51
|
Ghosh C, Gupta N, Mallick A, Santra MK, Basu S. Self-Assembled Glycosylated Chalcone–Boronic Acid Nanodrug Exhibits Anticancer Activity through Mitochondrial Impairment. ACS APPLIED BIO MATERIALS 2018; 1:347-355. [DOI: 10.1021/acsabm.8b00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Chandramouli Ghosh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra, India, 411008
| | - Neha Gupta
- Cancer and Epigenetic Lab, National Center for Cell Science (NCCS) Ganeshkhind, Pune, Maharashtra, India, 411007
| | - Abhik Mallick
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra, India, 411008
| | - Manas Kumar Santra
- Cancer and Epigenetic Lab, National Center for Cell Science (NCCS) Ganeshkhind, Pune, Maharashtra, India, 411007
| | - Sudipta Basu
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra, India, 411008
- Current address: Discipline of Chemistry, Indian Institute of Technology (IIT)-Gandhinagar, Palaj, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
52
|
Hammill JT, Scott DC, Min J, Connelly MC, Holbrook G, Zhu F, Matheny A, Yang L, Singh B, Schulman BA, Guy RK. Piperidinyl Ureas Chemically Control Defective in Cullin Neddylation 1 (DCN1)-Mediated Cullin Neddylation. J Med Chem 2018; 61:2680-2693. [PMID: 29547696 DOI: 10.1021/acs.jmedchem.7b01277] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We previously discovered and validated a class of piperidinyl ureas that regulate defective in cullin neddylation 1 (DCN1)-dependent neddylation of cullins. Here, we report preliminary structure-activity relationship studies aimed at advancing our high-throughput screen hit into a tractable tool compound for dissecting the effects of acute DCN1-UBE2M inhibition on the NEDD8/cullin pathway. Structure-enabled optimization led to a 100-fold increase in biochemical potency and modestly increased solubility and permeability as compared to our initial hit. The optimized compounds inhibit the DCN1-UBE2M protein-protein interaction in our TR-FRET binding assay and inhibit cullin neddylation in our pulse-chase NEDD8 transfer assay. The optimized compounds bind to DCN1 and selectively reduce steady-state levels of neddylated CUL1 and CUL3 in a squamous cell carcinoma cell line. Ultimately, we anticipate that these studies will identify early lead compounds for clinical development for the treatment of lung squamous cell carcinomas and other cancers.
Collapse
Affiliation(s)
- Jared T Hammill
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Daniel C Scott
- Howard Hughes Medical Institute , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States.,Department of Structural Biology , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Jaeki Min
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Michele C Connelly
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Gloria Holbrook
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Fangyi Zhu
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Amy Matheny
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Lei Yang
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - Bhuvanesh Singh
- Department of Surgery, Laboratory of Epithelial Cancer Biology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 United States
| | - Brenda A Schulman
- Howard Hughes Medical Institute , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States.,Department of Structural Biology , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| | - R Kiplin Guy
- Department of Chemical Biology and Theraputics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 United States
| |
Collapse
|
53
|
Preclinical evaluation of antitumor activity of the proteasome inhibitor MLN2238 (ixazomib) in hepatocellular carcinoma cells. Cell Death Dis 2018; 9:28. [PMID: 29348495 PMCID: PMC5833482 DOI: 10.1038/s41419-017-0195-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/17/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the common malignancies and is an increasingly important cause of cancer death worldwide. Surgery, chemotherapy, and radiation therapy extend the 5-year survival limit in HCC patients by only 6%. Therefore, there is a need to develop new therapeutic approaches for the treatment of this disease. The orally bioavailable proteasome inhibitor MLN2238 (ixazomib) has been demonstrated to have anticancer activity. In the present study, we investigated the preclinical therapeutic efficacy of MLN2238 in HCC cells through in vitro and in vivo models, and examined its molecular mechanisms of action. MLN2238 inhibited cell viability in human HCC cells HepG2, Hep3B, and SNU475 in a time- and dose-dependent manner. Flow cytometry analysis demonstrated that MLN2238 induced G2/M cell cycle arrest and cellular apoptosis in HCC cells. Cell cycle arrest was associated with increased expression levels of p21 and p27. MLN2238-induced apoptosis was confirmed by caspase-3/7 activation, PARP cleavage and caspase-dependent β-catenin degradation. In addition, MLN2238 activated ER stress genes in HCC cells and increased the expression of the stress-inducible gene nuclear protein-1. Furthermore, MLN2238 treatment induced upregulation of myeloid cell leukemia-1 (Mcl-1) protein, and Mcl-1 knockdown sensitized HCC cells to MLN2238 treatment, suggesting the contribution of Mcl-1 expression to MLN2238 resistance. This result was also confirmed using the novel Mcl-1 small molecule inhibitor A1210477. Association of A1210477 and MLN2238 determined synergistic antitumor effects in HCC cells. Finally, in vivo orally administered MLN2238 suppressed tumor growth of Hep3B cells in xenograft models in nude mice. In conclusion, our results offer hope for a new therapeutic opportunity in the treatment of HCC patients.
