1
|
Ma Z, Tang M, Yang L, Chen L. Distribution, metabolism, and excretion of [ 14C] purinostat mesylate, a novel selective HDAC I/IIb inhibitor, in rats analyzed by high-performance liquid chromatography coupled with LTQ orbitrap mass spectrometry/radioactivity monitoring. J Pharm Biomed Anal 2025; 261:116834. [PMID: 40164058 DOI: 10.1016/j.jpba.2025.116834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Purinostat Mesylate (PM) is a novel and highly efficient selective histone deacetylase (HDAC) I/IIb inhibitor for hematologic tumor treatment that was granted Investigational New Drug (IND) approval for clinical investigation by the National Medical Products Administration and is currently in phase IIb clinical trials for relapsed/refractory diffuse large B-cell lymphoma. In this paper, the excretion, distribution, and metabolism properties of this IND were researched by High-Performance Liquid Chromatography coupled with LTQ Orbitrap Mass Spectrometry/Radioactivity Monitoring (HPLC-LTQ-Orbitrap-MS/RAM) and liquid scintillation counting. Following a single intravenous dose of [14C] PM to rats, a total of 98.49 % of the dose was recovered from intact rats within 0-168 h post-dose, with 14.16 % in urine and 83.15 % in feces, most of which was recovered within the first 24 h post-dose. For bile duct cannulated rats, a total of 95.54 % of the dose was recovered, with 62.37 % in bile, 23.37 % in urine and 8.58 % in feces within 0-72 h post-dose, suggesting that [14C] PM was excreted mainly into feces via biliary excretion. [14C] PM was distributed widely and eliminated rapidly throughout the body, with the lung, liver, kidney and intestine as the main organs. Interestingly, slow elimination was observed in the spleen, which could benefit the functional restoration of the spleen in hematological tumors. In terms of metabolism, [14C] PM underwent an extensive metabolic transformation in rats. Fourteen metabolites were tentatively identified, with major phase I metabolic pathways encompassing reduction, N-dealkylation, and oxidative deamination. Concomitantly, the primary phase II metabolic routes involved acetylation and glucuronic acid conjugation. This study was the first comprehensive PM pharmacokinetic study utilizing [14C] isotope labeling technology.
Collapse
Affiliation(s)
- Ziyan Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Chengdu Zenitar Biomedical Technology Co., Ltd, Chengdu 610212, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linyu Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Chengdu Zenitar Biomedical Technology Co., Ltd, Chengdu 610212, China.
| |
Collapse
|
2
|
Huang L, Zhang J, Punnoose E, Xiao Z, Li W. Current status of drug development for patients with multiple myeloma: a review of comparison in China and the rest of world. Antib Ther 2023; 6:127-136. [PMID: 37324548 PMCID: PMC10262841 DOI: 10.1093/abt/tbad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Multiple myeloma (MM) is a highly heterogeneous malignancy. The treatment of MM has been significantly advanced in recent years. B cell maturation antigen (BCMA)-targeted immunotherapy and chimeric antigen receptor T (CAR-T) cell therapy have been approved for the treatment of relapsed and refractory MM (RRMM), which will be launched in China shortly. The CD38 (cluster of differentiation 38) antibody, daratumumab, improves the clinical outcomes both RRMM and newly diagnosed MM patients. The combination of daratumumab, bortezomib and dexamethasone achieved favorable outcomes as the first-line therapy in China. However, high-risk patients have limited benefits from these advanced therapeutics, and usually relapse early, progressing into aggressive end-stage MM. Therefore, novel therapies are sought to improve the cancer prognosis in these patients. This review furnishes an overview of the recent clinical developments of these novel drugs and compares the drug candidates under development in China to the rest of the world.
