1
|
Heppt MV, Wessely A, Hornig E, Kammerbauer C, Graf SA, Besch R, French LE, Matthies A, Kuphal S, Kappelmann-Fenzl M, Bosserhoff AK, Berking C. HDAC2 Is Involved in the Regulation of BRN3A in Melanocytes and Melanoma. Int J Mol Sci 2022; 23:ijms23020849. [PMID: 35055045 PMCID: PMC8778714 DOI: 10.3390/ijms23020849] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
The neural crest transcription factor BRN3A is essential for the proliferation and survival of melanoma cells. It is frequently expressed in melanoma but not in normal melanocytes or benign nevi. The mechanisms underlying the aberrant expression of BRN3A are unknown. Here, we investigated the epigenetic regulation of BRN3A in melanocytes and melanoma cell lines treated with DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC) inhibitors. DNMT and HAT inhibition did not significantly alter BRN3A expression levels, whereas panHDAC inhibition by trichostatin A led to increased expression. Treatment with the isoform-specific HDAC inhibitor mocetinostat, but not with PCI-34051, also increased BRN3A expression levels, suggesting that class I HDACs HDAC1, HDAC2, and HDAC3, and class IV HDAC11, were involved in the regulation of BRN3A expression. Transient silencing of HDACs 1, 2, 3, and 11 by siRNAs revealed that, specifically, HDAC2 inhibition was able to increase BRN3A expression. ChIP-Seq analysis uncovered that HDAC2 inhibition specifically increased H3K27ac levels at a distal enhancer region of the BRN3A gene. Altogether, our data suggest that HDAC2 is a key epigenetic regulator of BRN3A in melanocytes and melanoma cells. These results highlight the importance of epigenetic mechanisms in regulating melanoma oncogenes.
Collapse
Affiliation(s)
- Markus V. Heppt
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.W.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-85-35747
| | - Anja Wessely
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.W.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
| | - Eva Hornig
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337 Munich, Germany; (E.H.); (C.K.); (S.A.G.); (R.B.); (L.E.F.)
| | - Claudia Kammerbauer
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337 Munich, Germany; (E.H.); (C.K.); (S.A.G.); (R.B.); (L.E.F.)
| | - Saskia A. Graf
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337 Munich, Germany; (E.H.); (C.K.); (S.A.G.); (R.B.); (L.E.F.)
| | - Robert Besch
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337 Munich, Germany; (E.H.); (C.K.); (S.A.G.); (R.B.); (L.E.F.)
| | - Lars E. French
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337 Munich, Germany; (E.H.); (C.K.); (S.A.G.); (R.B.); (L.E.F.)
| | - Alexander Matthies
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.M.); (S.K.); (A.K.B.)
| | - Silke Kuphal
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.M.); (S.K.); (A.K.B.)
| | | | - Anja K. Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.M.); (S.K.); (A.K.B.)
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (A.W.); (C.B.)
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
| |
Collapse
|
2
|
Sangwan R, Rajan R, Mandal PK. HDAC as onco target: Reviewing the synthetic approaches with SAR study of their inhibitors. Eur J Med Chem 2018; 158:620-706. [DOI: 10.1016/j.ejmech.2018.08.073] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/09/2018] [Accepted: 08/26/2018] [Indexed: 02/06/2023]
|
3
|
Khan SA, Pruitt SL, Xuan L, Makris U, Gerber DE. How does autoimmune disease impact treatment and outcomes among patients with lung cancer? A national SEER-Medicare analysis. Lung Cancer 2017; 115:97-102. [PMID: 29290269 DOI: 10.1016/j.lungcan.2017.11.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/15/2017] [Accepted: 11/23/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The advent of cancer immunotherapy has made autoimmune disease in oncology populations clinically important. We analyzed the association of autoimmune disease with treatment and outcomes among lung cancer patients. METHODS Using linked Surveillance Epidemiology and End Results (SEER)-Medicare data, we identified lung cancer patients diagnosed between 1992 and 2009 with autoimmune diseases. We recorded number and timing of autoimmune disease diagnoses, lung cancer treatment, and markers of healthcare utilization including emergency department visits, hospitalizations, and outpatient visits. To account for potential lead-time bias, we used a matched case-control analysis wherein living and deceased patients were matched on survival time. We performed unadjusted and multivariable adjusted logistic regressions separately by cancer stage for all-cause and lung cancer-specific mortality. RESULTS Among 172,285 lung cancer patients, 23,084 (13.4%) had ≥1 autoimmune disease at any time. Overall, 10,927 patients (6.3%) had one autoimmune disease before cancer diagnosis; 9338 (5.4%) had two or more before cancer diagnosis; and 2819 (1.6%) had one or more after cancer diagnosis. Healthcare utilization was higher in the autoimmune disease population. Lung cancer treatment patterns were similar among patients with and without autoimmune disease and there was no significant association with mortality. CONCLUSIONS Among patients with lung cancer, autoimmune disease does not influence treatment patterns and is not associated with mortality.
