1
|
Tosun Ç, Wallabregue ALD, Mallerman M, Phillips SE, Edwards CM, Conway SJ, Hammond EM. Antibody-Based Imaging of Bioreductive Prodrug Release in Hypoxia. JACS AU 2023; 3:3237-3246. [PMID: 38034969 PMCID: PMC10685431 DOI: 10.1021/jacsau.3c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 12/02/2023]
Abstract
Regions of hypoxia occur in most tumors and are a predictor of poor patient prognosis. Hypoxia-activated prodrugs (HAPs) provide an ideal strategy to target the aggressive, hypoxic, fraction of a tumor, while protecting the normal tissue from toxicity. A key challenge associated with the development of novel HAPs, however, is the ability to visualize the delivery of the prodrug to hypoxic regions and determine where it has been activated. Here, we report a modified version of the commonly used nitroimidazole bioreductive group that incorporates the fluoroethyl epitope of the antibody-based hypoxia imaging agent, EF5. Attachment of this group to the red fluorescent dye, dicyanomethylene (DCM), enabled us to correlate the release of the DCM dye with imaging of the reduced bioreductive group using the EF5 antibody. This study confirmed that the antibody was imaging reduction and fragmentation of the pro-fluorophore. We next employed the modified bioreductive group to synthesize a new prodrug of the KDAC inhibitor Panobinostat, EF5-Pano. Release of EF5-Pano in hypoxic multiple myeloma cells was imaged using the EF5 antibody, and the presence of an imaging signal correlated with apoptosis and a reduction in cell viability. Therefore, EF5-Pano is an imageable HAP with a proven cytotoxic effect in multiple myeloma, which could be utilized in future in vivo experiments.
Collapse
Affiliation(s)
- Çağla Tosun
- Department
of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K.
| | - Antoine L. D. Wallabregue
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Maxim Mallerman
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Sarah E. Phillips
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Claire M. Edwards
- Nuffield
Department of Surgical Sciences, University
of Oxford, Oxford OX3 7HE, U.K.
- Nuffield
Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, U.K.
| | - Stuart J. Conway
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
- Department
of Chemistry & Biochemistry, University
of California, 607 Charles
E. Young Drive East, Los Angeles, California CA90095, United States
| | - Ester M. Hammond
- Department
of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K.
| |
Collapse
|
2
|
He Y, Jiang D, Zhang K, Zhu Y, Zhang J, Wu X, Xia J, Zhu Y, Zou L, Hu J, Cui Y, Zhou W, Chen F. Chidamide, a subtype-selective histone deacetylase inhibitor, enhances Bortezomib effects in multiple myeloma therapy. J Cancer 2021; 12:6198-6208. [PMID: 34539893 PMCID: PMC8425211 DOI: 10.7150/jca.61602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/15/2021] [Indexed: 12/25/2022] Open
Abstract
Drug resistance is the major cause for disease relapse and patient death in multiple myeloma (MM). It is an urgent need to develop new therapies to overcome drug resistance in MM. Chidamide (CHI), a novel oral HDAC inhibitor targeting HDAC1, 2, 3 and 10, has shown potential therapeutic effect in MM. In this study, we determined that CHI exhibited significant anti-tumor effect on MM cells both in vitro and in vivo, which was positively correlated with the expression of HDAC1. Meanwhile, CHI enhanced Bortezomib (BTZ) effects synergistically in MM cells and a combination of CHI with BTZ induced myeloma cell apoptosis and G0/G1 arrest in vitro and in vivo. Mechanistically, the synergistic anti-tumor effect of CHI and BTZ was related with the increased production of reactive oxygen species (ROS) dependent DNA damage and the changes of cell apoptosis and cycle pathways. Our data indicate that CHI may be a suitable drug to sensitize BTZ in MM cells, which provides novel insight into the therapy for MM patients.
Collapse
Affiliation(s)
- Yanjuan He
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duanfeng Jiang
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Kaixuan Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinghong Zhu
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jingyu Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xuan Wu
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jiliang Xia
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yan Zhu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lang Zou
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Hu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yajuan Cui
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Fangping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Bertamini L, Bonello F, Boccadoro M, Bringhen S. New drugs in early development for treating multiple myeloma: all that glitters is not gold. Expert Opin Investig Drugs 2020; 29:989-1004. [PMID: 32434394 DOI: 10.1080/13543784.2020.1772753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The last twenty years have introduced new therapeutic agents for multiple myeloma (MM); these include proteasome inhibitors (PIs), immunomodulatory drugs (IMDs) and monoclonal antibodies (mAbs). However, MM remains incurable, hence there is an unmet need for new agents for the treatment of advanced refractory disease. New agents could also be used in early lines to achieve improved, more sustained remission. AREAS COVERED We review the most promising agents investigated in early-phase trials for the treatment of MM and provide an emphasis on new agents directed against well-known targets (new PIs, IMDs and anti-CD38 mAbs). Drugs that work through distinct and numerous mechanisms of action (e.g. pro-apoptotic agents and tyrosine kinase inhibitors) and innovative immunotherapeutic approaches are also described. The paper culminates with our perspective on therapeutic approaches on the horizon for this disease. EXPERT OPINION IMD iberdomide and the export protein inhibitor selinexor demonstrated efficacy in heavily pretreated patients who had no other therapeutic options. We expect that immunotherapy with anti-BCMA BTEs and ADCs will revolutionize the approach to treating the early stages of the disease. Data on venetoclax in t(11;14)-positive patients may pave the way for personalized therapy. Not all new agents under early clinical evaluation will be investigated in regulatory phase III trials; one of the most important challenges is to identify those that could make a difference.
Collapse
Affiliation(s)
- Luca Bertamini
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Francesca Bonello
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| | - Sara Bringhen
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino , Torino, Italy
| |
Collapse
|
4
|
Gawel JM, Shouksmith AE, Raouf YS, Nawar N, Toutah K, Bukhari S, Manaswiyoungkul P, Olaoye OO, Israelian J, Radu TB, Cabral AD, Sina D, Sedighi A, de Araujo ED, Gunning PT. PTG-0861: A novel HDAC6-selective inhibitor as a therapeutic strategy in acute myeloid leukaemia. Eur J Med Chem 2020; 201:112411. [PMID: 32615502 DOI: 10.1016/j.ejmech.2020.112411] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/25/2022]
Abstract
Dysregulated Histone Deacetylase (HDAC) activity across multiple human pathologies have highlighted this family of epigenetic enzymes as critical druggable targets, amenable to small molecule intervention. While efficacious, current approaches using non-selective HDAC inhibitors (HDACi) have been shown to cause a range of undesirable clinical toxicities. To circumvent this, recent efforts have focused on the design of highly selective HDACi as a novel therapeutic strategy. Beyond roles in regulating transcription, the unique HDAC6 (with two catalytic domains) regulates the deacetylation of α-tubulin; promoting growth factor-controlled cell motility, cell division, and metastatic hallmarks. Recent studies have linked aberrant HDAC6 function in various hematological cancers including acute myeloid leukaemia and multiple myeloma. Herein, we report the discovery, in vitro characterization, and biological evaluation of PTG-0861 (JG-265), a novel HDAC6-selective inhibitor with strong isozyme-selectivity (∼36× ) and low nanomolar potency (IC50 = 5.92 nM) against HDAC6. This selectivity profile was rationalized via in silico docking studies and also observed in cellulo through cellular target engagement. Moreover, PTG-0861 achieved relevant potency against several blood cancer cell lines (e.g. MV4-11, MM1S), whilst showing limited cytotoxicity against non-malignant cells (e.g. NHF, HUVEC) and CD-1 mice. In examining compound stability and cellular permeability, PTG-0861 revealed a promising in vitro pharmacokinetic (PK) profile. Altogether, in this study we identified a novel and potent HDAC6-selective inhibitor (∼4× more selective than current clinical standards - citarinostat, ricolinostat), which achieves cellular target engagement, efficacy in hematological cancer cells with a promising safety profile and in vitro PK.