Collapse
Affiliation(s)
- Lei Huang
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai 201203, China
| | - Jingyu Zhang
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai 201203, China
| | - Elizabeth Punnoose
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA 94080, USA
| | - Zhenyu Xiao
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai 201203, China
| | - Wenjin Li
- To whom correspondence should be addressed. Wenjin Li, OBD China Lab, Room 2.57, Building 5, Lane 371, Lishizhen Road, Pudong Shanghai, China. Tel: +86 21 2894 6650.
| |
Collapse
|
3
|
Huang PA, Beedie SL, Chau CH, Venzon DJ, Gere S, Kazandjian D, Korde N, Mailankody S, Landgren O, Figg WD. Cereblon gene variants and clinical outcome in multiple myeloma patients treated with lenalidomide. Sci Rep 2019; 9:14884. [PMID: 31619706 PMCID: PMC6795854 DOI: 10.1038/s41598-019-51446-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
Carfilzomib-lenalidomide-dexamethasone (KRd) therapy has yielded promising results in patients with newly diagnosed multiple myeloma (NDMM). Cereblon (CRBN) is the direct molecular target of lenalidomide and genetic polymorphisms in CRBN have been associated with lenalidomide efficacy. In this study, we assessed the correlation of five single nucleotide variants (SNVs) in the CRBN gene with clinical response and outcomes in patients with NDMM administered KRd therapy with lenalidomide maintenance, achieving favorable trial endpoints in a prospective Phase II study (NCT01402284). Of the observed SNVs, no associations with KRd therapy response were found in this patient cohort, although strong trends in hypoalbuminemia grade and hyperbilirubinemia grade emerged across the CRBN rs1672753 genotype (P = 0.0008) and the rs1714327 genotype (P = 0.0010), respectively. Our results do not provide conclusive support for the predictive utility of CRBN gene polymorphisms as potential biomarkers of clinical response to lenalidomide-based therapy in our patient population. However, these findings remain to be validated in prospective studies using larger patient populations.
Collapse
Affiliation(s)
- Phoebe A Huang
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shaunna L Beedie
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Cindy H Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David J Venzon
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, MD, USA
| | - Sheryl Gere
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Dickran Kazandjian
- Myeloma Program, Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
4
|
Mouse Monoclonal Antibodies Generated from Full Length Human Cereblon: Detection of Cereblon Protein in Patients with Multiple Myeloma. Int J Mol Sci 2017; 18:ijms18091999. [PMID: 28926977 PMCID: PMC5618648 DOI: 10.3390/ijms18091999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 01/18/2023] Open
Abstract
Immunomodulatory drugs (IMiDs) are profoundly active compounds in the treatment of patients with multiple myeloma (MM). However, despite the fact that treatment with IMiDs has dramatically improved survival for patients with MM, the majority of MM patients develop IMiDs resistance over time. We have found that expression of functional cereblon is required for IMiDs' action. In addition, it has been reported that cells expressing high levels of cereblon are resistant to proteasome inhibitor, implying that patients with high levels of cereblon should be resistant to proteasome inhibitor. If the above conclusions are correct, cereblon could be considered as a biomarker to determine which standard regimens should be used to treat patients with MM. Unfortunately, the conclusions mentioned above have not been clinically confirmed. In order to confirm these conclusions, we have generated three highly specific mouse monoclonal antibodies (mAbs) against full-length human cereblon. These mAbs can be used to do western blot, immunoprecipitation and immunohistochemistry staining. In addition, their epitopes have been precisely determined and the peptides covering their epitopes completely blocked the antibody binding to cereblon in western blot analysis or in immunohistochemistry staining of MM patients' specimens.
Collapse
|
5
|
Cosenza M, Civallero M, Marcheselli L, Sacchi S, Pozzi S. Ricolinostat, a selective HDAC6 inhibitor, shows anti-lymphoma cell activity alone and in combination with bendamustine. Apoptosis 2017; 22:827-840. [PMID: 28315173 PMCID: PMC5401712 DOI: 10.1007/s10495-017-1364-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Histone deacetylase inhibitors (HDACis) have emerged as a new class of anticancer agents, targeting the biological process including cell cycle and apoptosis. We investigated and explained the anticancer effects of an HDAC6 inhibitor, ricolinostat alone and in combination with bendamustine in lymphoma cell lines. Cell viability was measured by MTT assay. Apoptosis, reactive oxygen species (ROS) generation, Bcl-2 protein expression, cell cycle progression and tubuline expression were determined by flow cytometry. The effects of ricolinostat alone and in combination on the caspases, PI3K/Akt, Bcl-2 pathways, ER stress and UPR were assessed by immunoblotting. Ricolinostat shows anti lymphoma activity when used as single agent and its capability to induce apoptosis is synergistically potentiated by the bendamustine in lymphoma cell lines. Drug combination reduced the proportion of cells in the G0/G1 and S phases and caused an increase of "sub-G0/G1" peak. The synergistic effect accompanied with the increased ROS, activation of caspase-8, -9, and -3, the cleavage of PARP and modulated by Bcl-2 proteins family. In addition, the exposure of ricolinostat induced the acetylation level of α-tubulin, the extend of which was not further modified by bendamustine. Finally, the apoptosis effect of ricolinostat/bendamustine may be mediated by a corresponding effect on microtubule stabilization. Our data suggest that ricolinostat in combination with bendamustine may be a novel combination with potential for use as an antitumor agent in lymphoma.
