101
|
Voorhees PM, Gasparetto C, Moore DT, Winans D, Orlowski RZ, Hurd DD. Final Results of a Phase 1 Study of Vorinostat, Pegylated Liposomal Doxorubicin, and Bortezomib in Relapsed or Refractory Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:424-432. [PMID: 28655599 DOI: 10.1016/j.clml.2017.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/07/2017] [Accepted: 05/04/2017] [Indexed: 11/17/2022]
Abstract
INTRODUCTION/BACKGROUND Deacetylase inhibitors have synergistic activity in combination with proteasome inhibitors and anthracyclines in preclinical models of multiple myeloma (MM). We therefore evaluated the safety and efficacy of the deacetylase inhibitor vorinostat in combination with pegylated liposomal doxorubicin (PLD) and bortezomib in relapsed/refractory MM. PATIENTS AND METHODS Thirty-two patients were treated with PLD and bortezomib in combination with escalating doses of vorinostat on days 4 to 11 or 1 to 14. RESULTS The maximum tolerated dose of vorinostat was 400 mg on days 4 to 11. Neutropenia and thrombocytopenia attributable to protocol therapy were seen in 59% and 94% of patients, of which 37% and 47% were of grade 3 or higher severity, respectively. Constitutional and gastrointestinal adverse events of all grades were common, the majority of which were less than grade 3 in severity. The overall response rate (partial response rate or better) was 65% and the clinical benefit rate (minimal response rate or better) 74%. The overall response rate was 83%, 71%, and 45% for patients with bortezomib-naive, -sensitive, and -refractory MM, respectively. The median progression-free survival was 13.9 months and the 3-year overall survival 77%. Whole blood proteasome activity assays demonstrated a potential impact of vorinostat on the chymotryptic-like activity of the proteasome. CONCLUSION Further evaluation of PLD, bortezomib, and deacetylase inhibitor combinations is warranted, with special attention directed toward strategies to improve tolerability.
Collapse
Affiliation(s)
- Peter M Voorhees
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC.
| | - Cristina Gasparetto
- Division of Hematological Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC
| | - Dominic T Moore
- Division of Hematology-Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Diane Winans
- Division of Hematology-Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX
| | - David D Hurd
- Section on Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center of Wake Forest University, Winston-Salem, NC
| |
Collapse
|
102
|
Matondo M, Marcellin M, Chaoui K, Bousquet-Dubouch MP, Gonzalez-de-Peredo A, Monsarrat B, Burlet-Schiltz O. Determination of differentially regulated proteins upon proteasome inhibition in AML cell lines by the combination of large-scale and targeted quantitative proteomics. Proteomics 2017; 17:1600089. [PMID: 27709814 PMCID: PMC5396343 DOI: 10.1002/pmic.201600089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 09/05/2016] [Accepted: 10/14/2016] [Indexed: 01/08/2023]
Abstract
The ubiquitin-proteasome pathway (UPP) plays a critical role in the degradation of proteins implicated in cell cycle control, signal transduction, DNA damage response, apoptosis and immune response. Proteasome inhibitors can inhibit the growth of a broad spectrum of human cancer cells by altering the balance of intracellular proteins. However, the targets of these compounds in acute myeloid leukemia (AML) cells have not been fully characterized. Herein, we combined large-scale quantitative analysis by SILAC-MS and targeted quantitative proteomic analysis in order to identify proteins regulated upon proteasome inhibition in two AML cell lines displaying different stages of maturation: immature KG1a cells and mature U937 cells. In-depth data analysis enabled accurate quantification of more than 7000 proteins in these two cell lines. Several candidates were validated by selected reaction monitoring (SRM) measurements in a large number of samples. Despite the broad range of proteins known to be affected by proteasome inhibition, such as heat shock (HSP) and cell cycle proteins, our analysis identified new differentially regulated proteins, including IL-32, MORF family mortality factors and apoptosis inducing factor SIVA, a target of p53. It could explain why proteasome inhibitors induce stronger apoptotic responses in immature AML cells.
Collapse
Affiliation(s)
- Mariette Matondo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, France
| | - Marlène Marcellin
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, France
| | - Karima Chaoui
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, France
| | | | - Anne Gonzalez-de-Peredo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, France
| | - Bernard Monsarrat
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, France
| |
Collapse
|
103
|
Gu JJ, Kaufman GP, Mavis C, Czuczman MS, Hernandez-Ilizaliturri FJ. Mitotic catastrophe and cell cycle arrest are alternative cell death pathways executed by bortezomib in rituximab resistant B-cell lymphoma cells. Oncotarget 2017; 8:12741-12753. [PMID: 28055975 PMCID: PMC5355050 DOI: 10.18632/oncotarget.14405] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
The ubiqutin-proteasome system (UPS) plays a role in rituximab-chemotherapy resistance and bortezomib (BTZ) possesses caspase-dependent (i.e. Bak stabilization) and a less characterized caspase-independent mechanism-of-action(s). Here, we define BTZ-induced caspase-independent cell death pathways. A panel of rituximab-sensitive (RSCL), rituximab-resistant cell lines (RRCL) and primary tumor cells derived from lymphoma patients (N = 13) were exposed to BTZ. Changes in cell viability, cell-cycle, senescence, and mitotic index were quantified. In resting conditions, RRCL exhibits a low-proliferation rate, accumulation of cells in S-phase and senescence. Exposure of RRCL to BTZ reduces cell senescence, induced G2-M phase cell-cycle arrest, and is associated with mitotic catastrophe. BTZ stabilized p21, CDC2, and cyclin B in RRCL and in primary tumor cells. Transient p21 knockdown alleviates BTZ-induced senescence inhibition, G2-M cell cycle blockade, and mitotic catastrophe. Our data suggest that BTZ can induce apoptosis or mitotic catastrophe and that p21 has a pivotal role in BTZ activity against RRCL.
Collapse
Affiliation(s)
- Juan J Gu
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Gregory P Kaufman
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, Celgene Corporation, Summit, NJ, USA
| | - Cory Mavis
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Francisco J Hernandez-Ilizaliturri
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
104
|
Momose I, Watanabe T. Tyropeptins, proteasome inhibitors produced by Kitasatospora sp. MK993-dF2. J Antibiot (Tokyo) 2017; 70:542-550. [PMID: 28196975 DOI: 10.1038/ja.2017.9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/17/2022]
Abstract
Tyropeptins are new proteasome inhibitors isolated from the culture broth of Kitasatospora sp. MK993-dF2. Tyropeptins permeate cell membranes, inhibit intracellular proteasomes and reduce the degradation of ubiquitinated proteins in mammalian cells. We performed structure-based drug design and structure-activity relationship studies on tyropeptin derivatives to obtain valuable information of derivatives. Among the synthesized tyropeptin derivatives, some boronic acid derivatives exhibited potent antitumor effects against human multiple myeloma. In this review, we summarize the discovery of tyropeptins and the development of tyropeptin derivatives.
Collapse
Affiliation(s)
- Isao Momose
- Institute of Microbial Chemistry (BIKAKEN), Shizuoka, Japan
| | | |
Collapse
|
105
|
Durie BGM, Hoering A, Abidi MH, Rajkumar SV, Epstein J, Kahanic SP, Thakuri M, Reu F, Reynolds CM, Sexton R, Orlowski RZ, Barlogie B, Dispenzieri A. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial. Lancet 2017; 389:519-527. [PMID: 28017406 PMCID: PMC5546834 DOI: 10.1016/s0140-6736(16)31594-x] [Citation(s) in RCA: 674] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/09/2016] [Accepted: 08/26/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Lenalidomide plus dexamethasone is a reference treatment for patients with newly diagnosed myeloma. The combination of the proteasome inhibitor bortezomib with lenalidomide and dexamethasone has shown significant efficacy in the setting of newly diagnosed myeloma. We aimed to study whether the addition of bortezomib to lenalidomide and dexamethasone would improve progression-free survival and provide better response rates in patients with previously untreated multiple myeloma who were not planned for immediate autologous stem-cell transplant. METHODS In this randomised, open-label, phase 3 trial, we recruited patients with newly diagnosed multiple myeloma aged 18 years and older from participating Southwest Oncology Group (SWOG) and National Clinical Trial Network (NCTN) institutions (both inpatient and outpatient settings). Key inclusion criteria were presence of CRAB (C=calcium elevation; R=renal impairment; A=anaemia; B=bone involvement) criteria with measurable disease (measured by assessment of free light chains), Eastern Cooperative Oncology Group (ECOG) performance status of 0-3, haemoglobin concentration 9 g/dL or higher, absolute neutrophil count 1 × 103 cells per mm3 or higher, and a platelet count of 80 000/mm3 or higher. We randomly assigned (1:1) patients to receive either an initial treatment of bortezomib with lenalidomide and dexamethasone (VRd group) or lenalidomide and dexamethasone alone (Rd group). Randomisation was stratified based on International Staging System stage (I, II, or III) and intent to transplant (yes vs no). The VRd regimen was given as eight 21-day cycles. Bortezomib was given at 1·3 mg/m2 intravenously on days 1, 4, 8, and 11, combined with oral lenalidomide 25 mg daily on days 1-14 plus oral dexamethasone 20 mg daily on days 1, 2, 4, 5, 8, 9, 11, and 12. The Rd regimen was given as six 28-day cycles. The standard Rd regimen consisted of 25 mg oral lenalidomide once a day for days 1-21 plus 40 mg oral dexamethasone once a day on days 1, 8, 15, and 22. The primary endpoint was progression-free survival using a prespecified one-sided stratified log rank test at a significance level of 0·02. Analyses were intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00644228. FINDINGS Between April, 2008, and February, 2012, we randomly assigned 525 patients at 139 participating institutions (264 to VRd and 261 to Rd). In the randomly assigned patients, 21 patients in the VRd group and 31 in the Rd group were deemed ineligible based mainly on missing, insufficient, or early or late baseline laboratory data. Median progression-free survival was significantly improved in the VRd group (43 months vs 30 months in the Rd group; stratified hazard ratio [HR] 0·712, 96% CI 0·56-0·906; one-sided p value 0·0018). The median overall survival was also significantly improved in the VRd group (75 months vs 64 months in the Rd group, HR 0·709, 95% CI 0·524-0·959; two-sided p value 0·025). The rates of overall response (partial response or better) were 82% (176/216) in the VRd group and 72% (153/214) in the Rd group, and 16% (34/216) and 8% (18/214) of patients who were assessable for response in these respective groups had a complete response or better. Adverse events of grade 3 or higher were reported in 198 (82%) of 241 patients in the VRd group and 169 (75%) of 226 patients in the Rd group; 55 (23%) and 22 (10%) patients discontinued induction treatment because of adverse events, respectively. There were no treatment-related deaths in the Rd group, and two in the VRd group. INTERPRETATION In patients with newly diagnosed myeloma, the addition of bortezomib to lenalidomide and dexamethasone resulted in significantly improved progression-free and overall survival and had an acceptable risk-benefit profile. FUNDING NIH, NCI, NCTN, Millennium Pharmaceuticals, Takeda Oncology Company, and Celgene Corporation.
