101
|
Structure-Activity Relationships (SARs) of α-Ketothioamides as Inhibitors of Phosphoglycerate Dehydrogenase (PHGDH). Pharmaceuticals (Basel) 2020; 13:ph13020020. [PMID: 31979167 PMCID: PMC7168936 DOI: 10.3390/ph13020020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/04/2020] [Accepted: 01/14/2020] [Indexed: 12/11/2022] Open
Abstract
For many years now, targeting deregulation within cancer cells’ metabolism has appeared as a promising strategy for the development of more specific and efficient cancer treatments. Recently, numerous reports highlighted the crucial role of the serine synthetic pathway, and particularly of the phosphoglycerate dehydrogenase (PHGDH), the first enzyme of the pathway, to sustain cancer progression. Yet, because of very weak potencies usually in cell-based settings, the inhibitors reported so far failed to lay ground on the potential of this approach. In this paper, we report a structure–activity relationship study of a series of α-ketothioamides that we have recently identified. Interestingly, this study led to a deeper understanding of the structure–activity relationship (SAR) in this series and to the identification of new PHGDH inhibitors. The activity of the more potent compounds was confirmed by cellular thermal shift assays and in cell-based experiments. We hope that this research will eventually provide a new entry point, based on this promising chemical scaffold, for the development of therapeutic agents targeting PHGDH.
Collapse
|
102
|
Xie H, Gu Y, Wang W, Wang X, Ye X, Xin C, Lu M, Reddy BA, Shu P. Silencing of SENP2 in Multiple Myeloma Induces Bortezomib Resistance by Activating NF-κB Through the Modulation of IκBα Sumoylation. Sci Rep 2020; 10:766. [PMID: 31964975 PMCID: PMC6972929 DOI: 10.1038/s41598-020-57698-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/20/2019] [Indexed: 01/09/2023] Open
Abstract
The proteasome inhibitor bortezomib is the most successfully applied chemotherapeutic drug for treating multiple myeloma. However, its clinical efficacy reduced due to resistance development. The underlying molecular mechanisms of bortezomib resistance are poorly understood. In this study, by combining in silico analysis and sgRNA library based drug resistance screening assay, we identified SENP2 (Sentrin/SUMO-specific proteases-2) as a bortezomib sensitive gene and found its expression highly downregulated in bortezomib resistant multiple myeloma patient's samples. Furthermore, down regulation of SENP2 in multiple myeloma cell line RPMI8226 alleviated bortezomib induced cell proliferation inhibition and apoptosis, whereas, overexpression of SENP2 sensitized these cells to bortezomib treatment. We further demonstrate that knockdown of SENP2 in RPMI8226 cells increased SUMO2 conjugated IκBα that resulted in the activation of NF-κB. Taken together, we report that silencing of SENP2 and consequent activation of NF-κB through the modulation of IκBα sumoylation as a novel mechanism inducing bortezomib resistance in multiple myeloma.
Collapse
Affiliation(s)
- Hongyi Xie
- Clinical Laboratory, the People's Hospital of Beilun District, Beilun Branch Hospital of the First Affiliated Hospital of Medical School Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - Yuanliang Gu
- Department of prevention and health care, the People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - Wenjuan Wang
- Department of Hematology & Oncology, the People's Hospital of Beilun District, Beilun Branch Hospital of the First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - Xuyao Wang
- Molecular Laboratory, the People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - Xiaojuan Ye
- Department of Hematology & Oncology, the People's Hospital of Beilun District, Beilun Branch Hospital of the First Affiliated Hospital of Medical School of Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - Chao Xin
- Clinical Laboratory, the People's Hospital of Beilun District, Beilun Branch Hospital of the First Affiliated Hospital of Medical School Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - Mengjiao Lu
- Clinical Laboratory, the People's Hospital of Beilun District, Beilun Branch Hospital of the First Affiliated Hospital of Medical School Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China
| | - B Ashok Reddy
- Division of Oncology, Liveon Biolabs, 46 & 47, Water tank Road, KIADB-Phase II, Antharasanahalli, Tumakuru, Karnataka, PIN-572106, India.
| | - Peng Shu
- Molecular Laboratory, the People's Hospital of Beilun District, Beilun Branch Hospital of The First Affiliated Hospital of Medical School Zhejiang University, 1288 Lushan East Road, Beilun District, Ningbo, 315800, China.
| |
Collapse
|
103
|
Saavedra-García P, Martini F, Auner HW. Proteasome inhibition in multiple myeloma: lessons for other cancers. Am J Physiol Cell Physiol 2019; 318:C451-C462. [PMID: 31875696 DOI: 10.1152/ajpcell.00286.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cellular protein homeostasis (proteostasis) depends on the controlled degradation of proteins that are damaged or no longer required by the ubiquitin-proteasome system (UPS). The 26S proteasome is the principal executer of substrate-specific proteolysis in eukaryotic cells and regulates a myriad of cellular functions. Proteasome inhibitors were initially developed as chemical tools to study proteasomal function but rapidly became widely used anticancer drugs that are now used at all stages of treatment for the bone marrow cancer multiple myeloma (MM). Here, we review the mechanisms of action of proteasome inhibitors that underlie their preferential toxicity to MM cells, focusing on endoplasmic reticulum stress, depletion of amino acids, and effects on glucose and lipid metabolism. We also discuss mechanisms of resistance to proteasome inhibition such as autophagy and metabolic rewiring and what lessons we may learn from the success and failure of proteasome inhibition in MM for treating other cancers with proteostasis-targeting drugs.
Collapse
Affiliation(s)
- Paula Saavedra-García
- Cancer Cell Metabolism Group, Hugh and Josseline Langmuir Centre for Myeloma Research, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Francesca Martini
- Department of Translational Research on New Technologies in Medicine and Surgery, Hematology Unit, Ospedale Santa Chiara, Pisa, Italy
| | - Holger W Auner
- Cancer Cell Metabolism Group, Hugh and Josseline Langmuir Centre for Myeloma Research, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
104
|
Mofers A, Perego P, Selvaraju K, Gatti L, Gullbo J, Linder S, D'Arcy P. Analysis of determinants for in vitro resistance to the small molecule deubiquitinase inhibitor b-AP15. PLoS One 2019; 14:e0223807. [PMID: 31639138 PMCID: PMC6804958 DOI: 10.1371/journal.pone.0223807] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/27/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND b-AP15/VLX1570 are small molecule inhibitors of the ubiquitin specific peptidase 14 (USP14) and ubiquitin carboxyl-terminal hydrolase 5 (UCHL5) deubiquitinases (DUBs) of the 19S proteasome. b-AP15/VLX1570 have been shown to be cytotoxic to cells resistant to bortezomib, raising the possibility that this class of drugs can be used as a second-line therapy for treatment-resistant multiple myeloma. Limited information is available with regard to potential resistance mechanisms to b-AP15/VLX1570. RESULTS We found that b-AP15-induced cell death is cell-cycle dependent and that non-cycling tumor cells may evade b-AP15-induced cell death. Such non-cycling cells may re-enter the proliferative state to form colonies of drug-sensitive cells. Long-term selection of cells with b-AP15 resulted in limited drug resistance (~2-fold) that could be reversed by buthionine sulphoximine, implying altered glutathione (GSH) metabolism as a resistance mechanism. In contrast, drug uptake and overexpression of drug efflux transporters were found not to be associated with b-AP15 resistance. CONCLUSIONS The proteasome DUB inhibitors b-AP15/VLX1570 are cell cycle-active. The slow and incomplete development of resistance towards these compounds is an attractive feature in view of future clinical use.