Collapse
|
54
|
de la Puente P, Luderer MJ, Federico C, Jin A, Gilson RC, Egbulefu C, Alhallak K, Shah S, Muz B, Sun J, King J, Kohnen D, Salama NN, Achilefu S, Vij R, Azab AK. Enhancing proteasome-inhibitory activity and specificity of bortezomib by CD38 targeted nanoparticles in multiple myeloma. J Control Release 2017; 270:158-176. [PMID: 29196043 DOI: 10.1016/j.jconrel.2017.11.045] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/29/2017] [Accepted: 11/27/2017] [Indexed: 01/03/2023]
Abstract
The establishment of more effective treatments that can circumvent chemoresistance in Multiple Myeloma (MM) is a priority. Although bortezomib (BTZ) is one of the most potent proteasome inhibitors available, still possesses limitations related to dose limiting side effects. Several strategies have been developed to improve the delivery of chemotherapies to MM by targeting different moieties expressed on MM cells to nanoparticle delivery systems (NPs), which have failed mainly due to their heterogeneous expression on these cells. Our goal was to test CD38 targeted chitosan NPs as novel targeting moiety for MM to improve the potency and efficacy of BTZ in MM cells and reduce the side effects in healthy tissue. We have showed preferential BTZ release in tumor-microenvironment, specific binding to MM cells, and an improved drug cellular uptake through BTZ diffusion from the surface and endocytosed NPs, which translated in enhanced proteasome inhibition and robust cytotoxic effect on MM cells when BTZ was administered through anti-CD38 chitosan NPs. Furthermore, the anti-CD38 chitosan NPs specifically delivered therapeutic agents to MM cells improving therapeutic efficacy and reducing side effects in vivo. The anti-CD38 chitosan NPs showed low toxicity profile allowing enhancement of proteasome-inhibitory activity and specificity of BTZ by endocytosis-mediated uptake of CD38 representing a promising therapy in MM.
Collapse
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Micah J Luderer
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Cinzia Federico
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Abbey Jin
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA
| | - Rebecca C Gilson
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Christopher Egbulefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Shruti Shah
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Justin King
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Daniel Kohnen
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Noha Nabil Salama
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA; Department of Pharmaceutics and Industrial Pharmacy, Cairo University Faculty of Pharmacy, Cairo, Egypt
| | - Samuel Achilefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Ravi Vij
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA.
| |
Collapse
|
55
|
Fukuda Y, Li Y, Segal RA. A Mechanistic Understanding of Axon Degeneration in Chemotherapy-Induced Peripheral Neuropathy. Front Neurosci 2017; 11:481. [PMID: 28912674 PMCID: PMC5583221 DOI: 10.3389/fnins.2017.00481] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Chemotherapeutic agents cause many short and long term toxic side effects to peripheral nervous system (PNS) that drastically alter quality of life. Chemotherapy-induced peripheral neuropathy (CIPN) is a common and enduring disorder caused by several anti-neoplastic agents. CIPN typically presents with neuropathic pain, numbness of distal extremities, and/or oversensitivity to thermal or mechanical stimuli. This adverse side effect often requires a reduction in chemotherapy dosage or even discontinuation of treatment. Currently there are no effective treatment options for CIPN. While the underlying mechanisms for CIPN are not understood, current data identify a “dying back” axon degeneration of distal nerve endings as the major pathology in this disorder. Therefore, mechanistic understanding of axon degeneration will provide insights into the pathway and molecular players responsible for CIPN. Here, we review recent findings that expand our understanding of the pathogenesis of CIPN and discuss pathways that may be shared with the axonal degeneration that occurs during developmental axon pruning and during injury-induced Wallerian degeneration. These mechanistic insights provide new avenues for development of therapies to prevent or treat CIPN.