Collapse
Affiliation(s)
- Justyna M Gawel
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada
| | - Andrew E Shouksmith
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada
| | - Yasir S Raouf
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Nabanita Nawar
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Krimo Toutah
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada
| | - Shazreh Bukhari
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Pimyupa Manaswiyoungkul
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Olasunkanmi O Olaoye
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Johan Israelian
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Tudor B Radu
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Aaron D Cabral
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Diana Sina
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Abootaleb Sedighi
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada
| | - Elvin D de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada
| | - Patrick T Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, 3359, Mississauga Road, Mississauga, Ontario, L5L 1C6, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada.
| |
Collapse
|
5
|
Zhao LM, Zhang JH. Histone Deacetylase Inhibitors in Tumor Immunotherapy. Curr Med Chem 2019; 26:2990-3008. [PMID: 28762309 DOI: 10.2174/0929867324666170801102124] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 04/26/2017] [Accepted: 06/27/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND With an increasing understanding of the antitumor immune response, considerable progress has been made in the field of tumor immunotherapy in the last decade. Inhibition of histone deacetylases represents a new strategy in tumor therapy and histone deacetylase inhibitors have been recently developed and validated as potential antitumor drugs. In addition to the direct antitumor effects, histone deacetylase inhibitors have been found to have the ability to improve tumor recognition by immune cells that may contribute to their antitumor activity. These immunomodolutory effects are desirable, and their in-depth comprehension will facilitate the design of novel regimens with improved clinical efficacy. OBJECTIVE Our goal here is to review recent developments in the application of histone deacetylase inhibitors as immune modulators in cancer treatment. METHODS Systemic compilation of the relevant literature in this field. RESULTS & CONCLUSION In this review, we summarize recent advances in the understanding of how histone deacetylase inhibitors alter immune process and discuss their effects on various cytokines. We also discuss the challenges to optimize the use of these inhibitors as immune modulators in cancer treatment. Information gained from this review will be valuable to this field and may be helpful for designing tumor immunotherapy trials involving histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Li-Ming Zhao
- School of Chemistry and Chemical Engineering, and Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.,State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Jie-Huan Zhang
- School of Chemistry and Chemical Engineering, and Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
6
|
Cengiz Seval G, Beksac M. A comparative safety review of histone deacetylase inhibitors for the treatment of myeloma. Expert Opin Drug Saf 2019; 18:563-571. [PMID: 31070945 DOI: 10.1080/14740338.2019.1615051] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Dysregulation of histone deacetylase (HDAC) activity is an epigenetic hallmark of multiple myeloma (MM), leading to aberrant gene expression and cellular signaling in myeloma cell growth, survival and resistance to therapy. Hyper-methylation at diagnosis is a frequent observation, which eventually may convert to hypo-methylation during advanced phases. AREAS COVERED A literature search on 'HDAC inhibitors' and 'multiple myeloma' was carried out using PubMed and Google Scholar in the preparation of this overview on clinical efficacy and safety data. EXPERT OPINION First-generation non-selective HDAC inhibitors have demonstrated minimal single-agent activity in refractory MM. Subsequently, combination therapy has proven an improvement in progression-free survival (PFS) but not response rates. The main concerns are associated with toxicities. Ongoing studies on new and more selective agents, i.e. Romidepsin or Ricolinostat, are promising in terms of better efficacy and less toxicity.
Collapse
Affiliation(s)
- Guldane Cengiz Seval
- a Department of Hematology , Ankara University School of Medicine, Cebeci Hospital , Mamak , Turkey
| | - Meral Beksac
- a Department of Hematology , Ankara University School of Medicine, Cebeci Hospital , Mamak , Turkey
| |
Collapse
|
7
|
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: 23] [Impact Index Per Article: 3.8] [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
|
8
|
Hillert EK, Brnjic S, Zhang X, Mazurkiewicz M, Saei AA, Mofers A, Selvaraju K, Zubarev R, Linder S, D'Arcy P. Proteasome inhibitor b-AP15 induces enhanced proteotoxicity by inhibiting cytoprotective aggresome formation. Cancer Lett 2019; 448:70-83. [PMID: 30768956 DOI: 10.1016/j.canlet.2019.02.003] [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] [Received: 11/08/2018] [Revised: 12/28/2018] [Accepted: 02/01/2019] [Indexed: 01/26/2023]
Abstract
Proteasome inhibitors have been shown to induce cell death in cancer cells by triggering an acute proteotoxic stress response characterized by accumulation of poly-ubiquitinated proteins, ER stress and the production of reactive oxygen species. The aggresome pathway has been described as an escape mechanism from proteotoxicity by sequestering toxic cellular aggregates. Here we show that b-AP15, a small-molecule inhibitor of proteasomal deubiquitinase activity, induces poly-ubiquitin accumulation in absence of aggresome formation. b-AP15 was found to affect organelle transport in treated cells, raising the possibility that microtubule-transport of toxic protein aggregates is inhibited, leading to enhanced cytotoxicity. In contrast to the antiproliferative effects of the clinically used proteasome inhibitor bortezomib, the effects of b-AP15 are not further enhanced by the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA). Our results suggest an inhibitory effect of b-AP15 on the transport of misfolded proteins, resulting in a lack of aggresome formation, and a strong proteotoxic stress response.
Collapse
Affiliation(s)
| | - Slavica Brnjic
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Xiaonan Zhang
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Amir Ata Saei
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Arjan Mofers
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Karthik Selvaraju
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Roman Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Stig Linder
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Padraig D'Arcy
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
9
|
Monoclonal Antibodies versus Histone Deacetylase Inhibitors in Combination with Bortezomib or Lenalidomide plus Dexamethasone for the Treatment of Relapsed or Refractory Multiple Myeloma: An Indirect-Comparison Meta-Analysis of Randomized Controlled Trials. J Immunol Res 2018; 2018:7646913. [PMID: 30050957 PMCID: PMC6040277 DOI: 10.1155/2018/7646913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/14/2018] [Indexed: 12/26/2022] Open
Abstract
During the past decades, agents with novel mechanisms of action, such as monoclonal antibodies (MAbs) and histone deacetylase inhibitors (HDACis) have been applied to treat relapsed or refractory multiple myeloma (RRMM). The treatment outcomes of MAbs versus HDACi in combination with bortezomib or lenalidomide plus dexamethasone remain unknown. We conducted this meta-analysis to compare indirectly the efficacy and safety of MAbs and HDACis in combination with bortezomib or lenalidomide plus dexamethasone. Six trials (eight articles) were included in the meta-analysis with 3270 RRMM patients enrolled. We synthesized hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS), risk ratios (RRs) for complete response (CR),very good partial response (VGPR), overall response (OR), progressive disease plus stable disease (PD + SD) and common at least grade 3 adverse events, and their corresponding 95%confidence intervals (95% CI). Treatment with MAbs in combination with bortezomib or lenalidomide plus dexamethasone resulted in longer PFS (HR 0.83, 95% CI: 0.66-0.98), fewer incidences of at least grade 3 thrombocytopenia (RR 0.35, 95% CI: 0.23-0.53), neutropenia (RR 0.70, 95% CI: 0.51-0.96), and sense of fatigue (RR 0.37, 95% CI: 0.17-0.82) than HDACis. The daratumumab plus bortezomib or lenalidomide and dexamethasone might significantly improve PFS in comparison with HDACis plus bortezomib or lenalidomide and dexamethasone (HR 0.55, 95% CI: 0.40-0.74). In conclusion, MAbs may be superior to HDACis in achieving longer PFS and may be better tolerated when in combination therapy with bortezomib or lenalidomide plus dexamethasone.
Collapse
|
10
|
Abstract
FDA and EMA approval of panobinostat offers an additional therapeutic option for multiple myeloma; however, adoption of panobinostat has been limited by its adverse event profile. Trials are ongoing to optimize the dosing of panobinostat and to identify its best partners, in order to fully realize the potential of this drug class.
Collapse
Affiliation(s)
- Andrew J Yee
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Noopur S Raje
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
The IgH 3' regulatory region and c-myc-induced B-cell lymphomagenesis. Oncotarget 2018; 8:7059-7067. [PMID: 27729620 PMCID: PMC5351691 DOI: 10.18632/oncotarget.12535] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/05/2016] [Indexed: 01/18/2023] Open
Abstract
Deregulation and mutations of c-myc have been reported in multiple mature B-cell malignancies such as Burkitt lymphoma, myeloma and plasma cell lymphoma. After translocation into the immunoglobulin heavy chain (IgH) locus, c-myc is constitutively expressed under the control of active IgH cis-regulatory enhancers. Those located in the IgH 3 regulatory region (3RR) are master control elements of transcription. Over the past decade numerous convincing demonstrations of 3RRs contribution to mature c-myc-induced lymphomagenesis have been made using transgenic models with various types of IgH-c-myc translocations and transgenes. This review highlights how IgH 3RR physiological functions play a critical role in c-myc deregulation during lymphomagenesis.
Collapse
|
12
|
Nijhof IS, van de Donk NWCJ, Zweegman S, Lokhorst HM. Current and New Therapeutic Strategies for Relapsed and Refractory Multiple Myeloma: An Update. Drugs 2018; 78:19-37. [PMID: 29188449 PMCID: PMC5756574 DOI: 10.1007/s40265-017-0841-y] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although survival of multiple myeloma patients has at least doubled during recent years, most patients eventually relapse, and treatment at this stage may be particularly complex. At the time of relapse, the use of alternative drugs to those given upfront is current practice. However, many new options are currently available for the treatment of relapsed multiple myeloma, including recently approved drugs, such as the second- and third-generation proteasome inhibitors carfilzomib and ixazomib, the immunomodulatory agent pomalidomide, the monoclonal antibodies daratumumab and elotuzumab and the histone deacetylase inhibitor panobinostat, but also new targeted agents are under active investigation (e.g. signal transduction modulators, kinesin spindle protein inhibitors, and inhibitors of NF-kB, MAPK, AKT). We here describe a new paradigm for the treatment of relapsed multiple myeloma. The final goal should be finding a balance among efficacy, toxicity, and cost and, at the end of the road, achieving long-lasting control of the disease and eventually even cure in a subset of patients.