Collapse
Affiliation(s)
- Arjan Mofers
- Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - Paola Perego
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Karthik Selvaraju
- Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - Laura Gatti
- Cerebrovascular Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Joachim Gullbo
- Department of Radiology, Oncology and Radiation Science, Section of Oncology, Uppsala University, Uppsala, Sweden
| | - Stig Linder
- Department of Medicine and Health, Linköping University, Linköping, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Padraig D'Arcy
- Department of Medicine and Health, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
105
|
Wei L, Lee D, Law CT, Zhang MS, Shen J, Chin DWC, Zhang A, Tsang FHC, Wong CLS, Ng IOL, Wong CCL, Wong CM. Genome-wide CRISPR/Cas9 library screening identified PHGDH as a critical driver for Sorafenib resistance in HCC. Nat Commun 2019; 10:4681. [PMID: 31615983 PMCID: PMC6794322 DOI: 10.1038/s41467-019-12606-7] [Citation(s) in RCA: 261] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
Sorafenib is the standard treatment for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance is common. By using genome-wide CRISPR/Cas9 library screening, we identify phosphoglycerate dehydrogenase (PHGDH), the first committed enzyme in the serine synthesis pathway (SSP), as a critical driver for Sorafenib resistance. Sorafenib treatment activates SSP by inducing PHGDH expression. With RNAi knockdown and CRISPR/Cas9 knockout models, we show that inactivation of PHGDH paralyzes the SSP and reduce the production of αKG, serine, and NADPH. Concomitantly, inactivation of PHGDH elevates ROS level and induces HCC apoptosis upon Sorafenib treatment. More strikingly, treatment of PHGDH inhibitor NCT-503 works synergistically with Sorafenib to abolish HCC growth in vivo. Similar findings are also obtained in other FDA-approved tyrosine kinase inhibitors (TKIs), including Regorafenib or Lenvatinib. In summary, our results demonstrate that targeting PHGDH is an effective approach to overcome TKI drug resistance in HCC. Resistance to the tyrosine kinase inhibitor Sorafenib, which is the standard treatment for advanced hepatocellular carcinoma, is a major clinical challenge. Here, the authors show that phosphoglycerate dehydrogenase, a key enzyme in the serine synthesis pathway, drives sorafenib resistance.
Collapse
Affiliation(s)
- Lai Wei
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Derek Lee
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cheuk-Ting Law
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Misty Shuo Zhang
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Jialing Shen
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Don Wai-Ching Chin
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Allen Zhang
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Felice Ho-Ching Tsang
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Ceci Lok-Sze Wong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Irene Oi-Lin Ng
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Carmen Chak-Lui Wong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong. .,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Chun-Ming Wong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong. .,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
106
|
Maekawa K, Ri M, Nakajima M, Sekine A, Ueda R, Tohkin M, Miyata N, Saito Y, Iida S. Serum lipidomics for exploring biomarkers of bortezomib therapy in patients with multiple myeloma. Cancer Sci 2019; 110:3267-3274. [PMID: 31444836 PMCID: PMC6778623 DOI: 10.1111/cas.14178] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/06/2019] [Accepted: 08/15/2019] [Indexed: 01/08/2023] Open
Abstract
Although the proteasome inhibitor bortezomib (BTZ) shows excellent efficacy in multiple myeloma (MM), a fraction of patients has a suboptimal or no response to this agent. In addition, BTZ-induced peripheral neuropathy (BiPN), a frequent side-effect of this therapy, limits its use in some patients. This study aimed to explore serum lipid biomarker candidates to predict the response to BTZ and the severity of BiPN. Fifty-nine serum samples were collected from patients with MM prior to receiving BTZ plus low-dose dexamethasone therapy. Serum levels of phospholipids, sphingolipids, neutral lipids, and polyunsaturated fatty acids and their oxidation products were measured by a comprehensive lipidomic study. Overall, 385 lipid metabolites were identified in patients' sera; lower levels of several glycerophospholipids, sphingolipids, and cholesteryl esters were associated with a poor treatment response. Metabolites related to platelet-activating factor biosynthesis and cholesterol metabolism appeared particularly relevant. Furthermore, several lysophosphatidylcholines, phosphatidylcholines, ceramides, neutral lipids, and oxidative fatty acids were significantly increased or decreased in patients with BiPN grades ranging from G0 to G3. Among these compounds, mediators reportedly inducing myelin breakdown and stimulating inflammatory responses were prominent. Although further study is necessary to validate these biomarker candidates, our results contribute to the development of predictive biomarkers for response to BTZ treatment, or ensuing severe BiPN, in patients with MM.