Collapse
Affiliation(s)
- Saad A Khan
- Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, United States; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, United States
| | - Sandi L Pruitt
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, United States; Department of Clinical Sciences, UT Southwestern Medical Center, Dallas, United States
| | - Lei Xuan
- Department of Clinical Sciences, UT Southwestern Medical Center, Dallas, United States
| | - Una Makris
- Department of Clinical Sciences, UT Southwestern Medical Center, Dallas, United States; Department of Internal Medicine, Division of Rheumatic Diseases, UT Southwestern Medical Center, Dallas, United States; VA North Texas Health Care System, Medical Service, Dallas, TX, United States
| | - David E Gerber
- Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, United States; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, United States; Department of Clinical Sciences, UT Southwestern Medical Center, Dallas, United States.
| |
Collapse
|
4
|
Histone Deacetylase Inhibitors as Anticancer Drugs. Int J Mol Sci 2017; 18:ijms18071414. [PMID: 28671573 PMCID: PMC5535906 DOI: 10.3390/ijms18071414] [Citation(s) in RCA: 814] [Impact Index Per Article: 116.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/11/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
Carcinogenesis cannot be explained only by genetic alterations, but also involves epigenetic processes. Modification of histones by acetylation plays a key role in epigenetic regulation of gene expression and is controlled by the balance between histone deacetylases (HDAC) and histone acetyltransferases (HAT). HDAC inhibitors induce cancer cell cycle arrest, differentiation and cell death, reduce angiogenesis and modulate immune response. Mechanisms of anticancer effects of HDAC inhibitors are not uniform; they may be different and depend on the cancer type, HDAC inhibitors, doses, etc. HDAC inhibitors seem to be promising anti-cancer drugs particularly in the combination with other anti-cancer drugs and/or radiotherapy. HDAC inhibitors vorinostat, romidepsin and belinostat have been approved for some T-cell lymphoma and panobinostat for multiple myeloma. Other HDAC inhibitors are in clinical trials for the treatment of hematological and solid malignancies. The results of such studies are promising but further larger studies are needed. Because of the reversibility of epigenetic changes during cancer development, the potency of epigenetic therapies seems to be of great importance. Here, we summarize the data on different classes of HDAC inhibitors, mechanisms of their actions and discuss novel results of preclinical and clinical studies, including the combination with other therapeutic modalities.
Collapse
|
5
|
Chhabra S. Novel Proteasome Inhibitors and Histone Deacetylase Inhibitors: Progress in Myeloma Therapeutics. Pharmaceuticals (Basel) 2017; 10:E40. [PMID: 28398261 PMCID: PMC5490397 DOI: 10.3390/ph10020040] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/28/2017] [Accepted: 04/04/2017] [Indexed: 01/01/2023] Open
Abstract
The unfolded protein response is responsible for the detection of misfolded proteins and the coordination of their disposal and is necessary to maintain the cellular homoeostasis. Multiple myeloma cells secrete large amounts of immunoglobulins, proteins that need to be correctly folded by the chaperone system. If this process fails, the misfolded proteins have to be eliminated by the two main garbage-disposal systems of the cell: proteasome and aggresome. The blockade of either of these systems will result in accumulation of immunoglobulins and other toxic proteins in the cytoplasm and cell death. The simultaneous inhibition of the proteasome, by proteasome inhibitors (PIs) and the aggresome, by histone deacetylase inhibitors (HDACi) results in a synergistic increase in cytotoxicity in myeloma cell lines. This review provides an overview of mechanisms of action of second-generation PIs and HDACi in multiple myeloma (MM), the clinical results currently observed with these agents and assesses the potential therapeutic impact of the different agents in the two classes. The second-generation PIs offer benefits in terms of increased efficacy, reduced neurotoxicity as off-target effect and may overcome resistance to bortezomib because of their different chemical structure, mechanism of action and biological properties. HDACi with anti-myeloma activity in clinical development discussed in this review include vorinostat, panobinostat and selective HDAC6 inhibitor, ricolinostat.