Collapse
Affiliation(s)
- Inger S Nijhof
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Niels W C J van de Donk
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Henk M Lokhorst
- Department of Hematology, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Poff A, Koutnik AP, Egan KM, Sahebjam S, D'Agostino D, Kumar NB. Targeting the Warburg effect for cancer treatment: Ketogenic diets for management of glioma. Semin Cancer Biol 2017; 56:135-148. [PMID: 29294371 DOI: 10.1016/j.semcancer.2017.12.011] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/07/2017] [Accepted: 12/29/2017] [Indexed: 12/29/2022]
Abstract
Gliomas are a highly heterogeneous tumor, refractory to treatment and the most frequently diagnosed primary brain tumor. Although the current WHO grading system (2016) demonstrates promise towards identifying novel treatment modalities and better prediction of prognosis over time, to date, existing targeted and mono therapy approaches have failed to elicit a robust impact on disease progression and patient survival. It is possible that tumor heterogeneity as well as specifically targeted agents fail because redundant molecular pathways in the tumor make it refractory to such approaches. Additionally, the underlying metabolic pathology, which is significantly altered during neoplastic transformation and tumor progression, is unaccounted for. With several molecular and metabolic pathways implicated in the carcinogenesis of CNS tumors, including glioma, we postulate that a systemic, broad spectrum approach to produce robust targeting of relevant and multiple molecular and metabolic regulation of growth and survival pathways, critical to the modulation of hallmarks of carcinogenesis, without clinically limiting toxicity, may provide a more sustained impact on clinical outcomes compared to the modalities of treatment evaluated to date. The objective of this review is to examine the emerging hallmark of reprogramming energy metabolism of the tumor cells and the tumor microenvironment during carcinogenesis, and to provide a rationale for exploiting this hallmark and its biological capabilities as a target for secondary chemoprevention and treatment of glioma. This review will primarily focus on interventions to induce ketosis to target the glycolytic phenotype of many cancers, with specific application to secondary chemoprevention of low grade glioma- to halt the progression of lower grade tumors to more aggressive subtypes, as evidenced by reduction in validated intermediate endpoints of disease progression including clinical symptoms.
Collapse
Affiliation(s)
- Angela Poff
- The University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B. Downs Blvd, MDC 8, Tampa, FL 33612, United States.
| | - Andrew P Koutnik
- The University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B. Downs Blvd, MDC 8, Tampa, FL 33612, United States.
| | - Kathleen M Egan
- Moffitt Cancer Center, H. Lee Moffitt Cancer Center and Research Institute, Department of Cancer Epidemiology, 12902 Magnolia Drive, MRC/CANCONT, Tampa, FL 22612-9497, United States.
| | - Solmaz Sahebjam
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Department of Cancer Epidemiology, 12902 Magnolia Drive, Tampa, FL 22612-9497, United States.
| | - Dominic D'Agostino
- The University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B. Downs Blvd, MDC 8, Tampa, FL 33612, United States.
| | - Nagi B Kumar
- Moffitt Cancer Center, H. Lee Moffitt Cancer Center and Research Institute, Department of Cancer Epidemiology, 12902 Magnolia Drive, MRC/CANCONT, Tampa, FL 22612-9497, United States.
| |
Collapse
|
14
|
Saintamand A, Ghazzaui N, Issaoui H, Denizot Y. [The IgH 3'RR: Doctor Jekyll and Mister Hyde of B-cell maturation and lymphomagenesis]. Med Sci (Paris) 2017; 33:963-970. [PMID: 29200394 DOI: 10.1051/medsci/20173311013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The four transcriptional enhancers located in the 3' regulatory region (3'RR) of the IgH locus control the late phases of B-cell maturation, namely IgH locus transcription, somatic hypermutation and class switch recombination. Doctor Jekyll by nature, the 3'RR acts as Mister Hyde in case of oncogenic translocation at the IgH locus taking under its transcriptional control the translocated oncogene. The aim of this review is to show this duality on the basis of the latest scientific advances in the structure and function of the 3'RR and to hIghlight the targeting of the 3'RR as a potential therapeutic approach in mature B-cell lymphomas.
Collapse
Affiliation(s)
- Alexis Saintamand
- UMR CNRS 7276, Université de Limoges, rue Pr Descottes, 87025 Limoges, France
| | - Nour Ghazzaui
- UMR CNRS 7276, Université de Limoges, rue Pr Descottes, 87025 Limoges, France
| | - Hussein Issaoui
- UMR CNRS 7276, Université de Limoges, rue Pr Descottes, 87025 Limoges, France
| | - Yves Denizot
- UMR CNRS 7276, Université de Limoges, rue Pr Descottes, 87025 Limoges, France
| |
Collapse
|
15
|
Haney SL, Allen C, Varney ML, Dykstra KM, Falcone ER, Colligan SH, Hu Q, Aldridge AM, Wright DL, Wiemer AJ, Holstein SA. Novel tropolones induce the unfolded protein response pathway and apoptosis in multiple myeloma cells. Oncotarget 2017; 8:76085-76098. [PMID: 29100294 PMCID: PMC5652688 DOI: 10.18632/oncotarget.18543] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022] Open
Abstract
Tropolones are small organic compounds with metal-directing moieties. Tropolones inhibit the proliferation of cancer cell lines, possibly through their effects on metalloenzymes such as select histone deacetylases (HDACs). Pan-HDAC inhibitors are therapeutically beneficial in the treatment of multiple myeloma, however there is interest in the use of more selective HDAC inhibitor therapy to minimize adverse side effects. We hypothesized that tropolones might have anti-myeloma activities. To this end, a series of novel α-substituted tropolones were evaluated for effects on multiple myeloma cells. While all tested tropolones showed some level of cytotoxicity, MO-OH-Nap had consistently low IC50 values between 1-11 μM in all three cell lines tested and was used for subsequent experiments. MO-OH-Nap was found to induce apoptosis in a concentration-dependent manner. Time course experiments demonstrated that MO-OH-Nap promotes caspase cleavage in a time frame that was distinct from the pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA). Furthermore, MO-OH-Nap- and SAHA-treated cells possess unique gene expression patterns, suggesting they promote apoptosis via different mechanisms. In particular, MO-OH-Nap increases the expression of markers associated with endoplasmic reticulum stress and the unfolded protein response. Synergistic cytotoxic effects were observed when cells were treated with the combination of MO-OH-Nap and the proteasome inhibitor bortezomib. However, treatment with MO-OH-Nap did not abrogate the bortezomib-induced increase in aggresomes, consistent with an HDAC6-independent mechanism for the observed synergy. Collectively, these finding support further investigation into the usefulness of α-substituted tropolones as anti-myeloma agents.
Collapse
Affiliation(s)
- Staci L. Haney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cheryl Allen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Michelle L. Varney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Eric R. Falcone
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Sean H. Colligan
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Qiang Hu
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Dennis L. Wright
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Andrew J. Wiemer
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Sarah A. Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
16
|
Abstract
Oral panobinostat (Farydak®), a potent nonselective histone deacetylase inhibitor, is approved in several countries for use in combination with bortezomib and dexamethasone in patients with multiple myeloma (MM) [USA] or relapsed and/or refractory MM (EU) who have received at least two prior treatment regimens, including bortezomib and an immunomodulatory drug (IMiD). In a pivotal phase III trial (PANORAMA 1) in patients with relapsed or relapsed and refractory MM who had received one to three previous lines of therapy, progression-free survival (PFS) was significantly prolonged with panobinostat plus bortezomib and dexamethasone compared with placebo plus bortezomib and dexamethasone. The significantly favourable effect of panobinostat- versus placebo-based treatment on PFS was also observed in a subgroup analysis of patients who had previously received an IMiD, bortezomib plus an IMiD, or at least two lines of treatment including bortezomib and an IMiD. Panobinostat plus bortezomib and dexamethasone had a generally manageable tolerability profile, with the most frequent grade 3-4 adverse events being myelosuppression, diarrhoea, asthenia or fatigue, peripheral neuropathy and pneumonia. Thus, panobinostat, in combination with bortezomib and dexamethasone, is a useful addition to the available treatment options for patients with relapsed or refractory MM.