Collapse
Affiliation(s)
- Maria Cosenza
- Program of Innovative Therapies in Oncology and Haematology, Department of Diagnostic Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71, 41124, Modena, Italy
| | - Monica Civallero
- Program of Innovative Therapies in Oncology and Haematology, Department of Diagnostic Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71, 41124, Modena, Italy
| | - Luigi Marcheselli
- Program of Innovative Therapies in Oncology and Haematology, Department of Diagnostic Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71, 41124, Modena, Italy
| | - Stefano Sacchi
- Program of Innovative Therapies in Oncology and Haematology, Department of Diagnostic Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71, 41124, Modena, Italy
| | - Samantha Pozzi
- Program of Innovative Therapies in Oncology and Haematology, Department of Diagnostic Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Via del Pozzo, 71, 41124, Modena, Italy.
| |
Collapse
|
6
|
Gomathinayagam S, Laface D, Houston-Cummings NR, Mangadu R, Moore R, Shandil I, Sharkey N, Li H, Stadheim TA, Zha D. In vivo anti-tumor efficacy of afucosylated anti-CS1 monoclonal antibody produced in glycoengineered Pichia pastoris. J Biotechnol 2015; 208:13-21. [PMID: 26015261 DOI: 10.1016/j.jbiotec.2015.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/21/2015] [Accepted: 05/13/2015] [Indexed: 11/15/2022]
Abstract
Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.
Collapse
Affiliation(s)
- Sujatha Gomathinayagam
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Drake Laface
- Biologics Discovery, Palo Alto, Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, United States
| | - Nga Rewa Houston-Cummings
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Ruban Mangadu
- Biologics Discovery, Palo Alto, Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, United States
| | - Renee Moore
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Ishaan Shandil
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Nathan Sharkey
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Huijuan Li
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Terrance A Stadheim
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Dongxing Zha
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States.
| |
Collapse
|
7
|
Chang X, Zhu Y, Shi C, Stewart AK. Mechanism of immunomodulatory drugs' action in the treatment of multiple myeloma. Acta Biochim Biophys Sin (Shanghai) 2014; 46:240-53. [PMID: 24374776 DOI: 10.1093/abbs/gmt142] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although immunomodulatory drugs (IMiDs), such as thalidomide, lenalidomide, and pomalidomide, are widely used in the treatment of multiple myeloma (MM), the molecular mechanism of IMiDs' action is largely unknown. In this review, we will summarize recent advances in the application of IMiDs in MM cancer treatment as well as their effects on immunomodulatory activities, anti-angiogenic activities, intervention of cell surface adhesion molecules between myeloma cells and bone marrow stromal cells, anti-inflammatory activities, anti-proliferation, pro-apoptotic effects, cell cycle arrest, and inhibition of cell migration and metastasis. In addition, the potential IMiDs' target protein, IMiDs' target protein's functional role, and the potential molecular mechanisms of IMiDs resistance will be discussed. We wish, by presentation of our naive discussion, that this review article will facilitate further investigation in these fields.