Collapse
Affiliation(s)
- Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, 9200 W Wisconsin Ave, Milwaukee, WI 53226, USA.
| |
Collapse
|
6
|
Matthews GM, de Matos Simoes R, Dhimolea E, Sheffer M, Gandolfi S, Dashevsky O, Sorrell JD, Mitsiades CS. NF-κB dysregulation in multiple myeloma. Semin Cancer Biol 2016; 39:68-76. [PMID: 27544796 DOI: 10.1016/j.semcancer.2016.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 12/29/2022]
Abstract
The nuclear factor-κB (NF-κB) transcription factor family plays critical roles in the pathophysiology of hematologic neoplasias, including multiple myeloma. The current review examines the roles that this transcription factor system plays in multiple myeloma cells and the nonmalignant accessory cells of the local microenvironment; as well as the evidence indicating that a large proportion of myeloma patients harbor genomic lesions which perturb diverse genes regulating the activity of NF-κB. This article also discusses the therapeutic targeting of the NF-κB pathway using proteasome inhibitors, a pharmacological class that has become a cornerstone in the therapeutic management of myeloma; and reviews some of the future challenges and opportunities for NF-κB-related research in myeloma.
Collapse
Affiliation(s)
- Geoffrey M Matthews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Ricardo de Matos Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Eugen Dhimolea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Sara Gandolfi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Jeffrey D Sorrell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States.
| |
Collapse
|
7
|
Haery L, Thompson RC, Gilmore TD. Histone acetyltransferases and histone deacetylases in B- and T-cell development, physiology and malignancy. Genes Cancer 2015; 6:184-213. [PMID: 26124919 PMCID: PMC4482241 DOI: 10.18632/genesandcancer.65] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 05/12/2015] [Indexed: 12/31/2022] Open
Abstract
The development of B and T cells from hematopoietic precursors and the regulation of the functions of these immune cells are complex processes that involve highly regulated signaling pathways and transcriptional control. The signaling pathways and gene expression patterns that give rise to these developmental processes are coordinated, in part, by two opposing classes of broad-based enzymatic regulators: histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs and HDACs can modulate gene transcription by altering histone acetylation to modify chromatin structure, and by regulating the activity of non-histone substrates, including an array of immune-cell transcription factors. In addition to their role in normal B and T cells, dysregulation of HAT and HDAC activity is associated with a variety of B- and T-cell malignancies. In this review, we describe the roles of HATs and HDACs in normal B- and T-cell physiology, describe mutations and dysregulation of HATs and HDACs that are implicated lymphoma and leukemia, and discuss HAT and HDAC inhibitors that have been explored as treatment options for leukemias and lymphomas.
Collapse
Affiliation(s)
- Leila Haery
- Department of Biology, Boston University, Boston, MA, USA
| | | | | |
Collapse
|
8
|
Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 2015; 20:3898-941. [PMID: 25738536 PMCID: PMC4372801 DOI: 10.3390/molecules20033898] [Citation(s) in RCA: 480] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 02/13/2015] [Accepted: 02/15/2015] [Indexed: 02/04/2023] Open
Abstract
Histone dacetylases (HDACs) are a group of enzymes that remove acetyl groups from histones and regulate expression of tumor suppressor genes. They are implicated in many human diseases, especially cancer, making them a promising therapeutic target for treatment of the latter by developing a wide variety of inhibitors. HDAC inhibitors interfere with HDAC activity and regulate biological events, such as cell cycle, differentiation and apoptosis in cancer cells. As a result, HDAC inhibitor-based therapies have gained much attention for cancer treatment. To date, the FDA has approved three HDAC inhibitors for cutaneous/peripheral T-cell lymphoma and many more HDAC inhibitors are in different stages of clinical development for the treatment of hematological malignancies as well as solid tumors. In the intensifying efforts to discover new, hopefully more therapeutically efficacious HDAC inhibitors, molecular modeling-based rational drug design has played an important role in identifying potential inhibitors that vary in molecular structures and properties. In this review, we summarize four major structural classes of HDAC inhibitors that are in clinical trials and different computer modeling tools available for their structural modifications as a guide to discover additional HDAC inhibitors with greater therapeutic utility.