Collapse
Affiliation(s)
- Yusuke Fukuda
- Department of Neurobiology, Harvard Medical SchoolBoston, MA, United States.,Department of Cancer Biology, Dana-Farber Cancer InstituteBoston, MA, United States
| | - Yihang Li
- Department of Neurobiology, Harvard Medical SchoolBoston, MA, United States.,Department of Cancer Biology, Dana-Farber Cancer InstituteBoston, MA, United States
| | - Rosalind A Segal
- Department of Neurobiology, Harvard Medical SchoolBoston, MA, United States.,Department of Cancer Biology, Dana-Farber Cancer InstituteBoston, MA, United States
| |
Collapse
|
56
|
Ronson A, Tvito A, Rowe JM. Treatment of Relapsed/Refractory Acute Lymphoblastic Leukemia in Adults. Curr Oncol Rep 2017; 18:39. [PMID: 27207612 DOI: 10.1007/s11912-016-0519-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Patients with relapsed and refractory acute lymphoblastic leukemia (ALL) have a dismal prognosis with less than 10 % of patients surviving 5 years. Most such patients cannot be rescued with currently available therapies, whatever the initial treatment they receive. Therefore, there is an urgent need for novel treatment options. Fortunately, over the past several years, an improved understanding of the biology of the disease has allowed the identification of rational molecular targets for therapeutic endeavors and the emergence of novel therapies has sparked great interest. This review will discuss the current treatment landscape for adult patients with relapsed and/or refractory ALL.
Collapse
Affiliation(s)
- Aharon Ronson
- Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bayit Street, Jerusalem, 91031, Israel
| | - Ariella Tvito
- Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bayit Street, Jerusalem, 91031, Israel
| | - Jacob M Rowe
- Department of Hematology, Shaare Zedek Medical Center, 12 Shmuel Bayit Street, Jerusalem, 91031, Israel. .,Technion, Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
57
|
Park S, Park JA, Yoo H, Park HB, Lee Y. Proteasome inhibitor-induced cleavage of HSP90 is mediated by ROS generation and caspase 10-activation in human leukemic cells. Redox Biol 2017; 13:470-476. [PMID: 28715732 PMCID: PMC5512190 DOI: 10.1016/j.redox.2017.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/07/2017] [Accepted: 07/11/2017] [Indexed: 12/26/2022] Open
Abstract
Heat shock protein 90 (HSP90) is a molecular chaperone that supports the stability of client proteins. The proteasome is one of the targets for cancer therapy, and studies are underway to use proteasome inhibitors as anti-cancer drugs. In this study, we found that HSP90 was cleaved to a 55 kDa protein after treatment with proteasome inhibitors including MG132 in leukemia cells but was not cleaved in other tissue-derived cells. HSP90 has two major isoforms (HSP90α and HSP90β), and both were cleaved by MG132 treatment. MG132 treatment also induced a decrease in HSP90 client proteins. MG132 treatment generated ROS, and the cleavage of HSP90 was blocked by a ROS scavenger, N-acetylcysteine (NAC). MG132 activated several caspases, and the activation was reduced by pretreatment with NAC. Based on an inhibitor study, the cleavage of HSP90 induced by MG132 was dependent on caspase 10 activation. Furthermore, active recombinant caspase 10 induced HSP90 cleavage in vitro. MG132 upregulated VDUP-1 expression and reduced the GSH levels implying that the regulation of redox-related proteins is involved. Taken all together, our results suggest that the cleavage of HSP90 by MG132 treatment is mediated by ROS generation and caspase 10 activation. HSP90 cleavage may provide an additional mechanism involved in the anti-cancer effects of proteasome inhibitors. Proteasome inhibitors induce cleavage of HSP90. MG132 induces ROS generation via VDUP-1 upregulation and GSH downregulation. ROS-mediated active caspase 10 cleaves HSP90.