Collapse
Affiliation(s)
- Xiubao Chang
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | | | | | | |
Collapse
|
8
|
de la Rubia J, Roig M. Bortezomib for previously untreated multiple myeloma. Expert Rev Hematol 2014; 4:381-98. [DOI: 10.1586/ehm.11.38] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Simmons JK, Patel J, Michalowski A, Zhang S, Wei BR, Sullivan P, Gamache B, Felsenstein K, Kuehl WM, Simpson RM, Zingone A, Landgren O, Mock BA. TORC1 and class I HDAC inhibitors synergize to suppress mature B cell neoplasms. Mol Oncol 2013; 8:261-72. [PMID: 24429254 DOI: 10.1016/j.molonc.2013.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/20/2013] [Accepted: 11/21/2013] [Indexed: 12/12/2022] Open
Abstract
Enhanced proliferative signaling and loss of cell cycle regulation are essential for cancer progression. Increased mitogenic signaling through activation of the mTOR pathway, coupled with deregulation of the Cyclin D/retinoblastoma (Rb) pathway is a common feature of lymphoid malignancies, including plasmacytoma (PCT), multiple myeloma (MM), Burkitt's lymphoma (BL), and mantle cell lymphoma (MCL). Here we evaluate the synergy of pharmacologically affecting both of these critical pathways using the mTOR inhibitor sirolimus and the histone deacetylase inhibitor entinostat. A dose-matrix screening approach found this combination to be highly active and synergistic in a panel of genetically diverse human MM cell lines. Synergy and activity was observed in mouse PCT and human BL and MCL cell lines tested in vitro, as well as in freshly isolated primary MM patient samples tested ex vivo. This combination had minimal effects on healthy donor cells and retained activity when tested in a co-culture system simulating the protective interaction of cancer cells with the tumor microenvironment. Combining sirolimus with entinostat enhanced cell cycle arrest and apoptosis. At the molecular level, entinostat increased the expression of cell cycle negative regulators including CDKN1A (p21) and CDKN2A (p16), while the combination decreased critical growth and survival effectors including Cyclin D, BCL-XL, BIRC5, and activated MAPK.
Collapse
Affiliation(s)
- John K Simmons
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Jyoti Patel
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Aleksandra Michalowski
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Patrick Sullivan
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Ben Gamache
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Kenneth Felsenstein
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - W Michael Kuehl
- Genetics Branch, National Cancer Institute, National Institutes of Health, USA
| | - R Mark Simpson
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA
| | - Adriana Zingone
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, USA
| | - Ola Landgren
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, USA.
| |
Collapse
|
10
|
A Phase I Study of Pomalidomide (CC-4047) in Combination with Cisplatin and Etoposide in Patients with Extensive-Stage Small-Cell Lung Cancer. J Thorac Oncol 2013; 8:423-8. [DOI: 10.1097/jto.0b013e318282707b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Cai B, Lyu H, Huang J, Wang S, Lee CK, Gao C, Liu B. Combination of bendamustine and entinostat synergistically inhibits proliferation of multiple myeloma cells via induction of apoptosis and DNA damage response. Cancer Lett 2013; 335:343-50. [PMID: 23459296 DOI: 10.1016/j.canlet.2013.02.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/19/2013] [Accepted: 02/22/2013] [Indexed: 01/06/2023]
Abstract
Bendamustine, a hybrid molecule of purine analog and alkylator, induces cell death by activation of apoptosis, DNA damage response, and mitotic catastrophe. Entinostat, a selective class I inhibitor of histone deacetylase (HDAC), exerts anti-tumor activity in various cancer types, including multiple myeloma (MM). We sought to determine the combinatorial effects of bendamustine and entinostat on MM cells. Cell growth assays showed that bendamustine or entinostat inhibited proliferation in a dose-dependent manner, and their combinations synergistically induced growth inhibition in all MM cells tested. An apoptotic-ELISA and western blot assays on PARP cleavage and caspase-8 and caspase-3 revealed that bendamustine in combination with entinostat exhibited a much more potent activity than either agent alone to promote the MM cells undergoing apoptosis in a dose-dependent manner. Flow cytometric analysis found that entinostat exhibited distinct effects on cell cycle progression in different lines and bendamustine mainly arrested the cells at S phase, whereas their combinations dramatically blocked the S cells entering G2/M phase. Furthermore, studies on DNA damage response indicated that phospho-histone H2A.X (P-H2A.X), a hall marker of DNA double strand break, along with phosphorylated CHK2 (P-CHK2) was significantly enhanced by the combinations of bendamustine and entinostat as compared to either agent alone. These molecular changes were correlated with the increases in mitotic catastrophe. Collectively, our data demonstrate that bendamustine in combination with entinostat exhibit potent anti-proliferative/anti-survival activity in MM cells via induction of apoptosis and DNA damage response. Regimens consisting of bendamustine and/or entinostat may represent novel therapeutic strategies against MM.