Collapse
Affiliation(s)
- Madhusoodanan Mottamal
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Shilong Zheng
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Tien L Huang
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
- College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Guangdi Wang
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| |
Collapse
|
9
|
Yang B, Yu D, Liu J, Yang K, Wu G, Liu H. Antitumor activity of SAHA, a novel histone deacetylase inhibitor, against murine B cell lymphoma A20 cells in vitro and in vivo. Tumour Biol 2015; 36:5051-61. [PMID: 25649979 DOI: 10.1007/s13277-015-3156-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/26/2015] [Indexed: 12/31/2022] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA; vorinostat), the second generation of histone deacetylase (HDAC) inhibitor, has been approved for the treatment of cutaneous manifestations of cutaneous T cell lymphoma (CTCL). It has also shown its anticancer activity over a large range of other hematological and solid malignancies, but few studies have been reported in B cell lymphoma. In this study, we aimed to investigate the antitumor activity of SAHA on murine B cell lymphoma cell line A20 cells. We treated A20 cells with different concentrations of SAHA. The effect of SAHA on the proliferation of A20 cells was studied by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium (MTT) assay in vitro; the anti-proliferation activity in vivo was evaluated by proliferating cell nuclear antigen (PCNA) of xenograft tumor tissues through immunocytochemical staining. Apoptosis were detected by Hoechst 33258 staining and Annexin V/propidium iodide (PI) double-labeled cytometry in vitro. The effect of SAHA on cell cycle of A20 cells was studied by a propidium iodide method. Autophagic cell death induced by SAHA was confirmed by transmission electron microscopy (TEM). Angiogenesis marker (CD31) was measured by immunocytochemical staining to investigate the anti-angiogenic effect of SAHA. Western blot was used to detect the expression of signaling pathway factors (phospho-AKT, phospho-ERK, AKT, ERK, Nur77, HIF-1α, and VEGF). Our results showed that SAHA inhibited the proliferation of A20 cells in a time- and dose-dependent manner, induced cell apoptosis and G0/G1 phase arrest of cell cycle, promoted autophagic cell death, and suppressed tumor progress in NCI-A20 cells nude mice xenograft model in vivo. SAHA decreased the activation of AKT (phospho-AKT: p-AKT) and ERK1/2 (phospho-ERK: p-ERK) proteins and inhibited the expression of pro-angiogenic factors (VEGF and HIF-1α), downregulated its downstream signaling factor (Nur77), which might be contributed to the antitumor mechanisms of SAHA.
Collapse
Affiliation(s)
- Bohan Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | | | | | | | | | | |
Collapse
|
10
|
Knopf KB, Duh MS, Lafeuille MH, Gravel J, Lefebvre P, Niculescu L, Ba-Mancini A, Ma E, Shi H, Comenzo RL. Meta-analysis of the efficacy and safety of bortezomib re-treatment in patients with multiple myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 14:380-8. [PMID: 25023616 DOI: 10.1016/j.clml.2014.03.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/28/2014] [Accepted: 03/17/2014] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Bortezomib is administered for a finite course; thus, patients might remain sensitive to bortezomib-based therapy at relapse. We report a meta-analysis of bortezomib-based retreatment in relapsed/refractory myeloma. PATIENTS AND METHODS A systematic literature review identified studies of bortezomib-based retreatment in relapsed/refractory myeloma. Proportions of bortezomib-refractory patients and additional prognostic factors were extracted and used in weighted stratified analyses of TTP and OS. Random-effect pooled estimates were calculated for overall response rate (ORR) and rates of common AEs. RESULTS Twenty-three studies (n = 1051 patients) were identified. Bortezomib was administered intravenously in all studies. Across studies in which data were available, pooled, weighted average ORR was 39.1% (95% confidence interval, 30.8%-47.4%), and pooled, weighted average median TTP and OS were 7.5 and 16.6 months, respectively. Patients with fewer previous therapies (≤ 4) and relapsed (not refractory) patients achieved higher ORRs, of 43.4% and 57.2%, respectively. Random-effects meta-regression analysis confirmed that relapsed patients were associated with a higher ORR by 28 to 41 percentage points versus refractory patients. In relapsed patients, median TTP and OS were 8.5 and 19.7 months, respectively. Common Grade 3/4 AEs included thrombocytopenia (35%), neutropenia (15%), anemia (14%), pneumonia (10%), and peripheral neuropathy (3%). CONCLUSION Based on these findings, bortezomib retreatment is well tolerated and appears efficacious in relapsed patients. In an era of new and emerging treatment options for relapsed and/or refractory myeloma, these data indicate that bortezomib retreatment might be a highly effective option in previously treated patients.