Collapse
Affiliation(s)
- Sangkyu Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Jeong-A Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Hwanmin Yoo
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Han-Bum Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
58
|
Mancuso MR, Neal JW. Novel systemic therapy against malignant pleural mesothelioma. Transl Lung Cancer Res 2017; 6:295-314. [PMID: 28713675 PMCID: PMC5504105 DOI: 10.21037/tlcr.2017.06.01] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022]
Abstract
Malignant pleural mesothelioma is an aggressive tumor of the pleura with an overall poor prognosis. Even with surgical resection, for which only a subset of patients are eligible, long term disease free survival is rare. Standard first-line systemic treatment consists of a platinum analog, an anti-metabolite, and sometimes anti-angiogenic therapy, but there is currently no well-established standard therapy for refractory or relapsed disease. This review focuses on efforts to develop improved systemic therapy for the treatment of malignant pleural mesothelioma (MPM) including cytotoxic systemic therapy, a variety of tyrosine kinase inhibitors and their downstream effector pathways, pharmacologic targeting of the epigenome, novel approaches to target proteins expressed on mesothelioma cells (such as mesothelin), arginine depletion therapy, and the emerging role of immunotherapy. Overall, these studies demonstrate the challenges of improving systemic therapy for MPM and highlight the need to develop therapeutic strategies to control this disease.
Collapse
Affiliation(s)
- Michael R Mancuso
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joel W Neal
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
59
|
Xu J, Ren X, Pathania AS, Fernandez GE, Tran A, Zhang Y, Moats RA, Shackleford GM, Erdreich-Epstein A. PID1 increases chemotherapy-induced apoptosis in medulloblastoma and glioblastoma cells in a manner that involves NFκB. Sci Rep 2017; 7:835. [PMID: 28400607 PMCID: PMC5429784 DOI: 10.1038/s41598-017-00947-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/20/2017] [Indexed: 01/28/2023] Open
Abstract
Phosphotyrosine Interaction Domain containing 1 (PID1; NYGGF4) inhibits growth of medulloblastoma, glioblastoma and atypical teratoid rhabdoid tumor cell lines. PID1 tumor mRNA levels are highly correlated with longer survival in medulloblastoma and glioma patients, suggesting their tumors may have been more sensitive to therapy. We hypothesized that PID1 sensitizes brain tumors to therapy. We found that PID1 increased the apoptosis induced by cisplatin and etoposide in medulloblastoma and glioblastoma cell lines. PID1 siRNA diminished cisplatin-induced apoptosis, suggesting that PID1 is required for cisplatin-induced apoptosis. Etoposide and cisplatin increased NFκB promoter reporter activity and etoposide induced nuclear translocation of NFκB. Etoposide also increased PID1 promoter reporter activity, PID1 mRNA, and PID1 protein, which were diminished by NFκB inhibitors JSH-23 and Bay117082. However, while cisplatin increased PID1 mRNA, it decreased PID1 protein. This decrease in PID1 protein was mitigated by the proteasome inhibitor, bortezomib, suggesting that cisplatin induced proteasome dependent degradation of PID1. These data demonstrate for the first time that etoposide- and cisplatin-induced apoptosis in medulloblastoma and glioblastoma cell lines is mediated in part by PID1, involves NFκB, and may be regulated by proteasomal degradation. This suggests that PID1 may contribute to responsiveness to chemotherapy.