Collapse
Affiliation(s)
- Keiko Maekawa
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyoto, Japan.,Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Division of Blood Transfusion, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan.,WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Akihiro Sekine
- Center for Preventive Medical Science, Chiba University, Chiba, Japan
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Masahiro Tohkin
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Naoki Miyata
- Institute of Drug Discovery Science, Nagoya City University, Nagoya, Japan
| | - Yoshiro Saito
- Division of Medical Safety Science, National Institute of Health Sciences, Kawasaki, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
107
|
Nishimura N, Radwan MO, Amano M, Endo S, Fujii E, Hayashi H, Ueno S, Ueno N, Tatetsu H, Hata H, Okamoto Y, Otsuka M, Mitsuya H, Matsuoka M, Okuno Y. Novel p97/VCP inhibitor induces endoplasmic reticulum stress and apoptosis in both bortezomib-sensitive and -resistant multiple myeloma cells. Cancer Sci 2019; 110:3275-3287. [PMID: 31368616 PMCID: PMC6778635 DOI: 10.1111/cas.14154] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 07/10/2019] [Accepted: 07/23/2019] [Indexed: 12/30/2022] Open
Abstract
p97/VCP is an endoplasmic reticulum (ER)‐associated protein that belongs to the AAA (ATPases associated with diverse cellular activities) ATPase family. It has a variety of cellular functions including ER‐associated protein degradation, autophagy, and aggresome formation. Recent studies have shown emerging roles of p97/VCP and its potential as a therapeutic target in several cancer subtypes including multiple myeloma (MM). We conducted a cell‐based compound screen to exploit novel small compounds that have cytotoxic activity in myeloma cells. Among approximately 2000 compounds, OSSL_325096 showed relatively strong antiproliferative activity in MM cell lines (IC50, 100‐500 nmol/L). OSSL_325096 induced apoptosis in myeloma cell lines, including a bortezomib‐resistant cell line and primary myeloma cells purified from patients. Accumulation of poly‐ubiquitinated proteins, PERK, CHOP, and IREα, was observed in MM cell lines treated with OSSL_325096, suggesting that it induces ER stress in MM cells. OSSL_325096 has a similar chemical structure to DBeQ, a known p97/VCP inhibitor. Knockdown of the gene encoding p97/VCP induced apoptosis in myeloma cells, accompanied by accumulation of poly‐ubiquitinated protein. IC50 of OSSL_325096 to myeloma cell lines were found to be lower (0.1‐0.8 μmol/L) than those of DBeQ (2‐5 μmol/L). In silico protein–drug‐binding simulation suggested possible binding of OSSL_325096 to the ATP binding site in the D2 domain of p97/VCP. In cell‐free ATPase assays, OSSL_325096 showed dose‐dependent inhibition of p97/VCP ATPase activity. Finally, OSSL_325096 inhibited the growth of subcutaneous myeloma cell tumors in vivo. The present data suggest that OSSL_325096 exerts anti‐myeloma activity, at least in part through p97/VCP inhibition.
Collapse
Affiliation(s)
- Nao Nishimura
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Mohamed O Radwan
- Faculty of Life Sciences, Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan.,Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Cairo, Egypt
| | - Masayuki Amano
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Shinya Endo
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Eri Fujii
- Faculty of Medical Sciences, Division of Informative Clinical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hironori Hayashi
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Shikiko Ueno
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Niina Ueno
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Hiro Tatetsu
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Hiroyuki Hata
- Faculty of Medical Sciences, Division of Informative Clinical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshinari Okamoto
- Faculty of Life Sciences, Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Masami Otsuka
- Faculty of Life Sciences, Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Hiroaki Mitsuya
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Masao Matsuoka
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Yutaka Okuno
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| |
Collapse
|
108
|
Choi J, Busino L. E3 ubiquitin ligases in B-cell malignancies. Cell Immunol 2019; 340:103905. [PMID: 30827673 PMCID: PMC6584052 DOI: 10.1016/j.cellimm.2019.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/05/2018] [Accepted: 02/19/2019] [Indexed: 12/21/2022]
Abstract
Ubiquitylation is a post-translational modification (PTM) that controls various cellular signaling pathways. It is orchestrated by a three-step enzymatic cascade know as the ubiquitin proteasome system (UPS). E3 ligases dictate the specificity to the substrates, primarily leading to proteasome-dependent degradation. Deregulation of the UPS components by various mechanisms contributes to the pathogenesis of cancer. This review focuses on E3 ligase-substrates pairings that are implicated in B-cell malignancies. Understanding the molecular mechanism of specific E3 ubiquitin ligases will present potential opportunities for the development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Jaewoo Choi
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Luca Busino
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
109
|
Tsvetkov P, Detappe A, Cai K, Keys HR, Brune Z, Ying W, Thiru P, Reidy M, Kugener G, Rossen J, Kocak M, Kory N, Tsherniak A, Santagata S, Whitesell L, Ghobrial IM, Markley JL, Lindquist S, Golub TR. Mitochondrial metabolism promotes adaptation to proteotoxic stress. Nat Chem Biol 2019; 15:681-689. [PMID: 31133756 PMCID: PMC8183600 DOI: 10.1038/s41589-019-0291-9] [Citation(s) in RCA: 365] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2022]
Abstract
The mechanisms by which cells adapt to proteotoxic stress are largely unknown, but key to understanding how tumor cells, particularly in vivo, are largely resistant to proteasome inhibitors. Analysis of cancer cell lines, mouse xenografts and patient-derived tumor samples all showed an association between mitochondrial metabolism and proteasome inhibitor sensitivity. When cells were forced to use oxidative phosphorylation rather than glycolysis, they became proteasome inhibitor-resistant. This mitochondrial state, however, creates a unique vulnerability: sensitivity to the small-molecule compound elesclomol. Genome-wide CRISPR/Cas9 screening showed that a single gene, encoding the mitochondrial reductase FDX1, could rescue elesclomol-induced cell death. Enzymatic function and NMR-based analyses further showed that FDX1 is the direct target of elesclomol, which promotes a unique form of copper-dependent cell death. These studies elucidate a fundamental mechanism by which cells adapt to proteotoxic stress and suggests strategies to mitigate proteasome inhibitor-resistance.
Collapse
Affiliation(s)
| | - Alexandre Detappe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kai Cai
- Biochemistry Department, University of Wisconsin-Madison, Madison, WI, USA
| | - Heather R Keys
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Zarina Brune
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Weiwen Ying
- OnTarget Pharmaceutical Consulting LLC, Lexington, MA, USA
| | - Prathapan Thiru
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Mairead Reidy
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Jordan Rossen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mustafa Kocak
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Nora Kory
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Luke Whitesell
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Molecular Genetics Department, University of Toronto, Toronto, ON, Canada
| | - Irene M Ghobrial
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - John L Markley
- Biochemistry Department, University of Wisconsin-Madison, Madison, WI, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Todd R Golub
- Broad Institute of Harvard and MIT, Cambridge, MA, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD, USA. .,Harvard Medical School, Boston, MA, USA. .,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
110
|
Janker L, Mayer RL, Bileck A, Kreutz D, Mader JC, Utpatel K, Heudobler D, Agis H, Gerner C, Slany A. Metabolic, Anti-apoptotic and Immune Evasion Strategies of Primary Human Myeloma Cells Indicate Adaptations to Hypoxia. Mol Cell Proteomics 2019; 18:936-953. [PMID: 30792264 PMCID: PMC6495257 DOI: 10.1074/mcp.ra119.001390] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Indexed: 12/26/2022] Open
Abstract
Multiple Myeloma (MM) is an incurable plasma cell malignancy primarily localized within the bone marrow (BM). It develops from a premalignant stage, monoclonal gammopathy of undetermined significance (MGUS), often via an intermediate stage, smoldering MM (SMM). The mechanisms of MM progression have not yet been fully understood, all the more because patients with MGUS and SMM already carry similar initial mutations as found in MM cells. Over the last years, increased importance has been attributed to the tumor microenvironment and its role in the pathophysiology of the disease. Adaptations of MM cells to hypoxic conditions in the BM have been shown to contribute significantly to MM progression, independently from the genetic predispositions of the tumor cells. Searching for consequences of hypoxia-induced adaptations in primary human MM cells, CD138-positive plasma cells freshly isolated from BM of patients with different disease stages, comprising MGUS, SMM, and MM, were analyzed by proteome profiling, which resulted in the identification of 6218 proteins. Results have been made fully accessible via ProteomeXchange with identifier PXD010600. Data previously obtained from normal primary B cells were included for comparative purposes. A principle component analysis revealed three clusters, differentiating B cells as well as MM cells corresponding to less and more advanced disease stages. Comparing these three clusters pointed to the alteration of pathways indicating adaptations to hypoxic stress in MM cells on disease progression. Protein regulations indicating immune evasion strategies of MM cells were determined, supported by immunohistochemical staining, as well as transcription factors involved in MM development and progression. Protein regulatory networks related to metabolic adaptations of the cells became apparent. Results were strengthened by targeted analyses of a selected panel of metabolites in MM cells and MM-associated fibroblasts. Based on our data, new opportunities may arise for developing therapeutic strategies targeting myeloma disease progression.