Collapse
Affiliation(s)
- Brian G M Durie
- Cedars-Sinai Samuel Oschin Cancer Center, Los Angeles, CA, USA.
| | | | - Muneer H Abidi
- Spectrum Health Cancer Center, Michigan State University, Grand Rapids, MI, USA
| | | | - Joshua Epstein
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stephen P Kahanic
- Siouxland Regional Cancer Center, Sanford NCORP of the Northern Central Plains, Sioux City, IA, USA
| | - Mohan Thakuri
- Southeast Clinical Oncology Research Consortium NCORP, Cancer Care of Western North Carolina, Asheville, NC, USA
| | - Frederic Reu
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | | | | | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bart Barlogie
- Myeloma Program, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
106
|
Bone marrow microenvironment-derived signals induce Mcl-1 dependence in multiple myeloma. Blood 2017; 129:1969-1979. [PMID: 28151428 DOI: 10.1182/blood-2016-10-745059] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/30/2017] [Indexed: 01/25/2023] Open
Abstract
Multiple myeloma is highly dependent on the bone marrow microenvironment until progressing to very advanced extramedullary stages of the disease such as plasma cell leukemia. Stromal cells in the bone marrow secrete a variety of cytokines that promote plasma cell survival by regulating antiapoptotic members of the Bcl-2 family including Mcl-1, Bcl-xL, and Bcl-2. Although the antiapoptotic protein on which a cell depends is typically consistent among normal cells of a particular phenotype, Bcl-2 family dependence is highly heterogeneous in multiple myeloma. Although normal plasma cells and most multiple myeloma cells require Mcl-1 for survival, a subset of myeloma is codependent on Bcl-2 and/or Bcl-xL We investigated the role of the bone marrow microenvironment in determining Bcl-2 family dependence in multiple myeloma. We used the Bcl-2/Bcl-xL inhibitor ABT-737 to study the factors regulating whether myeloma is Mcl-1 dependent, and thus resistant to ABT-737-induced apoptosis, or Bcl-2/Bcl-xL codependent, and thus sensitive to ABT-737. We demonstrate that bone marrow stroma is capable of inducing Mcl-1 dependence through the production of the plasma cell survival cytokine interleukin-6 (IL-6). IL-6 upregulates Mcl-1 transcription in a STAT3-dependent manner, although this occurred in a minority of the cells tested. In all cells, IL-6 treatment results in posttranslational modification of the proapoptotic protein Bim. Phosphorylation of Bim shifts its binding from Bcl-2 and Bcl-xL to Mcl-1, an effect reversed by MEK inhibition. Blocking IL-6 or downstream signaling restored Bcl-2/Bcl-xL dependence and may therefore represent a clinically useful strategy to enhance the activity of Bcl-2 inhibitors.
Collapse
|
107
|
Amanullah A, Upadhyay A, Chhangani D, Joshi V, Mishra R, Yamanaka K, Mishra A. Proteasomal Dysfunction Induced By Diclofenac Engenders Apoptosis Through Mitochondrial Pathway. J Cell Biochem 2017; 118:1014-1027. [DOI: 10.1002/jcb.25666] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/01/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Deepak Chhangani
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology Research Institute of Environmental Medicine; Nagoya University Furo-cho; Chikusa-ku Nagoya 464-8601 Japan
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| |
Collapse
|
108
|
Omari SA, Adams MJ, Geraghty DP. TRPV1 Channels in Immune Cells and Hematological Malignancies. ADVANCES IN PHARMACOLOGY 2017; 79:173-198. [DOI: 10.1016/bs.apha.2017.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
109
|
Anderson KC. Progress and Paradigms in Multiple Myeloma. Clin Cancer Res 2016; 22:5419-5427. [PMID: 28151709 PMCID: PMC5300651 DOI: 10.1158/1078-0432.ccr-16-0625] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022]
Abstract
Remarkable progress has been achieved in multiple myeloma, and patient median survival has been extended 3- to 4-fold. Specifically, there have been 18 newly approved treatments for multiple myeloma in the past 12 years, including seven in 2015, and the treatment paradigm and patient outcome have been transformed. The definition of patients benefitting from these therapies has been broadened. Response criteria now include minimal residual disease (MRD), assessed in bone marrow by multicolor flow cytometry or sequencing, and by imaging for extramedullary disease. Initial therapy for transplant candidates is a triplet incorporating novel therapies-that is, lenalidomide, bortezomib, and dexamethasone or cyclophosphamide, bortezomib, and dexamethasone. Lenalidomide maintenance until progression can prolong progression-free and overall survival in standard-risk multiple myeloma, with incorporation of proteasome inhibitor for high-risk disease. Studies are evaluating the value of early versus late transplant and MRD as a therapeutic goal to inform therapy. In nontransplant patients, triplet therapies are also preferred, with doublet therapy reserved for frail patients, and maintenance as described above. The availability of second-generation proteasome inhibitors (carfilzomib and ixazomib), immunomodulatory drugs (pomalidomide), histone deacetylase inhibitors (panobinostat), and monoclonal antibodies (elotuzumab and daratumumab) allows for effective combination therapies of relapsed disease as well. Finally, novel therapies targeting protein degradation, restoring autologous memory anti-multiple myeloma immunity, and exploiting genetic vulnerabilities show promise to improve patient outcome even further. Clin Cancer Res; 22(22); 5419-27. ©2016 AACR SEE ALL ARTICLES IN THIS CCR FOCUS SECTION, "MULTIPLE MYELOMA MULTIPLYING THERAPIES".
Collapse
Affiliation(s)
- Kenneth C Anderson
- Division of Hematologic Malignancy, Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
110
|
Kharfan-Dabaja M, Nishihori T, Reljic T, Hamadani M, Baz R, Ochoa-Bayona JL, Mhaskar R, Djulbegovic B, Kumar A. Three-drug versus two-drug induction therapy regimens for patients with transplant-eligible multiple myeloma. Hippokratia 2016. [DOI: 10.1002/14651858.cd010602.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Mohamed Kharfan-Dabaja
- H. Lee Moffitt Cancer Center, Division of Oncologic Sciences, University of South Florida; Department of Blood and Marrow Transplantation; Tampa Florida USA
| | - Taiga Nishihori
- Moffitt Cancer Center; Department of Blood and Marrow Transplantation; Tampa Florida USA
| | - Tea Reljic
- University of South Florida; Center for Evidence Based Medicine and Health Outcomes Research; 12901 Bruce B. Downs Blvd., MDC27 Tampa Florida USA 33612
| | - Mehdi Hamadani
- Mary Babb Randolph Cancer Center, West Virginia University; Blood and Marrow Transplantation Program; Morgantown USA
| | - Rachid Baz
- Moffitt Cancer Center; Department of Hematologic Malignancies; 12902 USF Magnolia Dr. Tampa Florida USA 33612
| | - José L. Ochoa-Bayona
- Moffitt Cancer Center; Department of Blood and Marrow Transplantation; Tampa Florida USA
| | - Rahul Mhaskar
- University of South Florida; Center for Evidence Based Medicine and Health Outcomes Research; 12901 Bruce B. Downs Blvd., MDC27 Tampa Florida USA 33612
| | - Benjamin Djulbegovic
- University of South Florida & Mofftt Cancer Center; USF Program for Comparative Effectiveness Research & Dpt of Hematology, Moffitt Cancer Ctr; Tampa Florida USA
| | - Ambuj Kumar
- University of South Florida; Center for Evidence Based Medicine and Health Outcomes Research; 12901 Bruce B. Downs Blvd., MDC27 Tampa Florida USA 33612
| |
Collapse
|
111
|
Harper JW, Bennett EJ. Proteome complexity and the forces that drive proteome imbalance. Nature 2016; 537:328-38. [PMID: 27629639 DOI: 10.1038/nature19947] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 07/29/2016] [Indexed: 12/28/2022]
Abstract
The cellular proteome is a complex microcosm of structural and regulatory networks that requires continuous surveillance and modification to meet the dynamic needs of the cell. It is therefore crucial that the protein flux of the cell remains in balance to ensure proper cell function. Genetic alterations that range from chromosome imbalance to oncogene activation can affect the speed, fidelity and capacity of protein biogenesis and degradation systems, which often results in proteome imbalance. An improved understanding of the causes and consequences of proteome imbalance is helping to reveal how these systems can be targeted to treat diseases such as cancer.
Collapse
Affiliation(s)
- J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Eric J Bennett
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
112
|
Offidani M, Corvatta L, Gentili S, Maracci L, Leoni P. Oral ixazomib maintenance therapy in multiple myeloma. Expert Rev Anticancer Ther 2016; 16:21-32. [PMID: 26588946 DOI: 10.1586/14737140.2016.1123627] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Continuous therapy has proven to be an effective therapeutic strategy to improve the outcome of both young and elderly multiple myeloma patients. Remarkably, lenalidomide and bortezomib showed to play a crucial role in this setting due to their safety profile allowing long-term exposure. Ixazomib, the first oral proteasome inhibitor to be evaluated in multiple myeloma, exerts substantial anti-myeloma activity as a single agent and particularly in combination with immunomodulatory drugs and it may be an attractive option for maintenance therapy. Here we address the issue of maintenance therapy as part of a therapeutic approach of multiple myeloma patients focusing on the potential role of ixazomib.