Collapse
Affiliation(s)
- Sarah L Greig
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
17
|
Sanchez L, Vesole DH, Richter JR, Biran N, Bilotti E, McBride L, Anand P, Ivanovski K, Siegel DS. A phase IIb trial of vorinostat in combination with lenalidomide and dexamethasone in patients with multiple myeloma refractory to previous lenalidomide-containing regimens. Br J Haematol 2016; 176:440-447. [DOI: 10.1111/bjh.14429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/22/2016] [Indexed: 01/13/2023]
Affiliation(s)
| | - David H. Vesole
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - Joshua R. Richter
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - Noa Biran
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - Elizabeth Bilotti
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - Laura McBride
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - Palka Anand
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - Kristin Ivanovski
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| | - David S. Siegel
- Division of Multiple Myeloma; John Theurer Cancer Center; Hackensack University Medical Center; Hackensack NJ USA
| |
Collapse
|
18
|
Pratt G. Panobinostat plus bortezomib and dexamethasone for relapsed myeloma. LANCET HAEMATOLOGY 2016; 3:e498-e499. [PMID: 27751706 DOI: 10.1016/s2352-3026(16)30133-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 11/25/2022]
Affiliation(s)
- Guy Pratt
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham B15 2TH, UK.
| |
Collapse
|
19
|
Yee AJ, Bensinger WI, Supko JG, Voorhees PM, Berdeja JG, Richardson PG, Libby EN, Wallace EE, Birrer NE, Burke JN, Tamang DL, Yang M, Jones SS, Wheeler CA, Markelewicz RJ, Raje NS. Ricolinostat plus lenalidomide, and dexamethasone in relapsed or refractory multiple myeloma: a multicentre phase 1b trial. Lancet Oncol 2016; 17:1569-1578. [PMID: 27646843 DOI: 10.1016/s1470-2045(16)30375-8] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Histone deacetylase (HDAC) inhibitors are an important new class of therapeutics for treating multiple myeloma. Ricolinostat (ACY-1215) is the first oral selective HDAC6 inhibitor with reduced class I HDAC activity to be studied clinically. Motivated by findings from preclinical studies showing potent synergistic activity with ricolinostat and lenalidomide, our goal was to assess the safety and preliminary activity of the combination of ricolinostat with lenalidomide and dexamethasone in relapsed or refractory multiple myeloma. METHODS In this multicentre phase 1b trial, we recruited patients aged 18 years or older with previously treated relapsed or refractory multiple myeloma from five cancer centres in the USA. Inclusion criteria included a Karnofsky Performance Status score of at least 70, measureable disease, adequate bone marrow reserve, adequate hepatic function, and a creatinine clearance of at least 50 mL per min. Exclusion criteria included previous exposure to HDAC inhibitors; previous allogeneic stem-cell transplantation; previous autologous stem-cell transplantation within 12 weeks of baseline; active systemic infection; malignancy within the last 5 years; known or suspected HIV, hepatitis B, or hepatitis C infection; a QTc Fridericia of more than 480 ms; and substantial cardiovascular, gastrointestinal, psychiatric, or other medical disorders. We gave escalating doses (from 40-240 mg once daily to 160 mg twice daily) of oral ricolinostat according to a standard 3 + 3 design according to three different regimens on days 1-21 with a conventional 28 day schedule of oral lenalidomide (from 15 mg [in one cohort] to 25 mg [in all other cohorts] once daily) and oral dexamethasone (40 mg weekly). Primary outcomes were dose-limiting toxicities, the maximum tolerated dose of ricolinostat in this combination, and the dose and schedule of ricolinostat recommended for further phase 2 investigation. Secondary outcomes were the pharmacokinetics and pharmacodynamics of ricolinostat in this combination and the preliminary anti-tumour activity of this treatment. The trial is closed to accrual and is registered at ClinicalTrials.gov, number NCT01583283. FINDINGS Between July 12, 2012, and Aug 20, 2015, we enrolled 38 patients. We observed two dose-limiting toxicities with ricolinostat 160 mg twice daily: one (2%) grade 3 syncope and one (2%) grade 3 myalgia event in different cohorts. A maximum tolerated dose was not reached. We chose ricolinostat 160 mg once daily on days 1-21 of a 28 day cycle as the recommended dose for future phase 2 studies in combination with lenalidomide 25 mg and dexamethasone 40 mg. The most common adverse events were fatigue (grade 1-2 in 14 [37%] patients; grade 3 in seven [18%]) and diarrhoea (grade 1-2 in 15 [39%] patients; grade 3 in two [5%]). Our pharmacodynamic studies showed that at clinically relevant doses, ricolinostat selectively inhibits HDAC6 while retaining a low and tolerable level of class I HDAC inhibition. The pharmacokinetics of ricolinostat and lenalidomide were not affected by co-administration. In a preliminary assessment of antitumour activity, 21 (55% [95% CI 38-71]) of 38 patients had an overall response. INTERPRETATION The findings from this study provide preliminary evidence that ricolinostat is a safe and well tolerated selective HDAC6 inhibitor, which might partner well with lenalidomide and dexamethasone to enhance their efficacy in relapsed or refractory multiple myeloma. FUNDING Acetylon Pharmaceuticals.
Collapse
Affiliation(s)
- Andrew J Yee
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Jeffrey G Supko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Paul G Richardson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Ellen E Wallace
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Nicole E Birrer
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jill N Burke
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Min Yang
- Acetylon Pharmaceuticals, Boston, MA, USA
| | | | | | | | - Noopur S Raje
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Smith EM, Zhang L, Walker BA, Davenport EL, Aronson LI, Krige D, Hooftman L, Drummond AH, Morgan GJ, Davies FE. The combination of HDAC and aminopeptidase inhibitors is highly synergistic in myeloma and leads to disruption of the NFκB signalling pathway. Oncotarget 2016; 6:17314-27. [PMID: 26015393 PMCID: PMC4627310 DOI: 10.18632/oncotarget.1168] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/10/2013] [Indexed: 02/05/2023] Open
Abstract
There is a growing body of evidence supporting the use of epigenetic therapies in the treatment of multiple myeloma. We show the novel HDAC inhibitor CHR-3996 induces apoptosis in myeloma cells at concentrations in the nanomolar range and with apoptosis mediated by p53 and caspase pathways. In addition, HDAC inhibitors are highly synergistic, both in vitro and in vivo, with the aminopeptidase inhibitor tosedostat (CHR-2797). We demonstrate that the basis for this synergy is a consequence of changes in the levels of NFκB regulators BIRC3/cIAP2, A20, CYLD, and IκB, which were markedly affected by the combination. When co-administered the HDAC and aminopeptidase inhibitors caused rapid nuclear translocation of NFκB family members p65 and p52, following activation of both canonical and non-canonical NFκB signalling pathways. The subsequent up-regulation of inhibitors of NFκB activation (most significantly BIRC3/cIAP2) turned off the cytoprotective effects of the NFκB signalling response in a negative feedback loop. These results provide a rationale for combining HDAC and aminopeptidase inhibitors clinically for the treatment of myeloma patients and support the disruption of the NFκB signalling pathway as a therapeutic strategy.
Collapse
Affiliation(s)
- Emma M Smith
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| | - Lei Zhang
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| | - Brian A Walker
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| | - Emma L Davenport
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| | - Lauren I Aronson
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| | | | | | | | - Gareth J Morgan
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| | - Faith E Davies
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Cancer Therapeutics and Clinical Studies, The Institute of Cancer Research, London, UK
| |
Collapse
|
21
|
Maes K, De Smedt E, Kassambara A, Hose D, Seckinger A, Van Valckenborgh E, Menu E, Klein B, Vanderkerken K, Moreaux J, De Bruyne E. In vivo treatment with epigenetic modulating agents induces transcriptional alterations associated with prognosis and immunomodulation in multiple myeloma. Oncotarget 2016; 6:3319-34. [PMID: 25669970 PMCID: PMC4413656 DOI: 10.18632/oncotarget.3207] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/17/2014] [Indexed: 12/11/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) and DNA methyltransferase inhibitors (DNMTi) are in early clinical development for multiple myeloma (MM) therapy. Despite all encouraging pre-clinical data, clinical activity of HDACi and DNMTi is mostly lacking. To optimize the trials, characterization of the in vivo response towards HDACi and DNMTi will be crucial. Therefore, we investigated the transcriptional response after in vivo treatment with the HDACi quisinostat or DNMTi decitabine using the murine 5T33MM model. We identified 504 and 154 genes deregulated by quisinostat and decitabine, respectively. Of interest, MM patients' gene expression levels of 62 quisinostat- and 25 decitabine-deregulated genes were predictive for overall survival of patients. This prognostic information was implemented in a DNA methylation and histone acetylation score. A high score was related to a high proliferative and immature phenotype of MM cells. Furthermore, highly scored MM patients had an adverse overall survival. Interestingly, bio-informatic prediction tools revealed an association of quisinostat-deregulated genes with lymphocyte activation, proliferation, immune-effector mechanisms and T-helper-1 development. Overall, treatment of 5T33MM mice with epigenetic modulating agents led to the translation of gene signatures to predict overall survival of MM patients. HDACi mainly deregulated tumoral immunomodulatory pathways, supporting the rationale to combine HDACi with immunomodulatory therapies.