Collapse
Affiliation(s)
- Bo Cai
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Liu H, Tamashiro S, Baritaki S, Penichet M, Yu Y, Chen H, Berenson J, Bonavida B. TRAF6 activation in multiple myeloma: a potential therapeutic target. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:155-63. [PMID: 22440007 DOI: 10.1016/j.clml.2012.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 01/18/2012] [Accepted: 01/20/2012] [Indexed: 01/03/2023]
Abstract
Multiple myeloma (MM) is an incurable B-lymphocyte malignancy. New therapeutic options have become available during the past several years; however nearly all patients acquire resistance to currently available therapeutic agents. Mechanisms contributing to the pathogenesis and chemoresistance of MM include genetic abnormalities, chromosomal translocations, gene mutations, the interaction between MM cells and the bone marrow microenvironment, and defects in the apoptotic signaling pathways. Survival signaling pathways associated with the pathogenesis of MM and bone marrow stromal cells play crucial roles in promoting growth, survival, adhesion, immortalization, angiogenesis, and drug resistance. The receptor activator of nuclear factor-kappa B/receptor activator of nuclear factor-kappa B ligand/tumor necrosis factor receptor-associated factor (RANK/RANKL-TRAF6) signal pathway mediates osteolytic bone lesions through the activation of the NF-κB and Janus kinase/signal transducer and activator of transcription (JNK) pathways in osteoclast precursor cells and thus contributes to the main clinical manifestations of bone disease. TRAF6 has also been identified as a ligase for Akt ubiquitination and membrane recruitment and its phosphorylation on growth factor stimulation. The inhibition of TRAF6 by silencing RNA or by decoy peptides decreases MM tumor cell proliferation and increases apoptosis as well as bone resorption. Some proteasome inhibitors and benzoxadiazole derivatives showed inhibitory effects on the activity and function of TRAF6. Overall, we propose that TRAF6 may be considered as a potential therapeutic target for the treatment of MM.
Collapse
Affiliation(s)
- Hong Liu
- Department of Microbiology, Immunology, and Molecular Genetics, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Abe M. Guest editorial: understanding the pathogenesis and the evolving treatment paradigm for multiple myeloma in the era of novel agents. Int J Hematol 2011; 94:307-309. [PMID: 21993876 DOI: 10.1007/s12185-011-0950-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Revised: 09/26/2011] [Accepted: 09/26/2011] [Indexed: 01/14/2023]
|
15
|
Offidani M, Corvatta L, Morabito F, Gentile M, Musto P, Leoni P, Palumbo A. How to treat patients with relapsed/refractory multiple myeloma: evidence-based information and opinions. Expert Opin Investig Drugs 2011; 20:779-93. [PMID: 21470070 DOI: 10.1517/13543784.2011.575060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Relapsed/refractory multiple myeloma (rrMM) remains a difficult condition to treat despite the availability of new drugs. This review aims to provide evidence to guide physicians in the choice of salvage therapy in certain subgroups of patients. AREAS COVERED The review attempts to present evidence-based information and suggest possible approaches based on data on previous therapies, previous remission duration and toxicity of previous treatments, patient's co-morbidities and disease characteristics at relapse. Unfortunately, little evidence is available; there are no large and/or randomized trials, direct comparisons of drugs or combinations for rrMM patients to draw any definite conclusion. EXPERT OPINION Almost all the studies presented here suggest that depth of response is a key factor also for patients with rrMM. Identifying the best approach between combinations and sequential therapies remains controversial. Several studies favor the former approach in early relapse as it leads to a higher complete response rate, regardless of previous therapies. However, in both strategies, achieving maximal response should always remain a main goal. Consolidation/maintenance therapy is beneficial both in combination and sequential therapies also in rrMM. Second generation new drugs, such as pomalidomide, carfilzomib, bendamustine and HDAC inhibitors, will probably expand the rescue possibilities also in this setting.