Collapse
Affiliation(s)
- Massimo Offidani
- a Azienda Ospedaliero-Universitaria , Ospedali Riuniti di Ancona , Ancona , Italy
| | - Laura Corvatta
- b Dipartimento di Medicina, UOC Medicina , Fabriano , Italy
| | - Silvia Gentili
- a Azienda Ospedaliero-Universitaria , Ospedali Riuniti di Ancona , Ancona , Italy
| | - Laura Maracci
- a Azienda Ospedaliero-Universitaria , Ospedali Riuniti di Ancona , Ancona , Italy
| | - Pietro Leoni
- a Azienda Ospedaliero-Universitaria , Ospedali Riuniti di Ancona , Ancona , Italy
| |
Collapse
|
113
|
Al-Hujaily EM, Oldham RAA, Hari P, Medin JA. Development of Novel Immunotherapies for Multiple Myeloma. Int J Mol Sci 2016; 17:E1506. [PMID: 27618026 PMCID: PMC5037783 DOI: 10.3390/ijms17091506] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a disorder of terminally differentiated plasma cells characterized by clonal expansion in the bone marrow (BM). It is the second-most common hematologic malignancy. Despite significant advances in therapeutic strategies, MM remains a predominantly incurable disease emphasizing the need for the development of new treatment regimens. Immunotherapy is a promising treatment modality to circumvent challenges in the management of MM. Many novel immunotherapy strategies, such as adoptive cell therapy and monoclonal antibodies, are currently under investigation in clinical trials, with some already demonstrating a positive impact on patient survival. In this review, we will summarize the current standards of care and discuss major new approaches in immunotherapy for MM.
Collapse
Affiliation(s)
- Ensaf M Al-Hujaily
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Robyn A A Oldham
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Parameswaran Hari
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jeffrey A Medin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
114
|
Abstract
Background. Multiple myeloma accounts for 10% of all haematologic malignancies worldwide. In Europe, over 10 000 new cases and nearly 8000 deaths were attributed to multiple myeloma in 2000. Unlike other malignancies, in which surgery and radiation are important treatment modalities, myeloma is exclusively treated with stem cell transplantation and drug therapy, requiring pharmacists to stay abreast of new developments. The melphalan-prednisolone and vincristine-doxorubicin-dexamethasone (VAD) regimens, which have been standard treatments for multiple myeloma over the past few decades, have yielded responses without real survival benefits. Transplantation utilizing high-dose chemotherapy has produced the only meaningful survival benefits for patients with multiple myeloma, but many patients are not candidates for this aggressive treatment option. More effective therapies for multiple myeloma are needed. Objective. To address the mechanisms of action, safety, and efficacy of novel approaches to the treatment of myeloma involving bortezomib, thalidomide and its analogues, lenalidomide and CC-4047 (ActimidTM), and arsenic trioxide as single agents or in combination regimens. Data sources. Published preclinical and primary clinical trial results, as well as scientific or clinical meeting abstracts. The author determined the relevance and subsequent inclusion of the data. Conclusions. Bortezomib is approved in the US and Europe as single-agent therapy for the treatment of relapsed or refractory multiple myeloma. Thalidomide, its analogues, and arsenic trioxide have demonstrated activity and are under investigation in this disease. Further clinical trials of the efficacy and toxicity of these novel agents are ongoing and will further define optimal combinations and sequencing with conventional therapies.
Collapse
|
115
|
Molecular and cellular basis for the unique functioning of Nrf1, an indispensable transcription factor for maintaining cell homoeostasis and organ integrity. Biochem J 2016; 473:961-1000. [PMID: 27060105 DOI: 10.1042/bj20151182] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/26/2016] [Indexed: 12/30/2022]
Abstract
The consensuscis-regulatory AP-1 (activator protein-1)-like AREs (antioxidant-response elements) and/or EpREs (electrophile-response elements) allow for differential recruitment of Nrf1 [NF-E2 (nuclear factor-erythroid 2)-related factor 1], Nrf2 and Nrf3, together with each of their heterodimeric partners (e.g. sMaf, c-Jun, JunD or c-Fos), to regulate different sets of cognate genes. Among them, NF-E2 p45 and Nrf3 are subject to tissue-specific expression in haemopoietic and placental cell lineages respectively. By contrast, Nrf1 and Nrf2 are two important transcription factors expressed ubiquitously in various vertebrate tissues and hence may elicit putative combinational or competitive functions. Nevertheless, they have de facto distinct biological activities because knockout of their genes in mice leads to distinguishable phenotypes. Of note, Nrf2 is dispensable during development and growth, albeit it is accepted as a master regulator of antioxidant, detoxification and cytoprotective genes against cellular stress. Relative to the water-soluble Nrf2, less attention has hitherto been drawn to the membrane-bound Nrf1, even though it has been shown to be indispensable for embryonic development and organ integrity. The biological discrepancy between Nrf1 and Nrf2 is determined by differences in both their primary structures and topovectorial subcellular locations, in which they are subjected to distinct post-translational processing so as to mediate differential expression of ARE-driven cytoprotective genes. In the present review, we focus on the molecular and cellular basis for Nrf1 and its isoforms, which together exert its essential functions for maintaining cellular homoeostasis, normal organ development and growth during life processes. Conversely, dysfunction of Nrf1 results in spontaneous development of non-alcoholic steatohepatitis, hepatoma, diabetes and neurodegenerative diseases in animal models.
Collapse
|
116
|
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: 40] [Impact Index Per Article: 4.4] [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
|
117
|
The life cycle of the 26S proteasome: from birth, through regulation and function, and onto its death. Cell Res 2016; 26:869-85. [PMID: 27444871 PMCID: PMC4973335 DOI: 10.1038/cr.2016.86] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The 26S proteasome is a large, ∼2.5 MDa, multi-catalytic ATP-dependent protease complex that serves as the degrading arm of the ubiquitin system, which is the major pathway for regulated degradation of cytosolic, nuclear and membrane proteins in all eukaryotic organisms.
Collapse
|
118
|
Shukla N, Somwar R, Smith RS, Ambati S, Munoz S, Merchant M, D'Arcy P, Wang X, Kobos R, Antczak C, Bhinder B, Shum D, Radu C, Yang G, Taylor BS, Ng CKY, Weigelt B, Khodos I, de Stanchina E, Reis-Filho JS, Ouerfelli O, Linder S, Djaballah H, Ladanyi M. Proteasome Addiction Defined in Ewing Sarcoma Is Effectively Targeted by a Novel Class of 19S Proteasome Inhibitors. Cancer Res 2016; 76:4525-34. [PMID: 27256563 DOI: 10.1158/0008-5472.can-16-1040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/09/2016] [Indexed: 01/05/2023]
Abstract
Ewing sarcoma is a primitive round cell sarcoma with a peak incidence in adolescence that is driven by a chimeric oncogene created from the fusion of the EWSR1 gene with a member of the ETS family of genes. Patients with metastatic and recurrent disease have dismal outcomes and need better therapeutic options. We screened a library of 309,989 chemical compounds for growth inhibition of Ewing sarcoma cells to provide the basis for the development of novel therapies and to discover vulnerable pathways that might broaden our understanding of the pathobiology of this aggressive sarcoma. This screening campaign identified a class of benzyl-4-piperidone compounds that selectively inhibit the growth of Ewing sarcoma cell lines by inducing apoptosis. These agents disrupt 19S proteasome function through inhibition of the deubiquitinating enzymes USP14 and UCHL5. Functional genomic data from a genome-wide shRNA screen in Ewing sarcoma cells also identified the proteasome as a node of vulnerability in Ewing sarcoma cells, providing orthologous confirmation of the chemical screen findings. Furthermore, shRNA-mediated silencing of USP14 or UCHL5 in Ewing sarcoma cells produced significant growth inhibition. Finally, treatment of a xenograft mouse model of Ewing sarcoma with VLX1570, a benzyl-4-piperidone compound derivative currently in clinical trials for relapsed multiple myeloma, significantly inhibited in vivo tumor growth. Overall, our results offer a preclinical proof of concept for the use of 19S proteasome inhibitors as a novel therapeutic strategy for Ewing sarcoma. Cancer Res; 76(15); 4525-34. ©2016 AACR.
Collapse
Affiliation(s)
- Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Romel Somwar
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roger S Smith
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sri Ambati
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stanley Munoz
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Melinda Merchant
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Padraig D'Arcy
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Xin Wang
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Rachel Kobos
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christophe Antczak
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Bhavneet Bhinder
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - David Shum
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Constantin Radu
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Guangbin Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York. Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Inna Khodos
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stig Linder
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden. Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Hakim Djaballah
- High-Throughput Drug Screening Facility, Memorial Sloan Kettering Cancer Center, New YorkNew York
| | - Marc Ladanyi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
119
|
Zhang XD, Baladandayuthapani V, Lin H, Mulligan G, Li B, Esseltine DLW, Qi L, Xu J, Hunziker W, Barlogie B, Usmani SZ, Zhang Q, Crowley J, Hoering A, Shah JJ, Weber DM, Manasanch EE, Thomas SK, Li BZ, Wang HH, Zhang J, Kuiatse I, Tang JL, Wang H, He J, Yang J, Milan E, Cenci S, Ma WC, Wang ZQ, Davis RE, Yang L, Orlowski RZ. Tight Junction Protein 1 Modulates Proteasome Capacity and Proteasome Inhibitor Sensitivity in Multiple Myeloma via EGFR/JAK1/STAT3 Signaling. Cancer Cell 2016; 29:639-652. [PMID: 27132469 PMCID: PMC4983190 DOI: 10.1016/j.ccell.2016.03.026] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/26/2015] [Accepted: 03/25/2016] [Indexed: 01/15/2023]
Abstract
Proteasome inhibitors have revolutionized outcomes in multiple myeloma, but they are used empirically, and primary and secondary resistance are emerging problems. We have identified TJP1 as a determinant of plasma cell proteasome inhibitor susceptibility. TJP1 suppressed expression of the catalytically active immunoproteasome subunits LMP7 and LMP2, decreased proteasome activity, and enhanced proteasome inhibitor sensitivity in vitro and in vivo. This occurred through TJP1-mediated suppression of EGFR/JAK1/STAT3 signaling, which modulated LMP7 and LMP2 levels. In the clinic, high TJP1 expression in patient myeloma cells was associated with a significantly higher likelihood of responding to bortezomib and a longer response duration, supporting the use of TJP1 as a biomarker to identify patients most likely to benefit from proteasome inhibitors.