Collapse
Affiliation(s)
- Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva De Smedt
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alboukadel Kassambara
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, Montpellier, France
| | - Dirk Hose
- Medizinische Klinik, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Anja Seckinger
- Medizinische Klinik, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bernard Klein
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, Montpellier, France.,University of Montpellier 1, UFR de Médecine, Montpellier, France
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jérôme Moreaux
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UPR1142, Montpellier, France.,University of Montpellier 1, UFR de Médecine, Montpellier, France
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
22
|
Fernández-Rodríguez C, Salar A, Navarro A, Gimeno E, Pairet S, Camacho L, Ferraro M, Serrano S, Besses C, Bellosillo B, Sanchez-Gonzalez B. Anti-tumor activity of the combination of bendamustine with vorinostat in diffuse large B-cell lymphoma cells. Leuk Lymphoma 2016; 57:692-9. [DOI: 10.3109/10428194.2015.1063143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - Antonio Salar
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei d’Hematologia, Hospital del Mar, Barcelona, Spain
| | - Alfons Navarro
- Human Anatomy Unit, School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Eva Gimeno
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei d’Hematologia, Hospital del Mar, Barcelona, Spain
| | - Silvia Pairet
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Laura Camacho
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | - Sergi Serrano
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
| | - Carles Besses
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei d’Hematologia, Hospital del Mar, Barcelona, Spain
| | - Beatriz Bellosillo
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei de Patologia, Hospital del Mar, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Blanca Sanchez-Gonzalez
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei d’Hematologia, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
23
|
Mensah AA, Kwee I, Gaudio E, Rinaldi A, Ponzoni M, Cascione L, Fossati G, Stathis A, Zucca E, Caprini G, Bertoni F. Novel HDAC inhibitors exhibit pre-clinical efficacy in lymphoma models and point to the importance of CDKN1A expression levels in mediating their anti-tumor response. Oncotarget 2016; 6:5059-71. [PMID: 25671298 PMCID: PMC4467133 DOI: 10.18632/oncotarget.3239] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/25/2014] [Indexed: 01/03/2023] Open
Abstract
We investigated the pre-clinical activities of two novel histone deacetylase inhibitors (HDACi), ITF-A and ITF-B, in a large panel of pre-clinical lymphoma models. The two compounds showed a dose-dependent anti-proliferative activity in the majority of cell lines. Gene expression profiling (GEP) of diffuse large B-cell lymphoma (DLBCL) cells treated with the compounds showed a modulation of genes involved in chromatin structure, cell cycle progression, apoptosis, B-cell signaling, and genes encoding metallothioneins. Cell lines showed differences between the concentrations of ITF-A and ITF-B needed to cause anti-proliferative or cytotoxic activity, and cell cycle and apoptosis genes appeared implicated in determining the type of response. In particular, CDKN1A expression was higher in DLBCL cells that, to undergo apoptosis, required a much higher amount of drug than that necessary to induce only an anti-proliferative effect. In conclusion, the two novel HDACi ITF-A and ITF-B demonstrated anti-proliferative activity across different mature B-cell lymphoma cell lines. Basal CDKN1A levels appeared to be important in determining the gap between HDACi concentrations causing cell cycle arrest and those that lead to cell death.
Collapse
Affiliation(s)
- Afua Adjeiwaa Mensah
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Ivo Kwee
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,Dalle Molle Institute for Artificial Intelligence (IDSIA), Manno, Switzerland
| | - Eugenio Gaudio
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Andrea Rinaldi
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland
| | - Maurilio Ponzoni
- Unit of Lymphoid Malignancies, Department of Onco-Hematology, San Raffaele Scientific Institute, Milan, Italy
| | - Luciano Cascione
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Gianluca Fossati
- Preclinical R&D Department, Italfarmaco S.p.A., Cinisello Balsamo, Milan, Italy
| | - Anastasios Stathis
- IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Emanuele Zucca
- IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Gianluca Caprini
- Preclinical R&D Department, Italfarmaco S.p.A., Cinisello Balsamo, Milan, Italy
| | - Francesco Bertoni
- Lymphoma & Genomics Research Program, IOR Institute of Oncology Research, Bellinzona, Switzerland.,IOSI Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| |
Collapse
|
24
|
Suberoylanilide hydroxamic acid synergistically enhances the antitumor activity of etoposide in Ewing sarcoma cell lines. Anticancer Drugs 2015; 26:843-51. [PMID: 26053276 DOI: 10.1097/cad.0000000000000256] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ewing sarcomas (ES) are highly malignant tumors arising in bone and soft tissues. Given the poor outcome of affected patients with primary disseminated disease or at relapse, there is a clear need for new targeted therapies. The HDAC inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA, Vorinostat) inhibits ES tumor growth and induces apoptosis in vitro and in vivo. Thus, SAHA may be considered a novel treatment. However, it is most likely that not a single agent but a combination of agents with synergistic mechanisms will help improve the prognosis in high-risk ES patients. Therefore, the aim of the present study was to assess a putative synergistic effect of SAHA in combination with conventional chemotherapeutic agents. The antitumor activity of SAHA in combination with conventional chemotherapeutics (doxorubicin, etoposide, rapamycin, topotecan) was assessed using an MTT cell proliferation assay on five well-characterized ES cell lines (CADO-ES-1, RD-ES, TC-71, SK-ES-1, SK-N-MC) and a newly established ES cell line (DC-ES-15). SAHA antagonistically affected the antiproliferative effect of doxorubicin and topotecan in the majority of the ES cell lines, but synergistically enhanced the antiproliferative activity of etoposide. In functional analyses, pretreatment with SAHA significantly increased the effects of etoposide on apoptosis and clonogenicity. The in-vitro analyses presented in this work show that SAHA synergistically enhances the antitumor activity of etoposide in ES cells. Sequential treatment with etoposide combined with SAHA may represent a new therapeutic approach in ES.
Collapse
|
25
|
Mu S, Kuroda Y, Shibayama H, Hino M, Tajima T, Corrado C, Lin R, Waldron E, Binlich F, Suzuki K. Panobinostat PK/PD profile in combination with bortezomib and dexamethasone in patients with relapsed and relapsed/refractory multiple myeloma. Eur J Clin Pharmacol 2015; 72:153-61. [PMID: 26494130 PMCID: PMC4713719 DOI: 10.1007/s00228-015-1967-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/14/2015] [Indexed: 11/29/2022]
Abstract
PURPOSE Panobinostat, a potent pan-deacetylase inhibitor, improved progression-free survival (PFS) in patients with relapsed and refractory multiple myeloma when combined with bortezomib and dexamethasone in a phase 3 trial, PANORAMA-1. This study aims to explore exposure-response relationship for panobinostat in this combination in a phase 1 trial, B2207 and contrast with data from historical single-agent studies. METHODS Panobinostat plasma concentration-time profiles were obtained in patients from PANORAMA-1 (n = 12) and B2207 (n = 12) trials. Overall response rates (ORR) and major adverse events (AE) by panobinostat exposure were investigated in the B2207 trial. Panobinostat PK data from combination trials were contrasted with data from single-agent studies. RESULTS At maximum tolerated dose (MTD), the geometric mean of panobinostat area under curve from 0 to 24 h (AUC0-24) was 47.5 ng h/mL (77 % CV), and maximum plasma concentration (Cmax) was 8.1 ng/mL (90 % CV). These values were comparable with exposure data obtained in PANORAMA-1, but were 20 % lower than those without dexamethasone, and ∼ 50 % lower from single-agent trials, likely due to enzyme induction by dexamethasone. Higher levels of panobinostat exposure were associated with higher response rates and higher incidences of diarrhea and thrombocytopenia. CONCLUSIONS Apparent panobinostat exposure-AE and exposure-ORR relationships were observed when combined with bortezomib and dexamethasone in the treatment of patients with relapsed and refractory multiple myeloma. The addition of dexamethasone facilitated best response even though plasma exposure of panobinostat was reduced. Combination with a strong enzyme inducer should be avoided in future trials to prevent further reduction of panobinostat exposure.