Collapse
Affiliation(s)
- Kevin B Knopf
- California Pacific Medical Center, Sutter Health, San Francisco, CA.
| | | | | | | | | | | | | | - Esprit Ma
- Millennium: The Takeda Oncology Company, Cambridge, MA
| | - Hongliang Shi
- Millennium: The Takeda Oncology Company, Cambridge, MA
| | | |
Collapse
|
11
|
Vorinostat in combination with lenalidomide and dexamethasone in patients with relapsed or refractory multiple myeloma. Blood Cancer J 2014; 4:e182. [PMID: 24562384 PMCID: PMC3944659 DOI: 10.1038/bcj.2014.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 12/11/2013] [Indexed: 01/17/2023] Open
Abstract
The addition of vorinostat to lenalidomide/dexamethasone represents a novel combination therapy in multiple myeloma (MM), informed by laboratory studies suggesting synergy. This was a phase I, multicenter, open-label, non-randomized, dose-escalating study in patients with relapsed or relapsed and refractory MM. Clinical evaluation, electrocardiogram, laboratory studies and adverse events were obtained and assessed. The maximum-tolerated dose was not reached owing to a non-occurrence of two dose-limiting toxicities per six patients tested at any of the dosing levels. Patients tolerated the highest dose tested (Level 5) and this was considered the maximum administered dose: at 400 mg vorinostat on days 1-7 and 15-21, 25 mg lenalidomide on days 1-21 and 40 mg dexamethasone on days 1, 8, 15 and 22, per 28-day cycle. Drug-related adverse events were reported in 90% of patients serious adverse experiences were reported in 45% of the patients and 22% of all patients had adverse experiences considered, possibly related to study drug by the investigators. A confirmed partial response or better was reported for 14/30 patients (47%) evaluable for efficacy, including 31% of patients previously treated with lenalidomide. Vorinostat in combination with lenalidomide and dexamethasone proved tolerable with appropriate supportive care, with encouraging activity observed.
Collapse
|
12
|
Hrabeta J, Stiborova M, Adam V, Kizek R, Eckschlager T. Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2013; 158:161-9. [PMID: 24263215 DOI: 10.5507/bp.2013.085] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/12/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Despite recent success toward discovery of more effective anticancer drugs, chemoresistance remains a major cause of treatment failure. There is emerging evidence that epigenetics plays a key role in the development of the resistance. Epigenetic regulators such as histone acetyltransferases (HATs) and histone deacetylases (HDACs) play an important role in gene expression. The latter are found to be commonly linked with many types of cancers and influence cancer development. Overall, histone acetylation is being investigated as a therapeutic target because of its importance in regulating gene expression. This review summarizes mechanisms of the anticancer effects of histone deacetylase (HDAC) inhibitors and the results of clinical studies. RESULTS Different HDAC inhibitors induce cancer cell death by different mechanisms that include changes in gene expression and alteration of both histone and non-histone proteins. Enhanced histone acetylation in tumors results in modification of expression of genes involved in cell signaling. Inhibition of HDACs causes changed expression in 2-10 % of genes involved in important biological processes. The results of experiments and clinical studies demonstrate that combination of HDAC inhibitors with some anticancer drugs have synergistic or additive effects. CONCLUSIONS Even though many biological effects of HDAC inhibitors have been found, most of the mechanisms of their action remain unclear. In addition, their use in combination with other drugs and the combination regime need to be investigated. The discovery of predictive factors is also necessary. Finally, a key question is whether the pan-HDAC inhibitors or the selective inhibitors will be more efficient for different types of cancers.