Collapse
Affiliation(s)
- Xing-Ding Zhang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu 215123, China; Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, China
| | | | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George Mulligan
- Millennium: The Takeda Oncology Company, Cambridge, MA 02139, USA
| | - Bin Li
- Millennium: The Takeda Oncology Company, Cambridge, MA 02139, USA
| | | | - Lin Qi
- Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, China
| | - Jianliang Xu
- Institute of Molecular and Cell Biology, Singapore 138673, Republic of Singapore
| | - Walter Hunziker
- Institute of Molecular and Cell Biology, Singapore 138673, Republic of Singapore
| | - Bart Barlogie
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Saad Z Usmani
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Hematologic Oncology, Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC 28204, USA
| | - Qing Zhang
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Hematologic Oncology, Levine Cancer Institute, Carolinas Healthcare System, Charlotte, NC 28204, USA
| | - John Crowley
- Cancer Research and Biostatistics, Seattle, WA 98101, USA
| | - Antje Hoering
- Cancer Research and Biostatistics, Seattle, WA 98101, USA
| | - Jatin J Shah
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Donna M Weber
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sheeba K Thomas
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bing-Zong Li
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui-Han Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiexin Zhang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Isere Kuiatse
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jin-Le Tang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu 215123, China
| | - Hua Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jin He
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jing Yang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Enrico Milan
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan 20132, Italy
| | - Simone Cenci
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan 20132, Italy
| | - Wen-Cai Ma
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhi-Qiang Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard Eric Davis
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lin Yang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu 215123, China; Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, China.
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
120
|
Mitsiades CS. Tight Junction Protein 1: New Insights into Proteasome Inhibitor Resistance and Myeloma Pathophysiology. Cancer Cell 2016; 29:611-612. [PMID: 27165736 DOI: 10.1016/j.ccell.2016.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In this issue of Cancer Cell, Zhang et al. report that TJP1 suppresses EGFR/JAK1/STAT3-mediated signaling and increases the proteasome inhibitor sensitivity of myeloma cells by altering the cellular proteasome capacity versus proteasome load of undegraded intracellular proteins.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
121
|
Influence of Photodynamic Therapy on Apoptosis and Invasion of Human Cholangiocarcinoma QBC939 Cell Line. ACTA ACUST UNITED AC 2016; 30:252-9. [PMID: 26960307 DOI: 10.1016/s1001-9294(16)30009-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To investigate the effect of photodynamic therapy (PDT) mediated by hematoporphyrin derivative (HPD) on apoptosis and invasion of cholangiocarcinoma QBC939 cell lines. METHODS In vitro cultured cholangiocarcinoma QBC939 cell line was exposed to 2, 4, 6, 8, 10, 12, and 14 μg/ml HPD with 5, 10, and 15 J/cm2 light intensity, respectively. The optical density at 450 nm of the QBC939 cells was measured by CCK8 assay and its growth inhibition ratio was calculated. Flow cytometry and transwell migration assay were applied to detect cell apoptosis and invasion respectively. RT-PCR and immunocytochemistry analyses were used to detect expressions of vascular endothelial growth factor-C (VEGF-C), cyclooxygenase-2 (COX-2), and proliferating cell nuclear antigen (PCNA). Enzyme-linked immunosorbent assay (ELISA) was carried out to examine the secretion of VEGF-C and COX-2 in QBC939 cells. RESULTS Exposure to HPD-PDT can significantly suppress the growth of QBC939 cells (all P<0.05). HPD-PDT can promote apoptosis of QBC939 cells at the early stage. When the concentration of HPD was 2 μg/ml and light irradiation was 5 J/cm2, HPD-PDT had no obvious inhibitory effect on QBC939 cell growth, but can obviously inhibit cell invasion, and significant difference was observed between the HPD-PDT and control groups (P<0.01). The HPD-PDT can reduce the mRNA and protein expressions of VEGF-C, COX-2, and PCNA, and decrease the secretion of VEGF-C and COX-2 in QBC939 cells. CONCLUSION PDT could promote apoptosis and inhibit growth and invasion of cholangiocarcinoma cells QBC939 in vitro.
Collapse
|
122
|
Beyar-Katz O, Magidey K, Ben-Tsedek N, Alishekevitz D, Timaner M, Miller V, Lindzen M, Yarden Y, Avivi I, Shaked Y. Bortezomib-induced pro-inflammatory macrophages as a potential factor limiting anti-tumour efficacy. J Pathol 2016; 239:262-73. [DOI: 10.1002/path.4723] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 03/20/2016] [Accepted: 03/23/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Ofrat Beyar-Katz
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
- Department of Haematology and BMT; Rambam Health Care Campus; Haifa Israel
| | - Ksenia Magidey
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
| | - Neta Ben-Tsedek
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
| | - Dror Alishekevitz
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
| | - Michael Timaner
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
| | - Valeria Miller
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
| | - Moshit Lindzen
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| | - Yosef Yarden
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| | - Irit Avivi
- Department of Haematology and BMT; Rambam Health Care Campus; Haifa Israel
| | - Yuval Shaked
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine; Technion-Israel Institute of Technology; Haifa Israel
| |
Collapse
|
123
|
Abdel Malek MAY, Jagannathan S, Malek E, Sayed DM, Elgammal SA, Abd El-Azeem HG, Thabet NM, Driscoll JJ. Molecular chaperone GRP78 enhances aggresome delivery to autophagosomes to promote drug resistance in multiple myeloma. Oncotarget 2016; 6:3098-110. [PMID: 25605012 PMCID: PMC4413640 DOI: 10.18632/oncotarget.3075] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/17/2014] [Indexed: 11/25/2022] Open
Abstract
Despite the clinical benefit of the proteasome inhibitor bortezomib, multiple myeloma (MM) patients invariably relapse through poorly defined mechanisms. Myeloma cells inevitably develop chemoresistance that leads to disease relapse and patient-related deaths. Studies in tumor cell lines and biopsies obtained from patients refractory to therapy have revealed that myeloma cells adapt to stress by inducing expression of glucose-regulated protein 78 (GRP78), an endoplasmic reticulum (ER) chaperone with anti-apoptotic properties. Treatment of myeloma cells with bortezomib increased GRP78 levels and activated GRP78-dependent autophagy. Expression profiling indicated that GRP78-encoding HSPA5 was significantly upregulated in bortezomib-resistant cells. Co-treatment with the anti-diabetic agent metformin suppressed GRP78 and enhanced the anti-proliferative effect of bortezomib. Bortezomib treatment led to GRP78 co-localization with proteotoxic protein aggregates, known as aggresomes. Pharmacologic suppression, genetic ablation or mutational inactivation of GRP78 followed by bortezomib treatment led to the accumulation of aggresomes but impaired autophagy and enhanced anti-myeloma effect of bortezomib. GRP78 was co-immunoprecipitated with the KDEL receptor, an ER quality control regulator that binds proteins bearing the KDEL motif to mediate their retrieval from the Golgi complex back to the ER. Taken together, we demonstrate that inhibition of GRP78 functional activity disrupts autophagy and enhances the anti-myeloma effect of bortezomib.
Collapse
Affiliation(s)
- Mohamed A Y Abdel Malek
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sajjeev Jagannathan
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ehsan Malek
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Douaa M Sayed
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Sahar A Elgammal
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Hanan G Abd El-Azeem
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nabila M Thabet
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - James J Driscoll
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,University of Cincinnati Cancer Institute, Cincinnati, OH, USA
| |
Collapse
|
124
|
Mehdad A, Xavier Reis G, Souza AA, Barbosa JARG, Ventura MM, de Freitas SM. A Bowman-Birk inhibitor induces apoptosis in human breast adenocarcinoma through mitochondrial impairment and oxidative damage following proteasome 20S inhibition. Cell Death Discov 2016; 2:15067. [PMID: 27551492 PMCID: PMC4979482 DOI: 10.1038/cddiscovery.2015.67] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/19/2015] [Accepted: 12/03/2015] [Indexed: 12/29/2022] Open
Abstract
Proteasome inhibitors are emerging as a new class of chemopreventive agents and have gained huge importance as potential pharmacological tools in breast cancer treatment. Improved understanding of the role played by proteases and their specific inhibitors in humans offers novel and challenging opportunities for preventive and therapeutic intervention. In this study, we demonstrated that the Bowman-Birk protease inhibitor from Vigna unguiculata seeds, named black-eyed pea trypsin/chymotrypsin Inhibitor (BTCI), potently suppresses human breast adenocarcinoma cell viability by inhibiting the activity of proteasome 20S. BTCI induced a negative growth effect against a panel of breast cancer cells, with a concomitant cytostatic effect at the G2/M phase of the cell cycle and an increase in apoptosis, as observed by an augmented number of cells at the sub-G1 phase and annexin V-fluorescin isothiocyanate (FITC)/propidium iodide (PI) staining. In contrast, BTCI exhibited no cytotoxic effect on normal mammary epithelial cells. Moreover, the increased levels of intracellular reactive oxygen species (ROS) and changes in the mitochondrial membrane potential in cells treated with BTCI indicated mitochondrial damage as a crucial cellular event responsible for the apoptotic process. The higher activity of caspase in tumoral cells treated with BTCI in comparison with untreated cells suggests that BTCI induces apoptosis in a caspase-dependent manner. BTCI affected NF-kB target gene expression in both non invasive and invasive breast cancer cell lines, with the effect highly pronounced in the invasive cells. An increased expression of interleukin-8 (IL-8) in both cell lines was also observed. Taken together, these results suggest that BTCI promotes apoptosis through ROS-induced mitochondrial damage following proteasome inhibition. These findings highlight the pharmacological potential and benefit of BTCI in breast cancer treatment.