Collapse
Affiliation(s)
- Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Rupert L Mayer
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dominique Kreutz
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Johanna C Mader
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Kirsten Utpatel
- Department of Pathology, University Regensburg, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Hermine Agis
- Department of Oncology, University Clinic for Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria;.
| |
Collapse
|
111
|
Zhang J, Ye ZW, Townsend DM, Hughes-Halbert C, Tew KD. Racial disparities, cancer and response to oxidative stress. Adv Cancer Res 2019; 144:343-383. [PMID: 31349903 PMCID: PMC7104807 DOI: 10.1016/bs.acr.2019.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
At the intersection of genetics, biochemistry and behavioral sciences, there is a largely untapped opportunity to consider how ethnic and racial disparities contribute to individual sensitivity to reactive oxygen species and how these might influence susceptibility to various cancers and/or response to classical cancer treatment regimens that pervasively result in the formation of such chemical species. This chapter begins to explore these connections and builds a platform from which to consider how the disciplines can be strengthened further.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States.
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Chanita Hughes-Halbert
- Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
112
|
Akhtar S, Achkar IW, Siveen KS, Kuttikrishnan S, Prabhu KS, Khan AQ, Ahmed EI, Sahir F, Jerobin J, Raza A, Merhi M, Elsabah HM, Taha R, Omri HE, Zayed H, Dermime S, Steinhoff M, Uddin S. Sanguinarine Induces Apoptosis Pathway in Multiple Myeloma Cell Lines via Inhibition of the JaK2/STAT3 Signaling. Front Oncol 2019; 9:285. [PMID: 31058086 PMCID: PMC6478801 DOI: 10.3389/fonc.2019.00285] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/29/2019] [Indexed: 12/15/2022] Open
Abstract
Sanguinarine (SNG), a benzophenanthridine alkaloid, has displayed various anticancer abilities in several vivo and in vitro studies. However, the anticancer potential of SNG is yet to be established in multiple myeloma (MM), a mostly incurable malignancy of plasma cells. In this study, we aimed to investigate the potential anti-proliferative and pro-apoptotic activities of SNG in a panel of MM cell lines (U266, IM9, MM1S, and RPMI-8226). SNG treatment of MM cells resulted in a dose-dependent decrease in cell viability through mitochondrial membrane potential loss and activation of caspase 3, 9, and cleavage of PARP. Pre-treatment of MM cells with a universal caspase inhibitor, Z-VAD-FMK, prevented SNG mediated loss of cell viability, apoptosis, and caspase activation, confirming that SNG-mediated apoptosis is caspase-dependent. The SNG-mediated apoptosis appears to be resulted from suppression of the constitutively active STAT3 with a concomitant increase in expression of protein tyrosine phosphatase (SHP-1). SNG treatment of MM cells leads to down-regulation of the anti-apoptotic proteins including cyclin D, Bcl-2, Bclxl, and XIAP. In addition, it also upregulates pro-apoptotic protein, Bax. SNG mediated cellular DNA damage in MM cell lines by induction of oxidative stress through the generation of reactive oxygen species and depletion of glutathione. Finally, the subtoxic concentration of SNG enhanced the cytotoxic effects of anticancer drugs bortezomib (BTZ) by suppressing the viability of MM cells via induction of caspase-mediated apoptosis. Altogether our findings demonstrate that SNG induces mitochondrial and caspase-dependent apoptosis, generates oxidative stress, and suppresses MM cell lines proliferation. In addition, co-treatment of MM cell lines with sub-toxic doses of SNG and BTZ potentiated the cytotoxic activity. These results would suggest that SNG could be developed into therapeutic agent either alone or in combination with other anticancer drugs in MM.
Collapse
Affiliation(s)
- Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Iman W. Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kodappully S. Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Eiman I. Ahmed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Fairooz Sahir
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Jayakumar Jerobin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Hesham M. Elsabah
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Ruba Taha
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Halima El Omri
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology Venereology, Hamad Medical Corporation, Doha, Qatar
- Weill Cornell-Medicine, Doha, Qatar
- Weill Cornell-Medicine, Cornell University, New York, NY, United States
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
113
|
Rizzieri D, Paul B, Kang Y. Metabolic alterations and the potential for targeting metabolic pathways in the treatment of multiple myeloma. ACTA ACUST UNITED AC 2019; 5. [PMID: 31020046 PMCID: PMC6476731 DOI: 10.20517/2394-4722.2019.05] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Metabolism is defined as the collection of complex biochemical processes that living cells use to generate energy and maintain their growth and survival. Metabolism encompasses the synthesis and breakdown of glucose, fatty acids, and amino acids; the generation of energy (ATP); and oxidative phosphorylation. In cancer cells, metabolism can be commandeered to promote tumor growth and cellular proliferation. These alterations in metabolism have emerged as an additional hallmark of various cancers. In this review we focus on metabolic alterations in multiple myeloma (MM) - a malignancy of plasma cells - including derangements in glycolysis, gluconeogenesis, the tricarboxylic acid cycle, oxidative phosphorylation, and fatty acid/amino acid synthesis and degradation. Particular focus is given to metabolic alterations that contribute to myeloma cell growth, proliferation and drug resistance. Finally, novel approaches that target metabolic pathways for the treatment of MM are discussed.