Collapse
Affiliation(s)
- Jan Hrabeta
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
13
|
Kaufman JL, Fabre C, Lonial S, Richardson PG. Histone deacetylase inhibitors in multiple myeloma: rationale and evidence for their use in combination therapy. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2013; 13:370-6. [PMID: 23787122 DOI: 10.1016/j.clml.2013.03.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/14/2013] [Accepted: 03/27/2013] [Indexed: 01/07/2023]
Abstract
Multiple myeloma (MM) arises from abnormal proliferation and survival (ie, a high proliferative index and a low apoptotic index) of mature immunoglobulin-producing plasma cells in the bone marrow. Development of novel therapeutic options, such as proteasome inhibitors and immunomodulatory agents (IMiDs), has improved treatment outcomes. However, patients often develop relapsed and refractory MM, thus requiring alternative treatment approaches. Histone acetyltransferases and histone deacetylases (HDACs) control the acetylation status of proteins and affect a broad array of physiologic processes (eg, cell cycle, apoptosis, and protein folding) involved in cell growth and survival. The discovery that HDACs might have a role in various hematologic malignancies, including MM, has led to the development of HDAC inhibitors as potential antitumor agents. Preclinical evidence from studies of HDAC inhibitors in combination with proteasome inhibitors (eg, bortezomib and carfilzomib), other antimyeloma agents, including IMiDs (eg, lenalidomide), and cytotoxic agents (eg, melphalan, pegylated liposomal doxorubicin), provides a strong scientific rationale for the evaluation of these regimens. Results from early stage clinical trials further support the use of HDAC inhibitors as a therapeutic option for MM, in combination with current and emerging antimyeloma agents. In this review, we examine the role of protein acetylation that underlies the antimyeloma effects of HDAC inhibitors, discuss the preclinical rationale for the use of HDAC inhibitors in combination with other antimyeloma agents, and provide an overview of the current clinical evidence supporting the use of HDAC inhibitors as a therapeutic option in MM.
Collapse
Affiliation(s)
- Jonathan L Kaufman
- Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
| | | | | | | |
Collapse
|
14
|
Maes K, Menu E, Van Valckenborgh E, Van Riet I, Vanderkerken K, De Bruyne E. Epigenetic modulating agents as a new therapeutic approach in multiple myeloma. Cancers (Basel) 2013; 5:430-61. [PMID: 24216985 PMCID: PMC3730337 DOI: 10.3390/cancers5020430] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) is an incurable B-cell malignancy. Therefore, new targets and drugs are urgently needed to improve patient outcome. Epigenetic aberrations play a crucial role in development and progression in cancer, including MM. To target these aberrations, epigenetic modulating agents, such as DNA methyltransferase inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi), are under intense investigation in solid and hematological cancers. A clinical benefit of the use of these agents as single agents and in combination regimens has been suggested based on numerous studies in pre-clinical tumor models, including MM models. The mechanisms of action are not yet fully understood but appear to involve a combination of true epigenetic changes and cytotoxic actions. In addition, the interactions with the BM niche are also affected by epigenetic modulating agents that will further determine the in vivo efficacy and thus patient outcome. A better understanding of the molecular events underlying the anti-tumor activity of the epigenetic drugs will lead to more rational drug combinations. This review focuses on the involvement of epigenetic changes in MM pathogenesis and how the use of DNMTi and HDACi affect the myeloma tumor itself and its interactions with the microenvironment.