Collapse
Affiliation(s)
- A Mehdad
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - Giselle Xavier Reis
- Faculty of Medicine, Department of Molecular
Pathology, University of Brasilia, Brasilia, Brazil
| | - AA Souza
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - JARG Barbosa
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - MM Ventura
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| | - SM de Freitas
- Laboratory of Molecular Biophysics, Institute
of Biological Sciences, University of Brasilia, Brasilia,
Brazil
| |
Collapse
|
125
|
Shah JJ, Feng L, Thomas SK, Berkova Z, Weber DM, Wang M, Qazilbash MH, Champlin RE, Mendoza TR, Cleeland C, Orlowski RZ. Siltuximab (CNTO 328) with lenalidomide, bortezomib and dexamethasone in newly-diagnosed, previously untreated multiple myeloma: an open-label phase I trial. Blood Cancer J 2016; 6:e396. [PMID: 26871714 PMCID: PMC4771967 DOI: 10.1038/bcj.2016.4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/04/2016] [Indexed: 01/01/2023] Open
Abstract
The safety and efficacy of siltuximab (CNTO 328) was tested in combination with lenalidomide, bortezomib and dexamethasone (RVD) in patients with newly-diagnosed, previously untreated symptomatic multiple myeloma. Fourteen patients were enrolled in the study, eleven of whom qualified to receive therapy. A majority of patients (81.8%) completed the minimal number or more of the four required cycles, while two patients completed only three cycles. The maximum tolerated dose (MTD) of siltuximab with RVD was dose level -1 (siltuximab: 8.3 mg/kg; bortezomib: 1.3 mg/m(2); lenalidomide: 25 mg; dexamethasone: 20 mg). Serious adverse events were grade 3 pneumonia and grade 4 thrombocytopenia, and no deaths occurred during the study or with follow-up (median follow-up 28.1 months). An overall response rate, after 3-4 cycles of therapy, of 90.9% (95% confidence interval (CI): 58.7%, 99.8%) (9.1% complete response (95% CI: 0.2%, 41.3%), 45.5% very good partial response (95% CI: 16.7%, 76.6%) and 36.4% partial response (95% CI: 10.9%, 69.2%)) was seen. Two patients withdrew consent, and nine patients (81.8%) opted for autologous stem cell transplantation.
Collapse
Affiliation(s)
- J J Shah
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S K Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Z Berkova
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D M Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M H Qazilbash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R E Champlin
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T R Mendoza
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C Cleeland
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
126
|
Development of acute pulmonary hypertension after bortezomib treatment in a patient with multiple myeloma: a case report and the review of the literature. Am J Ther 2016; 22:e88-92. [PMID: 24100255 DOI: 10.1097/01.mjt.0000433941.91996.5f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Bortezomib is widely used in treatment of multiple myeloma. In recent years, severe bortezomib-induced lung injury has been reported. The clinical course is generally characterized with fever and dyspnea, followed by respiratory failure with pulmonary infiltrates. Herein, we report a 57-year-old man with newly diagnosed multiple myeloma admitted with dyspnea, fever, and hypotension on the third day of the first dose of bortezomib therapy. He had bilateral jugular venous distention, crackles at the bases of the lungs and hepatomegaly. Transthoracic echocardiography revealed acute pulmonary hypertension (PH) with an estimated pressure of 70 mm Hg. The perfusion scintigraphy ruled out pulmonary embolism, and microbiological examination was negative. On his course, fever, dyspnea, hypoxia, and pulmonary vascular pressure subsided rapidly. The sudden onset of PH and its rapid decrement without any treatment suggests bortezomib as the underlying cause. Subsequently, the patient did not respond to vincristine-doxorubicin-dexamethasone regimen and thalidomide. Bortezomib treatment was repeated, and no pulmonary adverse reactions occurred. Follow-up echocardiographies revealed pulmonary arterial pressures to be maximally of 35 mm Hg. To our knowledge, this is the first case of acute PH after front-line bortezomib therapy. In this report, we review bortezomib-related pulmonary complications in the literature and possible underlying mechanisms.
Collapse
|
127
|
de Moreuil C, Ianotto JC, Eveillard JR, Carrier M, Delluc A. [Multiple myeloma and venous thrombosis. Which thromboprophylaxis should be given?]. Rev Med Interne 2016; 37:473-9. [PMID: 26833146 DOI: 10.1016/j.revmed.2015.12.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/24/2015] [Accepted: 12/29/2015] [Indexed: 01/06/2023]
Abstract
Multiple myeloma is a malignant plasma cells dyscrasia that mainly affects patients older than 65 years. These patients are at a higher risk for venous thromboembolism (VTE) because of cancer status, intrinsic risk factors, and exposure to prothrombotic therapies. The risk for VTE appears higher during the first months of myeloma treatment and decreases over time. Exposure to immunomodulatory drugs (IMIDs) such as thalidomide or lenalidomide in association with high doses of dexamethasone or anthracyclin-based chemotherapy is associated with a four-fold increased risk for VTE. Low-dose aspirin, preventive-dose of low molecular weight heparin (LMWH) or vitamin K antagonists were tested for primary prevention of VTE in myeloma patients receiving chemotherapy. The International Myeloma Working Group (IMWG) suggests stratifying VTE risk to decide which patients should receive VTE prevention. Then, the IMWG suggests giving low-dose aspirin to low VTE risk patients and LMWH or vitamin K antagonists to patients at high risk for VTE. For daily practice, it seems reasonable to start preventive doses of LMWH for 3 to 6 months in ambulatory myeloma patients receiving combined therapy with IMID and in all myeloma patients admitted to hospital.
Collapse
Affiliation(s)
- C de Moreuil
- EA 3878 (GETBO), département de médecine interne et de pneumologie, hôpital de la Cavale-Blanche, CHRU de Brest, 29609 Brest cedex, France
| | - J-C Ianotto
- EA 3878 (GETBO), département de médecine interne et de pneumologie, hôpital de la Cavale-Blanche, CHRU de Brest, 29609 Brest cedex, France; Service d'hématologie clinique, institut de cancéro-hématologie, hôpital Morvan, CHRU de Brest, 29609 Brest cedex, France
| | - J-R Eveillard
- Service d'hématologie clinique, institut de cancéro-hématologie, hôpital Morvan, CHRU de Brest, 29609 Brest cedex, France
| | - M Carrier
- Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, 501, Smyth Road, K1H 8 L6 Ottawa, Ontario, Canada
| | - A Delluc
- EA 3878 (GETBO), département de médecine interne et de pneumologie, hôpital de la Cavale-Blanche, CHRU de Brest, 29609 Brest cedex, France.
| |
Collapse
|
128
|
Yong K, Cavet J, Johnson P, Morgan G, Williams C, Nakashima D, Akinaga S, Oakervee H, Cavenagh J. Phase I study of KW-2478, a novel Hsp90 inhibitor, in patients with B-cell malignancies. Br J Cancer 2015; 114:7-13. [PMID: 26695442 PMCID: PMC4716540 DOI: 10.1038/bjc.2015.422] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/12/2015] [Accepted: 10/26/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND KW-2478 is a novel, non-ansamycin, non-purine heat-shock protein 90 (Hsp90) inhibitor. METHODS In this phase I, multicentre study, KW-2478 was administered intravenously over 1 h at doses ranging from 14 to 176 mg m(-2) once daily on days 1-5 of a 14-day cycle in a standard 3+3 design in 27 patients (22 with multiple myeloma and 5 with non-Hodgkin lymphoma). Patients enrolled had relapsed/refractory disease previously treated with ⩾2 regimens. RESULTS There were no dose-limiting toxicities, thus the maximum-tolerated dose was not reached. KW-2478 was well tolerated and did not manifest significant retinal or ocular toxicity. The most common treatment-related adverse events were diarrhoea (33.3%), fatigue (29.6%), headache (25.9%), hypertension (22.2%), nausea (14.8%), vomiting (7.4%), and dizziness (7.4%). Plasma concentrations peaked at the end of infusion and decayed in a biphasic manner with a terminal half-life of ∼6 h. Target inhibition was inferred from the increase in Hsp70 levels in peripheral blood mononuclear cells at doses ⩾71 mg m(-2). Twenty-four of 25 (96%) evaluable patients showed stable disease, with five being free of disease progression for ⩾6 months. CONCLUSIONS Preliminary clinical response data were encouraging and warrant further investigation of KW-2478 in combination regimens for relapsed/refractory B-cell malignancies.
Collapse
Affiliation(s)
- K Yong
- UCL Cancer Institute, University College London, Huntley Street, London WC1E 6DD, UK
| | - J Cavet
- Department of Haematology, Christie Hospital/University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - P Johnson
- Cancer Research UK Clinical Centre, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - G Morgan
- Myeloma Institute for Research and Therapy, West Markham Street, Little Rock, AR 72205, USA
| | - C Williams
- Centre for Clinical Haematology, Nottingham University Hospital, Hucknall Road, Nottingham NG5 1PB, UK
| | - D Nakashima
- Kyowa Hakko Kirin Pharma Inc., Princeton, NJ, USA
| | - S Akinaga
- Kyowa Hakko Kirin Co. Ltd, Tokyo Research Triangle Park, Tokyo 194-8533, Japan
| | - H Oakervee
- Department of Haematology, St. Bartholomew's Hospital, West Smithfield, London SE24 9LG, UK
| | - J Cavenagh
- Department of Haematology, St. Bartholomew's Hospital, West Smithfield, London SE24 9LG, UK
| |
Collapse
|
129
|
Mechanisms of Drug Resistance in Relapse and Refractory Multiple Myeloma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:341430. [PMID: 26649299 PMCID: PMC4663284 DOI: 10.1155/2015/341430] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/24/2015] [Accepted: 10/21/2015] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable because most patients eventually relapse or become refractory to current treatments. Although the treatments have improved, the major problem in MM is resistance to therapy. Clonal evolution of MM cells and bone marrow microenvironment changes contribute to drug resistance. Some mechanisms affect both MM cells and microenvironment, including the up- and downregulation of microRNAs and programmed death factor 1 (PD-1)/PD-L1 interaction. Here, we review the pathogenesis of MM cells and bone marrow microenvironment and highlight possible drug resistance mechanisms. We also review a potential molecular targeting treatment and immunotherapy for patients with refractory or relapse MM.