Collapse
Affiliation(s)
- Song Mu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | | | | | | | | | | | - Rong Lin
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Edward Waldron
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | |
Collapse
|
26
|
Mixed galactolipid anomers accentuate apoptosis of multiple myeloma cells by inducing DNA damage. Carbohydr Res 2015; 408:114-8. [DOI: 10.1016/j.carres.2014.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/15/2014] [Accepted: 11/21/2014] [Indexed: 11/20/2022]
|
27
|
Wang X, Zhang M. Synergistic effects of valproic acid and arsenic trioxide on RPMI8226 cells in vitro and the possible underlying mechanisms. Mol Med Rep 2015; 12:1449-56. [PMID: 25815518 DOI: 10.3892/mmr.2015.3519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 02/27/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the synergistic effects of valproic acid (VPA) and arsenic trioxide (ATO) on the proliferation of RPMI8226 cells and the possible underlying mechanisms. Cell apoptosis was assessed by flow cytometry. The mRNA expression levels were analyzed by semi-quantitative polymerase chain reaction, and the protein expression levels were analyzed by western blotting. The histone acetylation and methylation states of the gene promoters were detected using a chromatin immunoprecipitation technique. The apoptotic rates of the RPMI8226 cells in the combined drug groups were significantly increased compared with those of the single drug groups (P<0.05). The mRNA and protein expression levels of Bcl-2 and the expression levels of HDAC1 mRNA and H3K9me2 protein decreased significantly in the combined groups compared with the single drug groups. The mRNA and protein expression levels of Bax, Caspase 8, Caspase 9 and LSD1, and the protein expression of acetylated H3 increased significantly in the combination groups compared with the single drug groups. Histone methylation and acetylation of the Bcl-2 and bax gene promoters were increased in the combination groups compared with the single drug groups. VPA and ATO had synergistic effects on the proliferation of RPMI8226 cells, which may have been associated with the decreased expression of Bcl-2 and the increased expression levels of Bcl-2-associated X protein, Caspase 8 and Caspase 9. Therefore, the expression levels of the Bcl-2 gene family may have been regulated by the levels of gene promoter methylation and acetylation.
Collapse
Affiliation(s)
- Xiaoning Wang
- Department of Hematology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mei Zhang
- Department of Hematology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
28
|
Hojjat-Farsangi M. Novel and emerging targeted-based cancer therapy agents and methods. Tumour Biol 2015; 36:543-56. [PMID: 25663495 DOI: 10.1007/s13277-015-3184-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/27/2015] [Indexed: 02/06/2023] Open
Abstract
After several decades of uncovering the cancer features and following the improvement of therapeutic agents, however cancer remains as one of the major reasons of mortality. Chemotherapy is one of the main treatment options and has significantly improved the overall survival of cancer patients, but chemotherapeutic agents are highly toxic for normal cells. Therefore, there is a great unmet medical need to develop new therapeutic principles and agents. Targeted-based cancer therapy (TBCT) agents and methods have revolutionized the cancer treatment efficacy. Monoclonal antibodies (mAbs) and small molecule inhibitors (SMIs) are among the most effective agents of TBCT. These drugs have improved the prognosis and survival of cancer patients; however, the therapeutic resistance has subdued the effects. Several mechanisms lead to drug resistance such as mutations in the drug targets, activation of compensatory pathways, and intrinsic or acquired resistance of cancer stem cells. Therefore, new modalities, improving current generation of inhibitors and mAbs, and optimizing the combinational therapy regimens are necessary to decrease the current obstacles in front of TBCT. Moreover, the success of new TBCT agents such as mAbs, SMIs, and immunomodulatory agents has sparked further therapeutic modalities with novel targets to inhibit. Due to the lack of cumulative information describing different agents and methods of TBCT, this review focuses on the most important agents and methods of TBCT that are currently under investigation.
Collapse
Affiliation(s)
- Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, SE-171 76, Stockholm, Sweden,
| |
Collapse
|
29
|
Surati M, Valla K, Shah KS, Panjic EH, Lonial S. Panobinostat for the treatment of multiple myeloma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.999665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
30
|
Clinical use of proteasome inhibitors in the treatment of multiple myeloma. Pharmaceuticals (Basel) 2014; 8:1-20. [PMID: 25545164 PMCID: PMC4381198 DOI: 10.3390/ph8010001] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 12/04/2014] [Indexed: 01/08/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy characterized by the clonal proliferation of neoplastic plasma cells. The use of proteasome inhibitors in the treatment of MM has led to significant improvements in outcomes. This article reviews data on the use of the two approved proteasome inhibitors (bortezomib and carlfilzomib), as well as newer agents under development. Emphasis is placed on the clinical use of proteasome inhibitors, including management of side effects and combination with other agents.
Collapse
|
31
|
Libby EN, Becker PS, Burwick N, Green DJ, Holmberg L, Bensinger WI. Panobinostat: a review of trial results and future prospects in multiple myeloma. Expert Rev Hematol 2014; 8:9-18. [PMID: 25410127 DOI: 10.1586/17474086.2015.983065] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Multiple myeloma is an incurable often devastating disease that is responsible for 1-2% of all cancers. Multiple myeloma is the second most common hematologic malignancy. Over the past two decades, advances in therapy have doubled life expectancy. Unfortunately, all patients ultimately relapse. Novel agents (immunomodulatory drugs and proteasome inhibitors) have changed the outlook for patients, but further breakthroughs are needed. Epigenetic treatments offer potential for advancing therapy by modifying oncogene responses. The acetylation status of various proteins can affect the availability of chromatin for transcription. This response may be modulated epigenetically to advantage using histone deacetylase inhibitors like panobinostat.
Collapse
Affiliation(s)
- Edward N Libby
- University of Washington School of Medicine - Medical Oncology, 825 Eastlake Ave E, Seattle, WA 98109, USA
| | | | | | | | | | | |
Collapse
|
32
|
Stewart MD, Ramani VC, Sanderson RD. Shed syndecan-1 translocates to the nucleus of cells delivering growth factors and inhibiting histone acetylation: a novel mechanism of tumor-host cross-talk. J Biol Chem 2014; 290:941-9. [PMID: 25404732 DOI: 10.1074/jbc.m114.608455] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The heparan sulfate proteoglycan syndecan-1 is proteolytically shed from the surface of multiple myeloma cells and is abundant in the bone marrow microenvironment where it promotes tumor growth, angiogenesis, and metastasis. In this study, we demonstrate for the first time that shed syndecan-1 present in the medium conditioned by tumor cells is taken up by bone marrow-derived stromal cells and transported to the nucleus. Translocation of shed syndecan-1 (sSDC1) to the nucleus was blocked by addition of exogenous heparin or heparan sulfate, pretreatment of conditioned medium with heparinase III, or growth of cells in sodium chlorate, indicating that sulfated heparan sulfate chains are required for nuclear translocation. Interestingly, cargo bound to sSDC1 heparan sulfate chains (i.e. hepatocyte growth factor) was transported to the nucleus along with sSDC1, and removal of heparan sulfate-bound cargo from sSDC1 abolished its translocation to the nucleus. Once in the nucleus, sSDC1 binds to the histone acetyltransferase enzyme p300, and histone acetyltransferase activity and histone acetylation are diminished. These findings reveal a novel function for shed syndecan-1 in mediating tumor-host cross-talk by shuttling growth factors to the nucleus and by altering histone acetylation in host cells. In addition, this work has broad implications beyond myeloma because shed syndecan-1 is present in high levels in many tumor types as well as in other disease states.
Collapse
Affiliation(s)
| | | | - Ralph D Sanderson
- From the Department of Pathology, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
33
|
Abdi J, Chen G, Chang H. Drug resistance in multiple myeloma: latest findings and new concepts on molecular mechanisms. Oncotarget 2014; 4:2186-207. [PMID: 24327604 PMCID: PMC3926819 DOI: 10.18632/oncotarget.1497] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In the era of new and mostly effective therapeutic protocols, multiple myeloma still tends to be a hard-to-treat hematologic cancer. This hallmark of the disease is in fact a sequel to drug resistant phenotypes persisting initially or emerging in the course of treatment. Furthermore, the heterogeneous nature of multiple myeloma makes treating patients with the same drug challenging because finding a drugable oncogenic process common to all patients is not yet feasible, while our current knowledge of genetic/epigenetic basis of multiple myeloma pathogenesis is outstanding. Nonetheless, bone marrow microenvironment components are well known as playing critical roles in myeloma tumor cell survival and environment-mediated drug resistance happening most possibly in all myeloma patients. Generally speaking, however; real mechanisms underlying drug resistance in multiple myeloma are not completely understood. The present review will discuss the latest findings and concepts in this regard. It reviews the association of important chromosomal translocations, oncogenes (e.g. TP53) mutations and deranged signaling pathways (e.g. NFκB) with drug response in clinical and experimental investigations. It will also highlight how bone marrow microenvironment signals (Wnt, Notch) and myeloma cancer stem cells could contribute to drug resistance in multiple myeloma.