Collapse
Affiliation(s)
- Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Ivan Van Riet
- Stem Cell Laboratory, Department Clinical Hematology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussel, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
| |
Collapse
|
15
|
Dolloff NG, Talamo G. Targeted Therapy of Multiple Myeloma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 779:197-221. [DOI: 10.1007/978-1-4614-6176-0_9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Trials with 'epigenetic' drugs: an update. Mol Oncol 2012; 6:657-82. [PMID: 23103179 DOI: 10.1016/j.molonc.2012.09.004] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/30/2012] [Indexed: 02/06/2023] Open
Abstract
Epigenetic inactivation of pivotal genes involved in correct cell growth is a hallmark of human pathologies, in particular cancer. These epigenetic mechanisms, including crosstalk between DNA methylation, histone modifications and non-coding RNAs, affect gene expression and are associated with disease progression. In contrast to genetic mutations, epigenetic changes are potentially reversible. Re-expression of genes epigenetically inactivated can result in the suppression of disease state or sensitization to specific therapies. Small molecules that reverse epigenetic inactivation, so-called epi-drugs, are now undergoing clinical trials. Accordingly, the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for cancer treatment have approved some of these drugs. Here, we focus on the biological features of epigenetic molecules, analyzing the mechanism(s) of action and their current use in clinical practice.
Collapse
|
17
|
Bianchi G, Ghobrial IM. Molecular mechanisms of effectiveness of novel therapies in multiple myeloma. Leuk Lymphoma 2012; 54:229-41. [DOI: 10.3109/10428194.2012.706287] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
18
|
Scholz B, Marschalek R. Epigenetics and blood disorders. Br J Haematol 2012; 158:307-22. [DOI: 10.1111/j.1365-2141.2012.09193.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 05/09/2012] [Indexed: 12/25/2022]
Affiliation(s)
- Bastian Scholz
- Institute of Pharmaceutical Biology/DCAL; Biocentre; Goethe-University; Frankfurt/Main; Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/DCAL; Biocentre; Goethe-University; Frankfurt/Main; Germany
| |
Collapse
|
19
|
Role of Histone Deacetylase Inhibitors in the Treatment of Lymphomas and Multiple Myeloma. Hematol Oncol Clin North Am 2012; 26:671-704, ix. [DOI: 10.1016/j.hoc.2012.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
20
|
Verbrugge I, Johnstone RW, Bots M. Promises and challenges of anticancer drugs that target the epigenome. Epigenomics 2012; 3:547-65. [PMID: 22126246 DOI: 10.2217/epi.11.82] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The occurrence of epigenetic aberrations in cancer and their role in promoting tumorigenesis has led to the development of various small molecule inhibitors that target epigenetic enzymes. In preclinical settings, many epigenetic inhibitors demonstrate promising activity against a variety of both hematological and solid tumors. The therapeutic efficacy of those inhibitors that have entered the clinic however, is restricted predominantly to hematological malignancies. Here we outline the observed epigenetic aberrations in various types of cancer and the clinical responses to epigenetic drugs. We furthermore discuss strategies to improve the responsiveness of both hematological and solid malignancies to epigenetic drugs.
Collapse
Affiliation(s)
- Inge Verbrugge
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne 3002, Victoria, Australia
| | | | | |
Collapse
|
21
|
Dell'Aversana C, Lepore I, Altucci L. HDAC modulation and cell death in the clinic. Exp Cell Res 2012; 318:1229-44. [PMID: 22336671 DOI: 10.1016/j.yexcr.2012.01.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 01/26/2012] [Accepted: 01/27/2012] [Indexed: 01/29/2023]
Abstract
Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are two opposing classes of enzymes, which finely regulate the balance of histone acetylation affecting chromatin packaging and gene expression. Imbalanced acetylation has been associated with carcinogenesis and cancer progression. In contrast to genetic mutations, epigenetic changes are potentially reversible. This implies that epigenetic alterations are amenable to pharmacological interventions. Accordingly, some epigenetic-based drugs (epidrugs) have been approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for cancer treatment. Here, we focus on the biological features of HDAC inhibitors (HDACis), analyzing the mechanism(s) of action and their current use in clinical practice.