Collapse
|
130
|
Vallumsetla N, Paludo J, Kapoor P. Bortezomib in mantle cell lymphoma: comparative therapeutic outcomes. Ther Clin Risk Manag 2015; 11:1663-74. [PMID: 26609233 PMCID: PMC4644179 DOI: 10.2147/tcrm.s72943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mantle cell lymphoma (MCL) is an incurable, typically aggressive subtype of non-Hodgkin lymphoma, accounting for 4%–7% of newly diagnosed non-Hodgkin lymphoma cases. Chemoresistance commonly ensues in MCL, and patients with this heterogeneous disease invariably relapse, underscoring the unmet need for better therapies. Over the past few years, several novel agents with promising activity and unique mechanisms of action have been deemed effective in MCL. Bortezomib is a reversible proteasome inhibitor, approved as a single agent for patients with relapsed/refractory MCL who have received at least one prior line of therapy. Addition of bortezomib to chemoimmunotherapies has demonstrated good tolerability and superior efficacy, both in the upfront and salvage settings, and recently one such combination of bortezomib plus rituximab, cyclophosphamide, doxorubicin, and prednisone was approved as a frontline regimen in untreated patients with MCL. This review examines the role of bortezomib in a multitude of clinical settings and ongoing clinical trials designed to optimize its integration in the current treatment paradigms of MCL.
Collapse
Affiliation(s)
- Nishanth Vallumsetla
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jonas Paludo
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Prashant Kapoor
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
131
|
Das DS, Ray A, Song Y, Richardson P, Trikha M, Chauhan D, Anderson KC. Synergistic anti-myeloma activity of the proteasome inhibitor marizomib and the IMiD immunomodulatory drug pomalidomide. Br J Haematol 2015; 171:798-812. [PMID: 26456076 DOI: 10.1111/bjh.13780] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/13/2015] [Indexed: 12/22/2022]
Abstract
The proteasome inhibitor bortezomib is an effective therapy for the treatment of relapsed and refractory multiple myeloma (RRMM); however, prolonged treatment can be associated with toxicity, peripheral neuropathy and drug resistance. Our earlier studies showed that the novel proteasome inhibitor marizomib is distinct from bortezomib in its chemical structure, mechanisms of action and effects on proteasomal activities, and that it can overcome bortezomib resistance. Pomalidomide, like lenalidomide, has potent immunomodulatory activity and has been approved by the US Food and Drug Administration for the treatment of RRMM. Here, we demonstrate that combining low concentrations of marizomib with pomalidomide induces synergistic anti-MM activity. Marizomib plus pomalidomide-induced apoptosis is associated with: (i) activation of caspase-8, caspase-9, caspase-3 and PARP cleavage, (ii) downregulation of cereblon (CRBN), IRF4, MYC and MCL1, and (iii) suppression of chymotrypsin-like, caspase-like, and trypsin-like proteasome activities. CRBN-siRNA attenuates marizomib plus pomalidomide-induced MM cells death. Furthermore, marizomib plus pomalidomide inhibits the migration of MM cells and tumour-associated angiogenesis, as well as overcomes cytoprotective effects of bone marrow microenvironment. In human MM xenograft model studies, the combination of marizomib and pomalidomide is well tolerated, inhibits tumour growth and prolongs survival. These preclinical studies provide the rationale for on-going clinical trials of combined marizomib and pomalidomide to improve outcome in patients with RRMM.
Collapse
Affiliation(s)
- Deepika S Das
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Arghya Ray
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yan Song
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Paul Richardson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Dharminder Chauhan
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
132
|
Abstract
Mantle cell lymphoma (MCL) is a hematological malignancy with unfavorable prognosis. Bortezomib, a potent, selective and reversible inhibitor of the 26S proteasome, was shown to be active in MCL and is currently implemented in therapeutic combinations. Single-agent bortezomib has demonstrated clinical efficacy in relapsed and refractory MCL with objective response in up to 47% of the patients. However, complete remission rates are low and duration of response is relatively short. In previously untreated patients, the addition of bortezomib to induction chemotherapy is also promising. Further evaluation of bortezomib alone or in combination with other drugs for the treatment of MCL is warranted to improve the quality of life and survival of patients. This review explores bortezomib as therapy in patients with MCL.
Collapse
Affiliation(s)
- Tadeusz Robak
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, ul. Ciołkowskiego 2, Poland
| |
Collapse
|
133
|
Howland SW, Ng GXP, Chia SK, Rénia L. Investigating proteasome inhibitors as potential adjunct therapies for experimental cerebral malaria. Parasite Immunol 2015; 37:599-604. [PMID: 26366636 DOI: 10.1111/pim.12277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/08/2015] [Indexed: 12/12/2022]
Abstract
Aside from antimalarials, there is currently no treatment for cerebral malaria, a fulminant neurological complication of P. falciparum infection that is a leading cause of death in African children. In the mouse model of cerebral malaria, cross-presentation of parasite antigens by brain endothelial cells is thought to be a crucial late step in pathogenesis. We have investigated three proteasome inhibitors as potential adjunct therapies: bortezomib, carfilzomib and ONX-0914. Only carfilzomib, an irreversible inhibitor of both constitutive proteasomes and immunoproteasomes, was able to inhibit cross-presentation of malaria antigen by murine brain endothelial cells in vitro. To mimic the clinical setting, carfilzomib was co-administered with artesunate only when infected mice exhibited neurological defects. However, there was no improvement in survival compared to artesunate monotherapy. The treatment failure was explained by the inability of daily or twice daily bolus doses of carfilzomib to inhibit cross-presentation by brain endothelial cells in vivo. We also report here that bortezomib, which has been associated with neurological adverse events, accelerated death in ECM-infected mice. Future investigations of proteasome inhibitors for modulating cross-presentation during malaria infection should focus on sustained and targeted delivery to brain endothelial cells.
Collapse
Affiliation(s)
- S W Howland
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - G X P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - S K Chia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | - L Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
134
|
Overexpression of salivary-type amylase reduces the sensitivity to bortezomib in multiple myeloma cells. Int J Hematol 2015; 102:569-78. [DOI: 10.1007/s12185-015-1859-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 10/23/2022]
|
135
|
Lee KH, Jang AH, Yoo CG. 17-Allylamino-17-Demethoxygeldanamycin and the Enhancement of PS-341–Induced Lung Cancer Cell Death by Blocking the NF-κB and PI3K/Akt Pathways. Am J Respir Cell Mol Biol 2015; 53:412-421. [DOI: 10.1165/rcmb.2014-0186oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Kyoung-Hee Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and Medical Research Center, Seoul National University College of Medicine, Seoul South Korea
| | - An-Hee Jang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and Medical Research Center, Seoul National University College of Medicine, Seoul South Korea
| | - Chul-Gyu Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and Medical Research Center, Seoul National University College of Medicine, Seoul South Korea
| |
Collapse
|
136
|
Abstract
Under stressful conditions, the heat shock protein 90 (HSP90) molecular chaperone protects cellular proteins (client proteins) from degradation via the ubiquitin-proteasome pathway. HSP90 expression is upregulated in cancers, and this contributes to the malignant phenotype of increased proliferation and decreased apoptosis and maintenance of metastatic potential via conservation of its client proteins, including human epidermal growth factor receptor 2, anaplastic lymphoma kinase, androgen receptor, estrogen receptor, Akt, Raf-1, cell cycle proteins, and B-cell lymphoma 2 among others. Hence, inhibition of HSP90 leads to the simultaneous degradation of its many clients, thereby disrupting multiple oncogenic signaling cascades. This has sparked tremendous interest in the development of HSP90 inhibitors as an innovative anticancer strategy. Based on the wealth of compelling data from preclinical studies, a number of HSP90 inhibitors have entered into clinical testing. However, despite enormous promise and anticancer activity reported to date, none of the HSP90 inhibitors in development has been approved for cancer therapy, and the full potential of this class of agents is yet to be realized. This article provides a review on ganetespib, a small molecule HSP90 inhibitor that is currently under evaluation in a broad range of cancer types in combination with other therapeutic agents with the hope of further enhancing its efficacy and overcoming drug resistance. Based on our current understanding of the complex HSP90 machinery combined with the emerging data from these key clinical trials, ganetespib has the potential to be the first-in-class HSP90 inhibitor to be approved as a new anticancer therapy.
Collapse
Affiliation(s)
- Komal Jhaveri
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shanu Modi
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
137
|
Shah SP, Lonial S, Boise LH. When Cancer Fights Back: Multiple Myeloma, Proteasome Inhibition, and the Heat-Shock Response. Mol Cancer Res 2015; 13:1163-73. [PMID: 26013169 DOI: 10.1158/1541-7786.mcr-15-0135] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/13/2015] [Indexed: 01/01/2023]
Abstract
Multiple myeloma is a plasma cell malignancy with an estimated 26,850 new cases and 11,240 deaths in 2015 in the United States. Two main classes of agents are the mainstays of therapy-proteasome inhibitors (PI) and immunomodulatory drugs (IMiD). Other new targets are emerging rapidly, including monoclonal antibodies and histone deacetylase (HDAC) inhibitors. These therapeutic options have greatly improved overall survival, but currently only 15% to 20% of patients experience long-term progression-free survival or are cured. Therefore, improvement in treatment options is needed. One potential means of improving clinical options is to target resistance mechanisms for current agents. For example, eliminating the cytoprotective heat-shock response that protects myeloma cells from proteasome inhibition may enhance PI-based therapies. The transcription factor heat-shock factor 1 (HSF1) is the master regulator of the heat-shock response. HSF1 is vital in the proteotoxic stress response, and its activation is controlled by posttranslational modifications (PTM). This review details the mechanisms of HSF1 regulation and discusses leveraging that regulation to enhance PI activity.