Collapse
Affiliation(s)
- Jahangir Abdi
- Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
34
|
Tough DF, Lewis HD, Rioja I, Lindon MJ, Prinjha RK. Epigenetic pathway targets for the treatment of disease: accelerating progress in the development of pharmacological tools: IUPHAR Review 11. Br J Pharmacol 2014; 171:4981-5010. [PMID: 25060293 PMCID: PMC4253452 DOI: 10.1111/bph.12848] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/22/2014] [Accepted: 06/13/2014] [Indexed: 02/06/2023] Open
Abstract
The properties of a cell are determined both genetically by the DNA sequence of its genes and epigenetically through processes that regulate the pattern, timing and magnitude of expression of its genes. While the genetic basis of disease has been a topic of intense study for decades, recent years have seen a dramatic increase in the understanding of epigenetic regulatory mechanisms and a growing appreciation that epigenetic misregulation makes a significant contribution to human disease. Several large protein families have been identified that act in different ways to control the expression of genes through epigenetic mechanisms. Many of these protein families are finally proving tractable for the development of small molecules that modulate their function and represent new target classes for drug discovery. Here, we provide an overview of some of the key epigenetic regulatory proteins and discuss progress towards the development of pharmacological tools for use in research and therapy.
Collapse
Affiliation(s)
- David F Tough
- Immuno-Inflammation Therapy Area, GlaxoSmithKline R&D, Medicines Research Centre, Epinova DPU, Stevenage, UK
| | | | | | | | | |
Collapse
|
35
|
Bose P, Dai Y, Grant S. Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights. Pharmacol Ther 2014; 143:323-36. [PMID: 24769080 PMCID: PMC4117710 DOI: 10.1016/j.pharmthera.2014.04.004] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 04/10/2014] [Indexed: 02/05/2023]
Abstract
Initially regarded as "epigenetic modifiers" acting predominantly through chromatin remodeling via histone acetylation, HDACIs, alternatively referred to as lysine deacetylase or simply deacetylase inhibitors, have since been recognized to exert multiple cytotoxic actions in cancer cells, often through acetylation of non-histone proteins. Some well-recognized mechanisms of HDACI lethality include, in addition to relaxation of DNA and de-repression of gene transcription, interference with chaperone protein function, free radical generation, induction of DNA damage, up-regulation of endogenous inhibitors of cell cycle progression, e.g., p21, and promotion of apoptosis. Intriguingly, this class of agents is relatively selective for transformed cells, at least in pre-clinical studies. In recent years, additional mechanisms of action of these agents have been uncovered. For example, HDACIs interfere with multiple DNA repair processes, as well as disrupt cell cycle checkpoints, critical to the maintenance of genomic integrity in the face of diverse genotoxic insults. Despite their pre-clinical potential, the clinical use of HDACIs remains restricted to certain subsets of T-cell lymphoma. Currently, it appears likely that the ultimate role of these agents will lie in rational combinations, only a few of which have been pursued in the clinic to date. This review focuses on relatively recently identified mechanisms of action of HDACIs, with particular emphasis on those that relate to the DNA damage response (DDR), and discusses synergistic strategies combining HDACIs with several novel targeted agents that disrupt the DDR or antagonize anti-apoptotic proteins that could have implications for the future use of HDACIs in patients with cancer.
Collapse
Affiliation(s)
- Prithviraj Bose
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yun Dai
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA; Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA; Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA; Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
36
|
Oriol A, Motlló C. Nuevos fármacos en el tratamiento del mieloma múltiple. Med Clin (Barc) 2014; 143:268-74. [DOI: 10.1016/j.medcli.2013.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/30/2013] [Accepted: 10/03/2013] [Indexed: 10/25/2022]
|
37
|
Ludwig H, Sonneveld P, Davies F, Bladé J, Boccadoro M, Cavo M, Morgan G, de la Rubia J, Delforge M, Dimopoulos M, Einsele H, Facon T, Goldschmidt H, Moreau P, Nahi H, Plesner T, San-Miguel J, Hajek R, Sondergeld P, Palumbo A. European perspective on multiple myeloma treatment strategies in 2014. Oncologist 2014; 19:829-44. [PMID: 25063227 PMCID: PMC4122482 DOI: 10.1634/theoncologist.2014-0042] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
The treatment of multiple myeloma has undergone significant changes and has resulted in the achievement of molecular remissions, the prolongation of remission duration, and extended survival becoming realistic goals, with a cure being possible in a small but growing number of patients. In addition, nowadays it is possible to categorize patients more precisely into different risk groups, thus allowing the evaluation of therapies in different settings and enabling a better comparison of results across trials. Here, we review the evidence from clinical studies, which forms the basis for our recommendations for the management of patients with myeloma. Treatment approaches depend on "fitness," with chronological age still being an important discriminator for selecting therapy. In younger, fit patients, a short three drug-based induction treatment followed by autologous stem cell transplantation (ASCT) remains the preferred option. Consolidation and maintenance therapy are attractive strategies not yet approved by the European Medicines Agency, and a decision regarding post-ASCT therapy should only be made after detailed discussion of the pros and cons with the individual patient. Two- and three-drug combinations are recommended for patients not eligible for transplantation. Treatment should be administered for at least nine cycles, although different durations of initial therapy have only rarely been compared so far. Comorbidity and frailty should be thoroughly assessed in elderly patients, and treatment must be adapted to individual needs, carefully selecting appropriate drugs and doses. A substantial number of new drugs and novel drug classes in early clinical development have shown promising activity. Their introduction into clinical practice will most likely further improve treatment results.
Collapse
Affiliation(s)
- Heinz Ludwig
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Pieter Sonneveld
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Faith Davies
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Joan Bladé
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Mario Boccadoro
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Michele Cavo
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Gareth Morgan
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Javier de la Rubia
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Michel Delforge
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Meletios Dimopoulos
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Hermann Einsele
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Thierry Facon
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Hartmut Goldschmidt
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Philippe Moreau
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Hareth Nahi
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Torben Plesner
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Jesús San-Miguel
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Roman Hajek
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Pia Sondergeld
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| | - Antonio Palumbo
- Department of Oncology, Hematology and Palliative Care, Wilhelminenspital, Vienna, Austria; Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom; Department of Hematology, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clinic, Barcelona, Spain; Divisione di Ematologia dell'Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Ospedale Molinette, Turin, Italy; Institute of Hematology and Medical Oncology, Seragnoli, Bologna, Italy; Haemato-Oncology Unit, Royal Marsden Hospital, Surrey, United Kingdom; Hematology Service, University Hospital La Fe, Valencia, Spain; Department of Hematology, University Hospital, Leuven, Belgium; Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece; Universitätsklinik Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany; Service d'Hématologie, Centre Hospitalier Universitaire, Lille, France; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany; University Hospital, Nantes, France; Karolinska Institute, Stockholm, Sweden; Department of Hematology, Center Lillebaelt, University of Southern Denmark, Odense, Denmark; Clinica Universidad de Navarra, Centro Investigaciones Medicas Aplicada, Pamplona, Spain; Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic; University of Giessen, Giessen, Germany
| |
Collapse
|
38
|
Gkotzamanidou M, Sfikakis PP, Kyrtopoulos SA, Bamia C, Dimopoulos MA, Souliotis VL. Chromatin structure, transcriptional activity and DNA repair efficiency affect the outcome of chemotherapy in multiple myeloma. Br J Cancer 2014; 111:1293-304. [PMID: 25051404 PMCID: PMC4183844 DOI: 10.1038/bjc.2014.410] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/15/2014] [Accepted: 06/30/2014] [Indexed: 01/20/2023] Open
Abstract
Background: Melphalan is one of the most active chemotherapeutic agents in the treatment of multiple myeloma (MM). However, the mechanism underlying differential patient responses to melphalan therapy is unknown. Methods: Chromatin structure, transcriptional activity and DNA damage response signals were examined following ex vivo treatment with melphalan of both malignant bone marrow plasma cells (BMPCs) and peripheral blood mononuclear cells (PBMCs) of MM patients, responders (n=57) or non-responders (n=28) to melphalan therapy. PBMCs from healthy controls (n=25) were also included in the study. Results: In both BMPCs and PBMCs, the local chromatin looseness, transcriptional activity and repair efficiency of the transcribed strand (TS) were significantly higher in non-responders than in responders and lowest in healthy controls (all P<0.05). Moreover, we found that melphalan-induced apoptosis inversely correlated with the repair efficiency of the TS, with the duration of the inhibition of mRNA synthesis, phosphorylation of p53 at serine 15 and apoptosis rates being higher in responders than in non-responders (all P<0.001). Conclusions: Our findings provide a mechanistic basis for the link between DNA repair efficiency and response to melphalan therapy. Interestingly, the observation of these phenomena in PBMCs provides a novel approach for the prediction of response to anti-myeloma therapy.