Collapse
|
22
|
Abstract
Myeloma therapy has undergone significant advances in recent years resulting in a marked improvement in survival. Knowledge of the active pathways involved in myeloma pathogenesis has led to the discovery of novel agents and greatly expanded the potential armamentarium available for treatment. This better understanding of the disease and resistance mechanisms has resulted in new agent classes that are being evaluated in preclinical and early clinical studies. In addition, dosing for existing agents is being optimized, and they are being given in new combinations. In this article, we review experimental agents that are showing promise in multiple myeloma treatment. New biological agents in clinical trials hold the promise of efficacy through novel mechanisms of action, with a significant reduction of dose-limiting toxicities compared with classic cytotoxic chemotherapeutics. Second-generation proteasome inhibitors and immunomodulatory agents are furthest along in clinical development, and histone deacetylase inhibitors, heat shock protein 90 inhibitors, Akt inhibitors and monoclonal antibodies are some of the other agents entering later-phase clinical trials. We also review developments in targeting the myeloma stem cell as an exciting new treatment direction.
Collapse
Affiliation(s)
- Saad A. Khan
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Adam D. Cohen
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
23
|
Evidence-Based Mini-Review: Treatment Options for Patients with Relapsed/Refractory Myeloma Previously Treated with Novel Agents and High-Dose Chemotherapy and Autologous Stem-Cell Transplantation. Hematology 2010. [DOI: 10.1182/asheducation.v2010.1.310.3645859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
24
|
Watanabe T, Nagase K, Chosa M, Tobinai K. Schwann cell autophagy induced by SAHA, 17-AAG, or clonazepam can reduce bortezomib-induced peripheral neuropathy. Br J Cancer 2010; 103:1580-7. [PMID: 20959823 PMCID: PMC2990589 DOI: 10.1038/sj.bjc.6605954] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The proteasome inhibitor bortezomib has improved the survival of patients with multiple myeloma but bortezomib-induced peripheral neuropathy (BiPN) has emerged as a serious potential complication of this therapy. Animal studies suggest that bortezomib predominantly causes pathological changes in Schwann cells. A tractable system to evaluate combination drugs for use with bortezomib is essential to enable continuing clinical benefit from this drug. METHODS Rat schwannoma cells were pretreated with vincristine (VCR), histone deacetylase inhibitors, anticonvulsants, or a heat-shock protein 90 (HSP90) inhibitor. To then monitor aggresome formation as a result of proteasome inhibition and the activation of chaperone-mediated autophagy (CMA), we performed double-labelling immunofluorescent analyses of a cellular aggregation-prone protein marker. RESULTS Aggresome formation was interrupted by VCR, whereas combination treatments with bortezomib involving suberoylanilide hydroxamic acid, 17-allylamino-17-demethoxy-geldanamycin, or clonazepam appear to facilitate the disposal of unfolded proteins via CMA, inducing HSP70 and lysosome-associated membrane protein type 2A (LAMP-2A). CONCLUSIONS This schwannoma model can be used to test BiPN-reducing drugs. The present data suggest that aggresome formation in Schwann cells is a possible mechanism of BiPN, and drugs that induce HSP70 or LAMP-2A have the potential to alleviate this complication. Combination clinical trials are warranted to confirm the relevance of these observations.
Collapse
Affiliation(s)
- T Watanabe
- Hematology Division, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | | | | | | |
Collapse
|
25
|
Shah N, Lonial S. Evidence-based mini-review: treatment options for patients with relapsed/refractory myeloma previously treated with novel agents and high-dose chemotherapy and autologous stem-cell transplantation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2010; 2010:310-313. [PMID: 21239811 PMCID: PMC4086196 DOI: 10.1182/asheducation-2010.1.310] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
A 64 year-old man with a history of multiple myeloma presents with new back pain. He has a history of International Staging System stage 1, IgG kappa multiple myeloma with normal cytogenetics which was diagnosed 4 years ago when he presented with a pathological fracture of the left humerus. He was initially managed with mechanical stabilization and 4 cycles of bortezomib-dexamethasone, as well as 2 years of bisphosphanates. Following induction therapy he achieved a very good partial response (VGPR). He subsequently received high-dose melphalan and autologous stem cell transplantation (auto-SCT) and achieved a complete reponse (CR) post-transplant. He did not receive maintenance therapy and had been lost-to follow-up for about a year. He now presents 5 years after initial diagnosis with back pain and is noted to have a new lytic lesion with a compression fracture at T8. A serum protein electrophoresis demonstrates reappearance of his original monoclonal protein. After appropriate stabilization he comes to you to discuss additional treatment options.
Collapse
Affiliation(s)
- Nina Shah
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|