Collapse
Affiliation(s)
- Shardule P Shah
- Department of Hematology and Medical Oncology, Winship, Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, Georgia
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship, Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, Georgia
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship, Cancer Institute of Emory University and the Emory University School of Medicine, Atlanta, Georgia. Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
138
|
Teicher BA, Tomaszewski JE. Proteasome inhibitors. Biochem Pharmacol 2015; 96:1-9. [PMID: 25935605 DOI: 10.1016/j.bcp.2015.04.008] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/16/2015] [Indexed: 12/20/2022]
Abstract
Proteasome inhibitors have a 20 year history in cancer therapy. The first proteasome inhibitor, bortezomib (Velcade, PS-341), a break-through multiple myeloma treatment, moved rapidly through development from bench in 1994 to first approval in 2003. Bortezomib is a reversible boronic acid inhibitor of the chymotrypsin-like activity of the proteasome. Next generation proteasome inhibitors include carfilzomib and oprozomib which are irreversible epoxyketone proteasome inhibitors; and ixazomib and delanzomib which are reversible boronic acid proteasome inhibitors. Two proteasome inhibitors, bortezomib and carfilzomib are FDA approved drugs and ixazomib and oprozomib are in late stage clinical trials. All of the agents are potent cytotoxics. The disease focus for all the proteasome inhibitors is multiple myeloma. This focus arose from clinical observations made in bortezomib early clinical trials. Later preclinical studies confirmed that multiple myeloma cells were indeed more sensitive to proteasome inhibitors than other tumor cell types. The discovery and development of the proteasome inhibitor class of anticancer agents has progressed through a classic route of serendipity and scientific investigation. These agents are continuing to have a major impact in their treatment of hematologic malignancies and are beginning to be explored as potential treatment agent for non-cancer indications.
Collapse
Affiliation(s)
- Beverly A Teicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, United States.
| | - Joseph E Tomaszewski
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, United States
| |
Collapse
|
139
|
Sato A. The human immunodeficiency virus protease inhibitor ritonavir is potentially active against urological malignancies. Onco Targets Ther 2015; 8:761-8. [PMID: 25914545 PMCID: PMC4399512 DOI: 10.2147/ott.s79776] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The human immunodeficiency virus protease inhibitor ritonavir has recently been shown to have antineoplastic activity, and its use in urological malignancies is under investigation with an eye toward drug repositioning. Ritonavir is thought to exert its antineoplastic activity by inhibiting multiple signaling pathways, including the Akt and nuclear factor-kappaB pathways. It can increase the amount of unfolded proteins in the cell by inhibiting both the proteasome and heat shock protein 90. Combinations of ritonavir with agents that increase the amount of unfolded proteins, such as proteasome inhibitors, histone deacetylase inhibitors, or heat shock protein 90 inhibitors, therefore, induce endoplasmic reticulum stress cooperatively and thereby kill cancer cells effectively. Ritonavir is also a potent cytochrome P450 3A4 and P-glycoprotein inhibitor, increasing the intracellular concentration of combined drugs by inhibiting their degradation and efflux from cancer cells and thereby enhancing their antineoplastic activity. Furthermore, riotnavir’s antineoplastic activity includes modulation of immune system activity. Therapies using ritonavir are thus an attractive new approach to cancer treatment and, due to their novel mechanisms of action, are expected to be effective against malignancies that are refractory to current treatment strategies. Further investigations using ritonavir are expected to find new uses for clinically available drugs in the treatment of urological malignancies as well as many other types of cancer.
Collapse
Affiliation(s)
- Akinori Sato
- Department of Urology, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
140
|
Muñoz-Galván S, Gutierrez G, Perez M, Carnero A. MAP17 (PDZKIP1) Expression Determines Sensitivity to the Proteasomal Inhibitor Bortezomib by Preventing Cytoprotective Autophagy and NFκB Activation in Breast Cancer. Mol Cancer Ther 2015; 14:1454-65. [PMID: 25837675 DOI: 10.1158/1535-7163.mct-14-1053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/24/2015] [Indexed: 11/16/2022]
Abstract
MAP17 is a small nonglycosylated membrane protein that is overexpressed in a high percentage of carcinomas. High levels of MAP17 enhance the tumorigenic properties of tumor cells by increasing oxidative stress, which is dependent on Na(+)-coupled cotransport. Here, we show that MAP17 is associated with proteins involved in protein degradation and that proteasome inhibition induces autophagy. To analyze whether MAP17 could also alter this process, we used the proteasome inhibitor bortezomib (Velcade, PS-341), which is approved for the treatment of multiple myeloma and mantle cell lymphoma, although it has a high rate of resistance emergence and poor efficacy in solid tumors. We provide evidence that bortezomib induces a cytoprotective effect by activating autophagy and NFκB nuclear translocation, responses that are repressed in the presence of high levels of MAP17 both in vitro and in vivo. Indeed, patients with multiple myeloma treated with bortezomib showed higher response rates and a longer time to progression associated with increased levels of MAP17 expression. The MAP17-induced sensitivity to bortezomib is dependent on the oxidative status of the cells and the activity of Na(+)-coupled transporters because treatment with antioxidants or the inhibitor furosemide restores the cytoprotective activity induced by bortezomib. Therefore, bortezomib induces a prosurvival response through cytoprotective autophagy and NFκB nuclear translocation, which is repressed by high levels of MAP17. We propose that the levels of MAP17 could be used as a prognostic marker to predict the response to bortezomib in hematologic malignancies and in other tissues that are not commonly responsive to the drug.
Collapse
Affiliation(s)
- Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | | | - Marco Perez
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocio/Universidad de Sevilla/Consejo Superior de Investigaciones Cientificas, Seville, Spain.
| |
Collapse
|
141
|
Mitsiades CS. Therapeutic landscape of carfilzomib and other modulators of the ubiquitin-proteasome pathway. J Clin Oncol 2015; 33:782-5. [PMID: 25605842 PMCID: PMC4517049 DOI: 10.1200/jco.2014.55.5748] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
142
|
Li F, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. NF-κB in cancer therapy. Arch Toxicol 2015; 89:711-31. [PMID: 25690730 DOI: 10.1007/s00204-015-1470-4] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor kappa B (NF-κB) has attracted increasing attention in the field of cancer research from last few decades. Aberrant activation of this transcription factor is frequently encountered in a variety of solid tumors and hematological malignancies. NF-κB family members and their regulated genes have been linked to malignant transformation, tumor cell proliferation, survival, angiogenesis, invasion/metastasis, and therapeutic resistance. In this review, we highlight the diverse molecular mechanism(s) by which the NF-κB pathway is constitutively activated in different types of human cancers, and the potential role of various oncogenic genes regulated by this transcription factor in cancer development and progression. Additionally, various pharmacological approaches employed to target the deregulated NF-κB signaling pathway, and their possible therapeutic potential in cancer therapy is also discussed briefly.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, Cancer Science Institute, National University of Singapore, Singapore, 117597, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Huang Z, Peng S, Knoff J, Lee SY, Yang B, Wu TC, Hung CF. Combination of proteasome and HDAC inhibitor enhances HPV16 E7-specific CD8+ T cell immune response and antitumor effects in a preclinical cervical cancer model. J Biomed Sci 2015; 22:7. [PMID: 25591912 PMCID: PMC4298946 DOI: 10.1186/s12929-014-0111-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 12/31/2014] [Indexed: 11/15/2022] Open
Abstract
Background Bortezomib, a proteasome inhibitor and suberoylanilide hydroxamic acid (SAHA, also known as Vorinostat), a histone deacetylase inhibitor, have been recognized as potent chemotherapeutic drugs. Bortezomib and SAHA are FDA-approved for the treatment of cutaneous T cell lymphoma and multiple myeloma/mantle cell lymphoma, respectively. Furthermore, the combination of the bortezomib and SAHA has been tested in a variety of preclinical models and in clinical trials and may be ideal for the treatment of cancer. However, it remains unclear how this treatment strategy affects the host immune response against tumors. Results Here, we used a well-defined E6/E7-expressing tumor model to examine how the immune system can be motivated to act against tumor cells expressing tumor antigens. We demonstrate that the combination of bortezomib and SAHA elicits potent antitumor effects in TC-1 tumor-bearing mice. Additionally, we are the first to show that treatment with bortezomib and SAHA leads to tumor-specific immunity by rendering tumor cells more susceptible to killing by antigen-specific CD8+ T cells than treatment with either drug alone. Conclusions The current study serves an important foundation for the future clinical application of both drugs for the treatment of cervical cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12929-014-0111-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhuomin Huang
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Department of Gynecology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| | - Shiwen Peng
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Jayne Knoff
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Sung Yong Lee
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Department of Internal Medicine, Korea University Medical Center, Seoul, South Korea.
| | - Benjamin Yang
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| | - Tzyy-Choou Wu
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA. .,Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, USA. .,Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, USA. .,Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, CRB II Room 307, 1550 Orleans Street, 21231, Baltimore, MD, USA.
| |
Collapse
|
144
|
Murray MY, Zaitseva L, Auger MJ, Craig JI, MacEwan DJ, Rushworth SA, Bowles KM. Ibrutinib inhibits BTK-driven NF-κB p65 activity to overcome bortezomib-resistance in multiple myeloma. Cell Cycle 2015; 14:2367-75. [PMID: 25565020 DOI: 10.1080/15384101.2014.998067] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Multiple Myeloma (MM) is a haematologic malignancy characterized by the accumulation of clonal plasma cells in the bone marrow. Over the last 10-15 y the introduction of the proteasome-inhibitor bortezomib has improved MM prognosis, however relapse due to bortezomib-resistance is inevitable and the disease, at present, remains incurable. To model bortezomib-resistant MM we generated bortezomib-resistant MM cell lines (n = 4 ) and utilised primary malignant plasma cells from patients relapsing after bortezomib treatment (n = 6 ). We identified enhanced Bruton's tyrosine kinase (BTK) activity in bortezomib-resistant MM cells and found that inhibition of BTK, either pharmacologically with ibrutinib (0.5 μM) or via lenti-viral miRNA-targeted BTK interference, re-sensitized previously bortezomib-resistant MM cells to further bortezomib therapy at a physiologically relevant concentration (5 nM). Further analysis of pro-survival signaling revealed a role for the NF-κB p65 subunit in MM bortezomib-resistance, thus a combination of BTK and NF-κB p65 inhibition, either pharmacologically or via further lenti-viral miRNA NF-κB p65 interference, also restored sensitivity to bortezomib, significantly reducing cell viability (37.5 ± 6 .9 %, ANOVA P ≤ 0 .001). Accordingly, we propose the clinical evaluation of a bortezomib/ibrutinib combination therapy, including in patients resistant to single-agent bortezomib.