Collapse
Affiliation(s)
- Massimo Offidani
- Clinica di Ematologia, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
Huang J, Ding T, Yang M, Liu H, Sun X, Jin J. Antitumor activity and drug interactions of proteasome inhibitor Bortezomib in human high-risk myelodysplastic syndrome cells. Int J Hematol 2011; 93:482-493. [PMID: 21451957 DOI: 10.1007/s12185-011-0821-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 02/21/2011] [Accepted: 03/16/2011] [Indexed: 01/09/2023]
Abstract
The purpose of this study was to investigate the antitumor effects and drug interactions of the proteasome inhibitor Bortezomib against high-risk myelodysplastic syndrome (MDS) cells in vitro and in vivo. The high-risk MDS-derived MUTZ-1 cell line and bone marrow mononuclear cells from primary high-risk MDS patients were used to examine antitumor activity and drug interactions for Bortezomib. Apoptotic proteins, including caspase and Bcl-2 family members, as well as the protein FLIP, were studied. Phosphoinositide 3-kinase (PI3K)/Akt and MAPK signaling pathways were also examined. The PI3K inhibitor LY294002 was used to examine the involvement of the PI3K/Akt signaling pathway in the induction of apoptosis. Cytarabine (AraC) and daunorubicin (DNR) were used to test for synergistic effects between Bortezomib and chemotherapeutic agents. SCID mice xenografted with MUTZ-1 cells were used for in vivo study. We found that Bortezomib could induce growth arrest and apoptosis in high-risk MDS cells in vitro and in vivo. The mechanisms were related to decreased activation of the PI3K/Akt survival signaling pathway, but not the MAPK pathway, and involved inhibition of the NF-κB activity and downregulation of the Bcl-2/Bax and FLIPL/FLIPS ratios, triggering the activation of caspase cascades. This phenomenon was inhibited by the PI3K inhibitor LY294002. Bortezomib also acted synergistically with the chemotherapeutic agents AraC and DNR, which are associated with the inhibition of NF-κB activity. Our results demonstrate that Bortezomib can induce growth arrest and apoptosis of high-risk MDS cells and had a synergistic effect with two chemotherapeutic agents. Our findings provide new insights for the treatment of high-risk MDS, using either Bortezomib alone, or in combination with conventional antineoplastic agents.
Collapse
Affiliation(s)
- Jian Huang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Hematology, Diagnose and Treatment, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Combined Muti-organ Transplantation, Ministry of Public Health, #79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Ting Ding
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Hematology, Diagnose and Treatment, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Combined Muti-organ Transplantation, Ministry of Public Health, #79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Min Yang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Hematology, Diagnose and Treatment, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Combined Muti-organ Transplantation, Ministry of Public Health, #79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Hui Liu
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Hematology, Diagnose and Treatment, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Combined Muti-organ Transplantation, Ministry of Public Health, #79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Xin Sun
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Hematology, Diagnose and Treatment, Hangzhou, Zhejiang, People's Republic of China.,Key Lab of Combined Muti-organ Transplantation, Ministry of Public Health, #79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Jie Jin
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Lab of Hematology, Diagnose and Treatment, Hangzhou, Zhejiang, People's Republic of China. .,Key Lab of Combined Muti-organ Transplantation, Ministry of Public Health, #79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.
| |
Collapse
|
17
|
Affiliation(s)
- M A Dimopoulos
- Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece.
| | | |
Collapse
|
18
|
Abstract
In this issue of Blood, Richardson et al report on a landmark treatment regimen for newly diagnosed myeloma patients that for the first time combines lenalidomide and bortezomib. This effort required a team approach comprising 2 competing pharmaceutical companies (joining forces to study this promising regimen) and multiple academic medical centers.1
Collapse
|
19
|
Moreau P, Hulin C. Les nouveaux médicaments du myélome. ONCOLOGIE 2010. [DOI: 10.1007/s10269-010-1895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|