Collapse
Affiliation(s)
- Dustin Rizzieri
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Barry Paul
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| | - Yubin Kang
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
114
|
The selective HDAC6 inhibitor Nexturastat A induces apoptosis, overcomes drug resistance and inhibits tumor growth in multiple myeloma. Biosci Rep 2019; 39:BSR20181916. [PMID: 30782785 PMCID: PMC6430725 DOI: 10.1042/bsr20181916] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/12/2019] [Accepted: 02/18/2019] [Indexed: 01/25/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that produce a monoclonal immunoglobulin protein. Despite significant advances in the treatment of MM, challenges such as resistance to therapy remain. Currently, inhibition of histone deacetylases (HDACs) is emerging as a potential method for treating cancers. Numerous HDAC inhibitors are being studied for the use in monotherapy or in conjunction with other agents for MM. In the present study, we investigated the anti-myeloma effect of Nexturastat A (NexA), a novel selective HDAC6 inhibitor. We found that NexA impaired MM cells viability in a dose- and time-dependent manner. NexA also provoked a cell cycle arrest at the G1 phase in MM cells. Furthermore, NexA promoted apoptosis of MM cells via transcriptional activation of the p21 promoter, which may through its ability to up-regulate the H3Ac and H4Ac levels. Additionally, NexA could overcome bortezomib (BTZ) resistance in MM cells, and NexA in combination with BTZ had stronger efficacy. We also confirmed that NexA inhibited tumor growth in murine xenograft models of MM. These interesting findings provided the rationale for the future advancement of this novel HDAC6 inhibitor as a potential therapeutic anti-myeloma agent.
Collapse
|
115
|
Besse L, Besse A, Mendez-Lopez M, Vasickova K, Sedlackova M, Vanhara P, Kraus M, Bader J, Ferreira RB, Castellano RK, Law BK, Driessen C. A metabolic switch in proteasome inhibitor-resistant multiple myeloma ensures higher mitochondrial metabolism, protein folding and sphingomyelin synthesis. Haematologica 2019; 104:e415-e419. [PMID: 30792209 DOI: 10.3324/haematol.2018.207704] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Lenka Besse
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Andrej Besse
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Max Mendez-Lopez
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Katerina Vasickova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Miroslava Sedlackova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petr Vanhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marianne Kraus
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Jürgen Bader
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| | - Renan B Ferreira
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Ronald K Castellano
- Department of Chemistry, University of Florida, Gainesville, FL, USA.,UF-Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Brian K Law
- UF-Health Cancer Center, University of Florida, Gainesville, FL, USA.,Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, USA
| | - Christoph Driessen
- Experimental Oncology and Hematology, Department of Oncology and Hematology, St. Gallen Cantonal Hospital, St. Gallen, Switzerland
| |
Collapse
|
116
|
Kim Y, Lee J, Lee D, Othmer HG. Synergistic Effects of Bortezomib-OV Therapy and Anti-Invasive Strategies in Glioblastoma: A Mathematical Model. Cancers (Basel) 2019; 11:E215. [PMID: 30781871 PMCID: PMC6406513 DOI: 10.3390/cancers11020215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
It is well-known that the tumor microenvironment (TME) plays an important role in the regulation of tumor growth and the efficacy of anti-tumor therapies. Recent studies have demonstrated the potential of combination therapies, using oncolytic viruses (OVs) in conjunction with proteosome inhibitors for the treatment of glioblastoma, but the role of the TME in such therapies has not been studied. In this paper, we develop a mathematical model for combination therapies based on the proteosome inhibitor bortezomib and the oncolytic herpes simplex virus (oHSV), with the goal of understanding their roles in bortezomib-induced endoplasmic reticulum (ER) stress, and how the balance between apoptosis and necroptosis is affected by the treatment protocol. We show that the TME plays a significant role in anti-tumor efficacy in OV combination therapy, and illustrate the effect of different spatial patterns of OV injection. The results illustrate a possible phenotypic switch within tumor populations in a given microenvironment, and suggest new anti-invasion therapies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Junho Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
117
|
Abstract
BACKGROUND Oral cancer is one of the most frequently occurring cancers. Metabolic reprogramming is an important hallmark of cancer. Metabolomics characterizes all the small molecules in a biological sample, and a complete set of small molecules in such sample is referred as metabolome. Nuclear magnetic resonance spectroscopy and mass spectrometry are two widely used techniques in metabolomics studies. Increasing evidence demonstrates that metabolomics techniques can be used to explore the metabolic signatures in oral cancer. Elucidation of metabolic alterations in oral cancer is also important for the understanding of its pathological mechanisms. AIM OF REVIEW In this paper, we summarize the latest progress of metabolomics study in oral cancer and provide the suggestions for the future studies. KEY SCIENTIFIC CONCEPTS OF REVIEW The metabolomics studies in saliva, serum, and tumor tissues revealed the existence of metabolic signatures in bio-fluids and tissues of oral cancer, and several tumor-specific metabolites identified in individual study could discriminate oral cancer from healthy controls or precancerous lesions, which are potential biomarkers for the screening or early diagnosis of oral cancer. Metabolomics study of oral cancers in the future should aim to establish a routine procedure with high sensitivity, profile intracellular metabolites to find out the metabolic characteristics of tumor cells, and investigate the mechanism behind metabolomic alterations and the metabolic response of cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Xun Chen
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Dongsheng Yu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, 510055, People's Republic of China.
| |
Collapse
|
118
|
Harding T, Baughn L, Kumar S, Van Ness B. The future of myeloma precision medicine: integrating the compendium of known drug resistance mechanisms with emerging tumor profiling technologies. Leukemia 2019; 33:863-883. [PMID: 30683909 DOI: 10.1038/s41375-018-0362-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/25/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy that is considered mostly incurable in large part due to the inability of standard of care therapies to overcome refractory disease and inevitable drug-resistant relapse. The post-genomic era has been a productive period of discovery where modern sequencing methods have been applied to large MM patient cohorts to modernize our current perception of myeloma pathobiology and establish an appreciation for the vast heterogeneity that exists between and within MM patients. Numerous pre-clinical studies conducted in the last two decades have unveiled a compendium of mechanisms by which malignant plasma cells can escape standard therapies, many of which have potentially quantifiable biomarkers. Exhaustive pre-clinical efforts have evaluated countless putative anti-MM therapeutic agents and many of these have begun to enter clinical trial evaluation. While the palette of available anti-MM therapies is continuing to expand it is also clear that malignant plasma cells still have mechanistic avenues by which they can evade even the most promising new therapies. It is therefore becoming increasingly clear that there is an outstanding need to develop and employ precision medicine strategies in MM management that harness emerging tumor profiling technologies to identify biomarkers that predict efficacy or resistance within an individual's sub-clonally heterogeneous tumor. In this review we present an updated overview of broad classes of therapeutic resistance mechanisms and describe selected examples of putative biomarkers. We also outline several emerging tumor profiling technologies that have the potential to accurately quantify biomarkers for therapeutic sensitivity and resistance at genomic, transcriptomic and proteomic levels. Finally, we comment on the future of implementation for precision medicine strategies in MM and the clear need for a paradigm shift in clinical trial design and disease management.