Collapse
Affiliation(s)
- Megan Y Murray
- a Department of Molecular Haematology; Norwich Medical School ; University of East Anglia ; Norwich , UK
| | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
The debut of the proteasome inhibitor bortezomib (Btz; Velcade®) radically and immediately improved the treatment of multiple myeloma (MM), an incurable malignancy of the plasma cell. Therapeutic resistance is unavoidable, however, and represents a major obstacle to maximizing the clinical potential of the drug. To address this challenge, studies have been conducted to uncover the molecular mechanisms driving Btz resistance and to discover new targeted therapeutic strategies and combinations that restore Btz activity. This review discusses the literature describing molecular adaptations that confer Btz resistance with a primary disease focus on MM. Also discussed are the most recent advances in therapeutic strategies that overcome resistance, approaches that include redox-modulating agents, murine double minute 2 inhibitors, therapeutic monoclonal antibodies, and new epigenetic-targeted drugs like bromodomain and extra terminal domain inhibitors.
Collapse
Affiliation(s)
- Nathan G Dolloff
- Department of Cellular and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
146
|
Flaherty DP, Miller JR, Garshott DM, Hedrick M, Gosalia P, Li Y, Milewski M, Sugarman E, Vasile S, Salaniwal S, Su Y, Smith LH, Chung TDY, Pinkerton AB, Aubé J, Callaghan MU, Golden JE, Fribley AM, Kaufman RJ. Discovery of Sulfonamidebenzamides as Selective Apoptotic CHOP Pathway Activators of the Unfolded Protein Response. ACS Med Chem Lett 2014; 5:1278-1283. [PMID: 25530830 PMCID: PMC4266338 DOI: 10.1021/ml5003234] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/29/2014] [Indexed: 01/29/2023] Open
Abstract
![]()
Cellular proteins that fail to fold
properly result in inactive
or disfunctional proteins that can have toxic functions. The unfolded
protein response (UPR) is a two-tiered cellular mechanism initiated
by eukaryotic cells that have accumulated misfolded proteins within
the endoplasmic reticulum (ER). An adaptive pathway facilitates the
clearance of the undesired proteins; however, if overwhelmed, cells
trigger apoptosis by upregulating transcription factors such as C/EBP-homologous
protein (CHOP). A high throughput screen was performed directed at
identifying compounds that selectively upregulate the apoptotic CHOP
pathway while avoiding adaptive signaling cascades, resulting in a
sulfonamidebenzamide chemotype that was optimized. These efforts produced
a potent and selective CHOP inducer (AC50 = 0.8 μM;
XBP1 > 80 μM), which was efficacious in both mouse embryonic
fibroblast cells and a human oral squamous cell cancer cell line,
and demonstrated antiproliferative effects for multiple cancer cell
lines in the NCI-60 panel.
Collapse
Affiliation(s)
- Daniel P. Flaherty
- Delbert
M. Shankel Structural Biology Center, University of Kansas Specialized Chemistry Center, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Justin R. Miller
- Carmen
and Ann Adams Department of Pediatrics, Division of Hematology and
Oncology, and the Karmanos Cancer Institute Molecular Therapeutics
Group, Wayne State University, 2228 Elliman Building, 421 East
Canfield, Detroit, Michigan 48201, United States
| | - Danielle M. Garshott
- Carmen
and Ann Adams Department of Pediatrics, Division of Hematology and
Oncology, and the Karmanos Cancer Institute Molecular Therapeutics
Group, Wayne State University, 2228 Elliman Building, 421 East
Canfield, Detroit, Michigan 48201, United States
| | - Michael Hedrick
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Palak Gosalia
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Yujie Li
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Monika Milewski
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Eliot Sugarman
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, Florida 32827, United States
| | - Stefan Vasile
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, Florida 32827, United States
| | - Sumeet Salaniwal
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Ying Su
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Layton H. Smith
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, Florida 32827, United States
| | - Thomas D. Y. Chung
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Anthony B. Pinkerton
- Conrad
Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, United States
| | - Jeffrey Aubé
- Delbert
M. Shankel Structural Biology Center, University of Kansas Specialized Chemistry Center, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Michael U. Callaghan
- Carmen
and Ann Adams Department of Pediatrics, Division of Hematology and
Oncology, and the Karmanos Cancer Institute Molecular Therapeutics
Group, Wayne State University, 2228 Elliman Building, 421 East
Canfield, Detroit, Michigan 48201, United States
| | - Jennifer E. Golden
- Delbert
M. Shankel Structural Biology Center, University of Kansas Specialized Chemistry Center, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Andrew M. Fribley
- Carmen
and Ann Adams Department of Pediatrics, Division of Hematology and
Oncology, and the Karmanos Cancer Institute Molecular Therapeutics
Group, Wayne State University, 2228 Elliman Building, 421 East
Canfield, Detroit, Michigan 48201, United States
| | - Randal J. Kaufman
- Program
in Degenerative Disease Research, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
147
|
Paniagua Soriano G, De Bruin G, Overkleeft HS, Florea BI. Toward understanding induction of oxidative stress and apoptosis by proteasome inhibitors. Antioxid Redox Signal 2014; 21:2419-43. [PMID: 24437477 DOI: 10.1089/ars.2013.5794] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Proteasome inhibitors (PIs) are used in the clinic for the treatment of hematopoietic malignancies. PI inhibitors induce endoplasmatic reticulum (ER) stress and oxidative stress, disruption of signaling pathways, mitochondrial dysfunction, and, eventually, cell death by apoptosis. PIs designated as clinical candidates include natural product derivatives and compounds developed by rational design and feature a wide diversity of structural elements. The vast amount of literature on this topic underscores PIs significance in driving basic research alongside therapeutic benefit. RECENT ADVANCES Research in recent years has brought an in-depth insight into the molecular mechanisms of PI-induced apoptosis. However, there are some paradoxes and controversies in the literature. In this review, the advances and uncertainties, in particular on the time course events that make cells commit to apoptosis, are discussed. In addition, some mechanisms of evolved PI resistance are presented, and speculations on the difference in sensitivity between cell or tumor types are brought forward. The review concludes by giving an outlook of recent methods that may be employed to describe the system biology of how PIs impact cell survival decisions. CRITICAL ISSUES The biology of ER stress, reactive oxygen species (ROS) production, and apoptosis as induced by PIs is not well understood. Absorbed by the strong focus on PIs, one might overlook the importance of proteasome activity activators or modulators and the study of enzymatic pathways that lie up- or downstream from the proteasome function. FUTURE DIRECTIONS An increased understanding of the systems biology at mRNA and protein levels and the kinetics behind the interaction between PIs and cells is imperative. The design and synthesis of subunit specific inhibitors for each of the seven known proteasome activities and for the enzymes associated to proteasomes will aid in unraveling biology of the ubiquitin-proteasome system in relation to ER stress, ROS production, and apoptosis and will generate leads for therapeutic intervention.
Collapse
Affiliation(s)
- Guillem Paniagua Soriano
- Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre , Leiden, The Netherlands
| | | | | | | |
Collapse
|
148
|
Ashley JD, Stefanick JF, Schroeder VA, Suckow MA, Alves NJ, Suzuki R, Kikuchi S, Hideshima T, Anderson KC, Kiziltepe T, Bilgicer B. Liposomal carfilzomib nanoparticles effectively target multiple myeloma cells and demonstrate enhanced efficacy in vivo. J Control Release 2014; 196:113-21. [DOI: 10.1016/j.jconrel.2014.10.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/18/2014] [Accepted: 10/03/2014] [Indexed: 10/24/2022]
|
149
|
Carozzi VA, Chiorazzi A, Canta A, Meregalli C, Oggioni N, Cavaletti G, Marmiroli P. Chemotherapy-induced peripheral neurotoxicity in immune-deficient mice: new useful ready-to-use animal models. Exp Neurol 2014; 264:92-102. [PMID: 25450467 DOI: 10.1016/j.expneurol.2014.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/27/2014] [Accepted: 11/04/2014] [Indexed: 12/13/2022]
Abstract
Cisplatin, paclitaxel and bortezomib are effective chemotherapy drugs in cancer treatment. However, they share severe peripheral neurotoxicity (PN) as one of their major dose-limiting side effects, often impairing cancer patients' quality of life and sometimes being permanent. Even if preclinical oncology is largely based on the use of immune-deficient mice, rodent models used to study the chemotherapy-induced PN are available only in immune-competent animals. In this study we characterized for the first time the PN induced by these chemotherapies through neurophysiological, behavioral, morphological and morphometric studies in athymic nude mice, a commonly employed strain in the preclinical oncology. The animals, divided into four groups, were chronically treated with cisplatin, paclitaxel or bortezomib once or twice a week for 4 or 6 weeks or were left untreated. These schedules were tolerated, neurotoxic and in the range of antineoplastic effectiveness. Despite similarities, differences in the features of PN were evident if compared with immune-competent models under comparable regimens of treatment. The results of this study may provide a basis for future combined analysis of antineoplastic and neurotoxic effects of chemotherapy in the same animals.
Collapse
Affiliation(s)
- Valentina Alda Carozzi
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy; Young Against Pain (YAP) Italian Group, Italy.
| | - Alessia Chiorazzi
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy
| | - Annalisa Canta
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy
| | - Cristina Meregalli
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy
| | - Norberto Oggioni
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy
| | - Paola Marmiroli
- Experimental Neurology Unit and Center for Neuroscience of Milan, Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, MB, Italy
| |
Collapse
|
150
|
Ribatti D, Nico B, Vacca A. Multiple myeloma as a model for the role of bone marrow niches in the control of angiogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 314:259-82. [PMID: 25619720 DOI: 10.1016/bs.ircmb.2014.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bone marrow (BM) contains hematopoietic stem cells (HSCs) and nonhematopoietic cells. HSCs give rise to all types of mature blood cells, while the nonhematopoietic component includes osteoblasts/osteoclasts, endothelial cells (ECs), endothelial progenitor cells (EPCs), and mesenchymal stem cells (MSCs). These cells form specialized "niches" which are close to the vasculature ("vascular niche") or to the endosteum ("osteoblast niche"). The "vascular niche", rich in blood vessels where ECs and mural cells (pericytes and smooth muscle cells), create a microenvironment affecting the behavior of several stem and progenitor cells. The vessel wall acts as an independent niche for the recruitment of EPCs and MSCs. This chapter will focus on the description of the role of BM niches in the control of angiogenesis occurring during multiple myeloma progression.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Beatrice Nico
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Angelo Vacca
- Department of Internal Medicine and Oncology, University of Bari Medical School, Bari, Italy
| |
Collapse
|