Collapse
Affiliation(s)
- M Gkotzamanidou
- 1] Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA [2] Department of Clinical Therapeutics, University of Athens School of Medicine, 11528 Athens, Greece
| | - P P Sfikakis
- First Department of Propedeutic Medicine, University of Athens School of Medicine, 11527 Athens, Greece
| | - S A Kyrtopoulos
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - C Bamia
- Department of Hygiene, Epidemiology and Medical Statistics, University of Athens School of Medicine, 11527 Athens, Greece
| | - M A Dimopoulos
- Department of Clinical Therapeutics, University of Athens School of Medicine, 11528 Athens, Greece
| | - V L Souliotis
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| |
Collapse
|
39
|
Abstract
Multiple myeloma (MM) is a clonal B-cell malignancy characterized by aberrant expansion of plasma cells within bone marrow and extramedullary sites. It is one of the most common haematological malignancies; it accounts for 1.4% of all tumours and is responsible for 2% of cancer-related mortality. Over the last decades, the paradigm of MM therapy has changed dramatically - from the conventional combination of oral melphatan + prednisone, high-dose chemotherapy with stem cell (ASCT) support for younger patients to the present paradigm with the use of one (or more) of 3 major new targeted agents - the first-in class proteasome inhibitor bortezomib, the immunomodulatory drug thalidomide, and its more potent derivative lenalidomide. Their use as a part of initial therapy is associated with high overall response rates as well as high rates of complete response (CR), both for elderly patients unable to undergo ASCT and for younger patients treated prior to ASCT. Altogether, the advent of novel agents has resulted in a 50% improvement in median survival. Moreover, the development of new drug classes based on preclinical rationale and the introduction of next-generation agents are likely to further expand treatment options and improve outcomes for especially relapsed MM. This review highlights important historic landmarks as well as more recent events that have played an important role in the evolution of myeloma targeted therapy.
Collapse
|
40
|
Castelli R, Orofino N, Losurdo A, Gualtierotti R, Cugno M. Choosing treatment options for patients with relapsed/refractory multiple myeloma. Expert Rev Anticancer Ther 2013; 14:199-215. [PMID: 24329153 DOI: 10.1586/14737140.2014.863153] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma cell disorder that is still incurable using conventional treatments. Over the last decade, advances in front-line therapy have led to an increase in survival, but there are still some doubts in the case of relapsed/refractory disease. We searched the PubMed database for articles on treatment options for patients with relapsed/refractory MM published between 1996 and 2013. These treatments included hematopoietic cell transplantation (HCT), rechallenges using previous chemotherapy regimens, and trials of new regimens. The introduction of new agents such as the immunomodulatory drugs (IMIDs) thalidomide and lenalidomide, and the first-in-its-class proteasome inhibitor bortezomib, has greatly improved clinical outcomes in patients with relapsed/refractory MM, but not all patients respond and those that do may eventually relapse or become refractory to treatment. The challenge is therefore to select the optimal treatment for each patient by balancing efficacy and toxicity. To do this, it is necessary to consider disease-related factors, such as the quality and duration of responses to previous therapies, and the aggressiveness of the relapse, and patient-related factors such as age, comorbidities, performance status, pre-existing toxicities and cytogenetic patterns. The message from the trials reviewed in this article is that the new agents may be used to re-treat relapsed/refractory disease, and that the sequencing of their administration should be modulated on the basis of the various disease and patient-related factors. Moreover, our understanding of the pharmacology and molecular action of the new drugs will contribute to the possibility of developing tailored treatment.
Collapse
Affiliation(s)
- Roberto Castelli
- Department of Pathophysiology and Transplantation, Internal Medicine, University of Milan, Milan, Italy
| | | | | | | | | |
Collapse
|
41
|
Sripayap P, Nagai T, Hatano K, Kikuchi J, Furukawa Y, Ozawa K. Romidepsin overcomes cell adhesion-mediated drug resistance in multiple myeloma cells. Acta Haematol 2013; 132:1-4. [PMID: 24356056 DOI: 10.1159/000357213] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 11/10/2013] [Indexed: 12/12/2022]
|
42
|
|
43
|
Bruzzese F, Pucci B, Milone MR, Ciardiello C, Franco R, Chianese MI, Rocco M, Di Gennaro E, Leone A, Luciano A, Arra C, Santini D, Caraglia M, Budillon A. Panobinostat synergizes with zoledronic acid in prostate cancer and multiple myeloma models by increasing ROS and modulating mevalonate and p38-MAPK pathways. Cell Death Dis 2013; 4:e878. [PMID: 24157872 PMCID: PMC3920938 DOI: 10.1038/cddis.2013.406] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/11/2013] [Accepted: 09/13/2013] [Indexed: 11/23/2022]
Abstract
Patients with advanced prostate cancer (PCa) and multiple myeloma (MM) have limited long-term responses to available therapies. The histone deacetylase inhibitor panobinostat has shown significant preclinical and clinical anticancer activity in both hematological and solid malignancies and is currently in phase III trials for relapsed MM. Bisphosphonates (BPs), such as zoledronic acid (ZOL), inhibit osteoclast-mediated bone resorption and are indicated for the treatment of bone metastasis. BPs, including ZOL, have also shown anticancer activity in several preclinical and clinical studies. In the present report, we found a potent synergistic antiproliferative effect of panobinostat/ZOL treatment in three PCa and three MM cell lines as well as in a PCa ZOL-resistant subline, independently of p53/KRAS status, androgen dependency, or the schedule of administration. The synergistic effect was also observed in an anchorage-independent agar assay in both ZOL-sensitive and ZOL-resistant cells and was confirmed in vivo in a PCa xenograft model. The co-administration of the antioxidant N-acetyl-L-cysteine blocked the increased reactive oxygen species generation and apoptosis observed in the combination setting compared with control or single-agent treatments, suggesting that oxidative injury plays a functional role in the synergism. Proapoptotic synergy was also partially antagonized by the addition of geranyl-geraniol, which bypasses the inhibition of farnesylpyrophosphate synthase by ZOL in the mevalonate pathway, supporting the involvement of this pathway in the synergy. Finally, at the molecular level, the inhibition of basal and ZOL-induced activation of p38-MAPK by panobinostat in sensitive and ZOL-resistant cells and in tumor xenografts could explain, at least in part, the observed synergism.
Collapse
Affiliation(s)
- F Bruzzese
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale'-IRCCS, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Castelli R, Gualtierotti R, Orofino N, Losurdo A, Gandolfi S, Cugno M. Current and emerging treatment options for patients with relapsed myeloma. Clin Med Insights Oncol 2013; 7:209-19. [PMID: 24179412 PMCID: PMC3813615 DOI: 10.4137/cmo.s8014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma (MM) is a neoplastic disorder. It results from proliferation of clonal plasma cells in bone marrow with production of monoclonal proteins, which are detectable in serum or urine. MM is clinically characterized by destructive bone lesions, anemia, hypercalcemia and renal insufficiency. Its prognosis is severe, with a median survival after diagnosis of approximately 3 years due to frequent relapses. Treatments for patients with relapsed/refractory MM include hematopoietic cell transplantation, a rechallenge using a previous chemotherapy regimen or a trial of a new regimen. The introduction of new drugs such as thalidomide, lenalidomide and bortezomib has markedly improved MM outcomes. When relapse occurs, the clinician's challenge is to select the optimal treatment for each patient while balancing efficacy and toxicity. Patients with indolent relapse can be first treated with a 2-drug or a 3-drug combination. Patients with more aggressive relapse often require therapy with a combination of multiple active agents. Autologous stem cell transplantation should be considered as salvage therapy at first relapse for patients who have cryopreserved stem cells early in the disease course. The aim of this review is to provide an overview on the pharmacological and molecular action of treatments used for patients with relapsed/refractory multiple myeloma.
Collapse
Affiliation(s)
- Roberto Castelli
- Internal Medicine, Department of Pathophysiology and Transplantation, University of Milan, and Department of Medicine, IRCCS Fondazione Ca’ Granda Policlinico, Milan, Italy
| | - Roberta Gualtierotti
- Internal Medicine, Department of Pathophysiology and Transplantation, University of Milan, and Department of Medicine, IRCCS Fondazione Ca’ Granda Policlinico, Milan, Italy
| | - Nicola Orofino
- Internal Medicine, Department of Pathophysiology and Transplantation, University of Milan, and Department of Medicine, IRCCS Fondazione Ca’ Granda Policlinico, Milan, Italy
| | - Agnese Losurdo
- Internal Medicine, Department of Pathophysiology and Transplantation, University of Milan, and Department of Medicine, IRCCS Fondazione Ca’ Granda Policlinico, Milan, Italy
| | - Sara Gandolfi
- Internal Medicine, Department of Pathophysiology and Transplantation, University of Milan, and Department of Medicine, IRCCS Fondazione Ca’ Granda Policlinico, Milan, Italy
| | - Massimo Cugno
- Internal Medicine, Department of Pathophysiology and Transplantation, University of Milan, and Department of Medicine, IRCCS Fondazione Ca’ Granda Policlinico, Milan, Italy
| |
Collapse
|