Collapse
Affiliation(s)
- Taylor Harding
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, USA
| | - Linda Baughn
- Department of Laboratory Medicine and Pathology, Division of Laboratory Genetics, Mayo Clinic, Rochester, MN, USA
| | - Shaji Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic Rochester, Rochester, USA
| | - Brian Van Ness
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
119
|
Zhang Y, Yang L, Dai G, Cao H. Knockdown of PHGDH potentiates 5-FU cytotoxicity in gastric cancer cells via the Bcl-2/Bax/caspase-3 signaling pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:5869-5876. [PMID: 31949673 PMCID: PMC6963086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/15/2018] [Indexed: 06/10/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies in the world. Fluorouracil (5-FU) is widely used in the treatment of cancers, but resistance to 5-FU results in the failure of chemotherapy. Phosphoglycerate dehydrogenase (PHGDH) has been reported to play a vital role in the development of 5-FU resistance in cancer cells. However, the exact role of PHGDH and the underlying mechanisms for 5-FU resistance in GC cells remain elusive. In this study, PHGDH expression was much higher in the GC tissues of 5-FU-resistant patients than that in the GC tissues of 5-FU-sensitive patients. Moreover, the expression of PHGDH was obviously increased in BGC823/5-FU cells compared with that in BGC823 cells. 5-FU treatment significantly reduced the viability of BGC823/5-FU cells, in a dose- and time-dependent manner. Furthermore, 5-FU treatment inhibited the proliferation of BGC823/5-FU cells, as evidenced by decreased cell viability and reduced colony-forming ability. The knockdown of PHGDH made possible the inhibitory effect of 5-FU on the proliferation of BGC823/5-FU cells. Furthermore, 5-FU treatment promoted apoptosis of BGC823/5-FU cells, as indicated by increased numbers of TUNEL-positive cells and increased rates of apoptosis. Notably, the promoting effect of 5-FU on the apoptosis of BGC823/5-FU cells was markedly enhanced by PHGDH knockdown. Additionally, 5-FU treatment downregulated Bcl-2 expression and upregulated the expression of Bax and caspase-3, and this effect was remarkably enhanced by PHGDH knockdown. In conclusion, knockdown of PHGDH potentiates 5-FU cytotoxicity in GC cells via the Bcl-2/Bax/caspase-3 signaling pathway.
Collapse
Affiliation(s)
- Yunli Zhang
- Department of Abdominal Surgery, Zhejiang Cancer Hospital Hangzhou 310022, Zhejiang Province, China
| | - Litao Yang
- Department of Abdominal Surgery, Zhejiang Cancer Hospital Hangzhou 310022, Zhejiang Province, China
| | - Guangou Dai
- Department of Abdominal Surgery, Zhejiang Cancer Hospital Hangzhou 310022, Zhejiang Province, China
| | - Hu Cao
- Department of Abdominal Surgery, Zhejiang Cancer Hospital Hangzhou 310022, Zhejiang Province, China
| |
Collapse
|
120
|
Guerrero-Garcia TA, Gandolfi S, Laubach JP, Hideshima T, Chauhan D, Mitsiades C, Anderson KC, Richardson PG. The power of proteasome inhibition in multiple myeloma. Expert Rev Proteomics 2018; 15:1033-1052. [PMID: 30427223 DOI: 10.1080/14789450.2018.1543595] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Proteasome inhibitors (PIs) are therapeutic backbones of multiple myeloma treatment, with PI-based therapies being standards of care throughout the treatment algorithm. Proteasome inhibition affects multiple critical signaling pathways in myeloma cells and interacts synergistically with mechanisms of action of other conventional and novel agents, resulting in substantial anti-myeloma activity and at least additive effects. Areas covered: This review summarizes the biologic effects of proteasome inhibition in myeloma and provides an overview of the importance of proteasome inhibition to the current treatment algorithm. It reviews key clinical data on three PIs, specifically bortezomib, carfilzomib, and ixazomib; assesses ongoing phase 3 trials with these agents; and looks ahead to the increasingly broad role of both approved PIs and PIs under investigation in the frontline and relapsed settings. Expert commentary: Progress to date with PIs in multiple myeloma has been impressive, but there remain unmet needs and challenges, as well as increasing opportunities to optimize the use of these agents. Understanding discrepancies between PIs in terms of efficacy and safety profile is a key goal of ongoing research, along with proteomics-based efforts to identify potential biomarkers of sensitivity and resistance, thereby enabling increasingly personalized treatment approaches in the future.
Collapse
Affiliation(s)
| | - Sara Gandolfi
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Jacob P Laubach
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Teru Hideshima
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | | | | | | | - Paul G Richardson
- b Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| |
Collapse
|
121
|
Zaal EA, Berkers CR. The Influence of Metabolism on Drug Response in Cancer. Front Oncol 2018; 8:500. [PMID: 30456204 PMCID: PMC6230982 DOI: 10.3389/fonc.2018.00500] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/15/2018] [Indexed: 12/23/2022] Open
Abstract
Resistance to therapeutic agents, either intrinsic or acquired, is currently a major problem in the treatment of cancers and occurs in virtually every type of anti-cancer therapy. Therefore, understanding how resistance can be prevented, targeted and predicted becomes increasingly important to improve cancer therapy. In the last decade, it has become apparent that alterations in cellular metabolism are a hallmark of cancer cells and that a rewired metabolism is essential for rapid tumor growth and proliferation. Recently, metabolic alterations have been shown to play a role in the sensitivity of cancer cells to widely-used first-line chemotherapeutics. This suggests that metabolic pathways are important mediators of resistance toward anticancer agents. In this review, we highlight the metabolic alterations associated with resistance toward different anticancer agents and discuss how metabolism may be exploited to overcome drug resistance to classical chemotherapy.
Collapse
Affiliation(s)
- Esther A. Zaal
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Celia R. Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
122
|
Zhang L, Xu Y, Wang L, Liu H. Role of RACK1 on cell proliferation, adhesion, and bortezomib-induced apoptosis in multiple myeloma. Int J Biol Macromol 2018; 121:1077-1085. [PMID: 30315883 DOI: 10.1016/j.ijbiomac.2018.10.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/16/2018] [Accepted: 10/09/2018] [Indexed: 12/30/2022]
Abstract
Receptor for activated C kinase 1 (RACK1), a scaffold protein, plays a crucial role in the progression of various cancers. However, the biological function and underlying mechanism of RACK1 in multiple myeloma (MM) cells remain unclear. The present study aimed to explore the function of RACK1 on the cell proliferation, adhesion, and bortezomib-induced apoptosis in MM. We found that RACK1 was significantly overexpressed in myeloma cell lines and primary myeloma cells compared with normal bone marrow plasma cells. Moreover, immunofluorescence revealed that RACK1 was primarily expressed in the cytoplasm of MM cells. Knockdown of RACK1 impaired growth of MM cells, blocked entry into the S-phase of the cell cycle, and resulted in reduced cell adhesion rates. More importantly, knockdown of RACK1 decreased the proliferation of MM cells by activating P-P38 and P-ERK in the MAPK/ERK signaling pathway. We also found that altered expression of RACK1 is associated with bortezomib-mediated MM cell apoptosis. In summary, these results may provide a possible target for therapy in MM.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Ya Xu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Li Wang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, People's Republic of China.
