; 74:6622-31. [PMID: 10864676 PMCID: PMC112172 DOI: 10.1128/jvi.74.14.6622-6631.2000] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The production of the human papillomavirus type 16 (HPV-16) is intimately tied to the differentiation of the host epithelium that it infects. Infection occurs in the basal layer of the epithelium at a site of wounding, where the virus utilizes the host DNA replication machinery to establish itself as a low-copy-number episome. The productive stage of the HPV-16 life cycle occurs in the postmitotic suprabasal layers of the epithelium, where the virus amplifies its DNA to high copy number, synthesizes the capsid proteins (L1 and L2), encapsidates the HPV-16 genome, and releases virion particles as the upper layer of the epithelium is shed. Papillomaviruses are hypothesized to possess a mechanism to overcome the block in DNA synthesis that occurs in the differentiated epithelial cells, and the HPV-16 E7 oncoprotein has been suggested to play a role in this process. To determine whether E7 plays a role in the HPV-16 life cycle, an E7-deficient HPV-16 genome was created by inserting a translational termination linker (TTL) in the E7 gene of the full HPV-16 genome. This DNA was transfected into an immortalized human foreskin keratinocyte cell line shown previously to support the HPV-16 life cycle, and stable cell lines were obtained that harbored the E7-deficient HPV-16 genome episomally, the state of the genome found in normal infections. By culturing these cells under conditions which promote the differentiation of epithelial cells, we found E7 to be necessary for the productive stage of the HPV-16 life cycle. HPV-16 lacking E7 failed to amplify its DNA and expressed reduced amounts of the capsid protein L1, which is required for virus production. E7 appears to create a favorable environment for HPV-16 DNA synthesis by perturbing the keratinocyte differentiation program and inducing the host DNA replication machinery. These data demonstrate that E7 plays an essential role in the papillomavirus life cycle.
Collapse
Affiliation(s)
- E R Flores
- McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|