| |
Collapse
|
123
|
Abu el Maaty MA, Dabiri Y, Almouhanna F, Blagojevic B, Theobald J, Büttner M, Wölfl S. Activation of pro-survival metabolic networks by 1,25(OH) 2D 3 does not hamper the sensitivity of breast cancer cells to chemotherapeutics. Cancer Metab 2018; 6:11. [PMID: 30181873 PMCID: PMC6116450 DOI: 10.1186/s40170-018-0183-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/01/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We have previously identified 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], the bioactive form of vitamin D3, as a potent regulator of energy-utilization and nutrient-sensing pathways in prostate cancer cells. In the current study, we investigated the effects of 1,25(OH)2D3 on breast cancer (BCa) cell metabolism using cell lines representing distinct molecular subtypes, luminal (MCF-7 and T-47D), and triple-negative BCa (MDA-MB-231, MDA-MB-468, and HCC-1143). METHODS 1,25(OH)2D3's effect on BCa cell metabolism was evaluated by employing a combination of real-time measurements of glycolysis/oxygen consumption rates using a biosensor chip system, GC/MS-based metabolomics, gene expression analysis, and assessment of overall energy levels. The influence of treatment on energy-related signaling molecules was investigated by immunoblotting. RESULTS We show that 1,25(OH)2D3 significantly induces the expression and activity of the pentose phosphate pathway enzyme glucose-6-phosphate dehydrogenase (G6PD) in all BCa cell lines, however differentially influences glycolytic and respiratory rates in the same cells. Although 1,25(OH)2D3 treatment was found to induce seemingly anti-oxidant responses in MCF-7 cells, such as increased intracellular serine levels, and reduce the expression of its putative target gene thioredoxin-interacting protein (TXNIP), intracellular reactive oxygen species levels were found to be elevated. Serine accumulation in 1,25(OH)2D3-treated cells was not found to hamper the efficacy of chemotherapeutics, including 5-fluorouracil. Detailed analyses of the nature of TXNIP's regulation by 1,25(OH)2D3 included genetic and pharmacological inhibition of signaling molecules and metabolic enzymes including AMP-activated protein kinase and G6PD, as well as by studying the ITCH (E3 ubiquitin ligase)-TXNIP interaction. While these investigations demonstrated minimal involvement of such pathways in the observed non-canonical regulation of TXNIP, inhibition of estrogen receptor (ER) signaling by tamoxifen mirrored the reduction of TXNIP levels by 1,25(OH)2D3, demonstrating that the latter's negative regulation of ER expression is a potential mechanism of TXNIP modulation. CONCLUSIONS Altogether, we propose that regulation of energy metabolism contributes to 1,25(OH)2D3's anti-cancer effects and that combining 1,25(OH)2D3 with drugs targeting metabolic networks in tumor cells may lead to synergistic effects.
Collapse
Affiliation(s)
- Mohamed A. Abu el Maaty
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Yasamin Dabiri
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Fadi Almouhanna
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Biljana Blagojevic
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Jannick Theobald
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Michael Büttner
- Metabolomics Core Technology Platform, Center for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 360, 69120 Heidelberg, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| |
Collapse
|
124
|
Steiner N, Müller U, Hajek R, Sevcikova S, Borjan B, Jöhrer K, Göbel G, Pircher A, Gunsilius E. The metabolomic plasma profile of myeloma patients is considerably different from healthy subjects and reveals potential new therapeutic targets. PLoS One 2018; 13:e0202045. [PMID: 30096165 PMCID: PMC6086450 DOI: 10.1371/journal.pone.0202045] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/26/2018] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION Multiple myeloma (MM), a malignant plasma cell disorder, is still an incurable disease. Thus, the identification of novel therapeutic targets is of utmost importance. Here, we evaluated the peripheral blood-based metabolic profile of patients with MM. MATERIAL & METHODS Peripheral blood plasma levels of 188 endogenous metabolites, including amino acids, biogenic amines, acylcarnitines, glycerophospholipids, sphingomyelins, and hexoses were determined in patients with plasma cell dyscrasias: monoclonal gammopathy of undetermined significance, a precursor stage of MM (MGUS, n = 15), newly diagnosed MM, (NDMM, n = 32), relapsed/refractory MM (RRMM, n = 19) and in 25 healthy controls by mass spectrometry. RESULTS Patients with NDMM, RRMM and MGUS have a substantially different metabolomic profile than healthy controls. The amount of eight plasma metabolites significantly differs between the NDMM and MGUS group: free carnitine, acetylcarnitine, glutamate, asymmetric dimethylarginine (ADMA) and four phosphatidylcholine (PC) species. In addition, the levels of octadecanoylcarnitine, ADMA and six PCs were significantly different between RRMM and MGUS patients. 13 different concentrations of metabolites were found between RRMM and NDMM patients (free carnitine, acetylcarnitine, creatinine, five LysoPCs and PCs). Pathway analyses revealed a distinct metabolic profile with significant alterations in amino acid, lipid, and energy metabolism in healthy volunteers compared to MGUS/MM patients. CONCLUSION We identified different metabolic profiles in MGUS und MM patients in comparison to healthy controls. Thus, different metabolic processes, potentially the immunoregulation by indoleamine 2,3 dioxygenase-1 (IDO), which is involved in cancer development and progression supporting inflammatory processes in the tumor microenvironment and glutaminolysis, can serve as novel promising therapeutic targets in MM.
Collapse
Affiliation(s)
- Normann Steiner
- Laboratory for Tumor Biology & Angiogenesis, Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Innsbruck, Austria
- * E-mail:
| | - Udo Müller
- Biocrates Life Sciences AG, Innsbruck, Austria
| | - Roman Hajek
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Sabina Sevcikova
- Babak Myeloma Group, Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Bojana Borjan
- Laboratory for Tumor Biology & Angiogenesis, Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Innsbruck, Austria
| | - Karin Jöhrer
- Tyrolean Cancer Research Institute, Innsbruck, Austria
- Salzburg Cancer Research Institute, Laboratory for Immunological and Molecular Cancer Research, Salzburg, Austria
| | - Georg Göbel
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Pircher
- Laboratory for Tumor Biology & Angiogenesis, Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Innsbruck, Austria
| | - Eberhard Gunsilius
- Laboratory for Tumor Biology & Angiogenesis, Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
125
|
El Arfani C, De Veirman K, Maes K, De Bruyne E, Menu E. Metabolic Features of Multiple Myeloma. Int J Mol Sci 2018; 19:E1200. [PMID: 29662010 PMCID: PMC5979361 DOI: 10.3390/ijms19041200] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/07/2018] [Accepted: 04/10/2018] [Indexed: 01/19/2023] Open
Abstract
Cancer is known for its cellular changes contributing to tumour growth and cell proliferation. As part of these changes, metabolic rearrangements are identified in several cancers, including multiple myeloma (MM), which is a condition whereby malignant plasma cells accumulate in the bone marrow (BM). These metabolic changes consist of generation, inhibition and accumulation of metabolites and metabolic shifts in MM cells. Changes in the BM micro-environment could be the reason for such adjustments. Enhancement of glycolysis and glutaminolysis is found in MM cells compared to healthy cells. Metabolites and enzymes can be upregulated or downregulated and play a crucial role in drug resistance. Therefore, this review will focus on changes in glucose and glutamine metabolism linked with the emergence of drug resistance. Moreover, metabolites do not only affect other metabolic components to benefit cancer development; they also interfere with transcription factors involved in proliferation and apoptotic regulation.
Collapse
Affiliation(s)
- Chaima El Arfani
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| |
Collapse
|
126
|
Gandolfi S, Laubach JP, Hideshima T, Chauhan D, Anderson KC, Richardson PG. The proteasome and proteasome inhibitors in multiple myeloma. Cancer Metastasis Rev 2018; 36:561-584. [PMID: 29196868 DOI: 10.1007/s10555-017-9707-8] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteasome inhibitors are one of the most important classes of agents to have emerged for the treatment of multiple myeloma in the past two decades, and now form one of the backbones of treatment. Three agents in this class have been approved by the United States Food and Drug Administration-the first-in-class compound bortezomib, the second-generation agent carfilzomib, and the first oral proteasome inhibitor, ixazomib. The success of this class of agents is due to the exquisite sensitivity of myeloma cells to the inhibition of the 26S proteasome, which plays a critical role in the pathogenesis and proliferation of the disease. Proteasome inhibition results in multiple downstream effects, including the inhibition of NF-κB signaling, the accumulation of misfolded and unfolded proteins, resulting in endoplasmic reticulum stress and leading to the unfolded protein response, the downregulation of growth factor receptors, suppression of adhesion molecule expression, and inhibition of angiogenesis; resistance to proteasome inhibition may arise through cellular responses mediating these downstream effects. These multiple biologic consequences of proteasome inhibition result in synergistic or additive activity with other chemotherapeutic and targeted agents for myeloma, and proteasome inhibitor-based combination regimens have become established as a cornerstone of therapy throughout the myeloma treatment algorithm, incorporating agents from the other key classes of antimyeloma agents, including the immunomodulatory drugs, monoclonal antibodies, and histone deacetylase inhibitors. This review gives an overview of the critical role of the proteasome in myeloma and the characteristics of the different proteasome inhibitors and provides a comprehensive summary of key clinical efficacy and safety data with the currently approved proteasome inhibitors.
Collapse
Affiliation(s)
- Sara Gandolfi
- Dana-Farber Cancer Institute, 44 Binney Street, Dana 1B02, Boston, MA, 02115, USA
| | - Jacob P Laubach
- Dana-Farber Cancer Institute, 44 Binney Street, Dana 1B02, Boston, MA, 02115, USA
| | - Teru Hideshima
- Dana-Farber Cancer Institute, 44 Binney Street, Dana 1B02, Boston, MA, 02115, USA
| | - Dharminder Chauhan
- Dana-Farber Cancer Institute, 44 Binney Street, Dana 1B02, Boston, MA, 02115, USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute, 44 Binney Street, Dana 1B02, Boston, MA, 02115, USA
| | - Paul G Richardson
- Dana-Farber Cancer Institute, 44 Binney Street, Dana 1B02, Boston, MA, 02115, USA.
| |
Collapse
|
127
|
Yang X, Ye H, He M, Zhou X, Sun N, Guo W, Lin X, Huang H, Lin Y, Yao R, Wang H. LncRNA PDIA3P interacts with c-Myc to regulate cell proliferation via induction of pentose phosphate pathway in multiple myeloma. Biochem Biophys Res Commun 2018; 498:207-213. [PMID: 29501744 DOI: 10.1016/j.bbrc.2018.02.211] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 02/28/2018] [Indexed: 12/24/2022]
Abstract
Multiple myeloma (MM), the second most common hematologic malignancy, is an incurable disease characterized by the accumulation of malignant plasma cells within the bone marrow. Though great progresses have been made in understanding the mechanisms of MM, metabolic plasticity and drug resistance remain largely unknown. In this study, we found lncRNA Protein disulfide isomerase family A member 3 pseudogene 1 (PDIA3P) is highly expressed in MM and is associated with the survival rate of MM patients. PDIA3P regulates MM growth and drug resistance through Glucose 6-phosphate dehydrogenase (G6PD) and the pentose phosphate pathway (PPP). Mechanistically, we revealed that PDIA3P interacts with c-Myc to enhance its transactivation activity and binding to G6PD promoter, stimulating G6PD expression and PPP flux. Our study identified PDIA3P as a novel c-Myc interacting lncRNA and elucidated crucial roles for PDIA3P in metabolic regulation of MM, providing a potential therapeutic target for MM patients.
Collapse
Affiliation(s)
- Xiangchou Yang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Haihao Ye
- Department of Cardiology, Wenzhou TCM Hospital, Wenzhou, 325000, China
| | - Muqing He
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaohai Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ni Sun
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenjian Guo
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoji Lin
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - He Huang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ying Lin
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Rongxin Yao
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hong Wang
- Department of Rheumatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
128
|
Roeten MSF, Cloos J, Jansen G. Positioning of proteasome inhibitors in therapy of solid malignancies. Cancer Chemother Pharmacol 2018; 81:227-243. [PMID: 29184971 PMCID: PMC5778165 DOI: 10.1007/s00280-017-3489-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/19/2017] [Indexed: 12/13/2022]
Abstract
Targeting of the protein degradation pathway, in particular, the ubiquitin-proteasome system, has emerged as an attractive novel cancer chemotherapeutic modality. Although proteasome inhibitors have been most successfully applied in the treatment of hematological malignancies, they also received continuing interest for the treatment of solid tumors. In this review, we summarize the current positioning of proteasome inhibitors in the treatment of common solid malignancies (e.g., lung, colon, pancreas, breast, and head and neck cancer), addressing topics of their mechanism(s) of action, predictive factors and molecular mechanisms of resistance.
Collapse
Affiliation(s)
- Margot S F Roeten
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands.
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Location VUmc, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|