Collapse
Affiliation(s)
- Jingying Xu
- Saban Research Institute at Children's Hospital Los Angeles, Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Pediatrics, Los Angeles, California, 90027, USA
| | - Xiuhai Ren
- Saban Research Institute at Children's Hospital Los Angeles, Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Pediatrics, Los Angeles, California, 90027, USA
| | - Anup Singh Pathania
- Saban Research Institute at Children's Hospital Los Angeles, Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Pediatrics, Los Angeles, California, 90027, USA
| | - G Esteban Fernandez
- Saban Research Institute at Children's Hospital Los Angeles, Cellular Imaging Core, Los Angeles, California, 90027, USA
| | - Anthony Tran
- Saban Research Institute at Children's Hospital Los Angeles, Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Pediatrics, Los Angeles, California, 90027, USA
| | - Yifu Zhang
- Saban Research Institute at Children's Hospital Los Angeles, Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Pediatrics, Los Angeles, California, 90027, USA
| | - Rex A Moats
- Saban Research Institute at Children's Hospital Los Angeles, Department of Radiology, Los Angeles, California, 90027, USA
| | - Gregory M Shackleford
- Saban Research Institute at Children's Hospital Los Angeles, Department of Radiology, Los Angeles, California, 90027, USA
| | - Anat Erdreich-Epstein
- Saban Research Institute at Children's Hospital Los Angeles, Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Pediatrics, Los Angeles, California, 90027, USA. .,Keck School of Medicine, University of Southern California, Departments of Pediatrics and Pathology, Los Angeles, California, 90033, USA.
| |
Collapse
|
60
|
Sartakhti JS, Manshaei MH, Bateni S, Archetti M. Evolutionary Dynamics of Tumor-Stroma Interactions in Multiple Myeloma. PLoS One 2016; 11:e0168856. [PMID: 28030607 PMCID: PMC5193458 DOI: 10.1371/journal.pone.0168856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/01/2016] [Indexed: 01/08/2023] Open
Abstract
Cancer cells and stromal cells cooperate by exchanging diffusible factors that sustain tumor growth, a form of frequency-dependent selection that can be studied in the framework of evolutionary game theory. In the case of multiple myeloma, three types of cells (malignant plasma cells, osteoblasts and osteoclasts) exchange growth factors with different effects, and tumor-stroma interactions have been analysed using a model of cooperation with pairwise interactions. Here we show that a model in which growth factors have autocrine and paracrine effects on multiple cells, a more realistic assumption for tumor-stroma interactions, leads to different results, with implications for disease progression and treatment. In particular, the model reveals that reducing the number of malignant plasma cells below a critical threshold can lead to their extinction and thus to restore a healthy balance between osteoclast and osteoblast, a result in line with current therapies against multiple myeloma.
Collapse
Affiliation(s)
- Javad Salimi Sartakhti
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Mohammad Hossein Manshaei
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
- * E-mail: (MHM); (MA)
| | - Soroosh Bateni
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Marco Archetti
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
- * E-mail: (MHM); (MA)
| |
Collapse
|
61
|
Berenson A, Vardanyan S, David M, Wang J, Harutyunyan NM, Gottlieb J, Halleluyan R, Spektor TM, Udd KA, Eshaghian S, Nassir Y, Eades B, Swift R, Berenson JR. Outcomes of multiple myeloma patients receiving bortezomib, lenalidomide, and carfilzomib. Ann Hematol 2016; 96:449-459. [PMID: 27933373 DOI: 10.1007/s00277-016-2889-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 11/27/2016] [Indexed: 12/13/2022]
Abstract
New classes of drugs including the proteasome inhibitors (PI) bortezomib and, more recently, carfilzomib and the immunomodulatory agent lenalidomide have shown improved outcomes for multiple myeloma (MM) patients during the past decade. However, most of the studies reporting outcomes for patients receiving these drugs have relied on older data sets derived from large institutions that included patients not receiving their treatment at those facilities and represented only those eligible for clinical trials or were from sites where treatment options were limited. We have analyzed data from 258 MM patients who have received treatment with at least one of three agents: bortezomib, carfilzomib, and lenalidomide in a single clinic specializing in MM with respect to their responses and other outcomes to treatment regimens including these agents. Response rates were similar between these three drugs when used for the first time and again during subsequent treatment regimens. As expected, the clinical benefit rates (CBRs) were better for patients receiving their first treatment when compared to their use in subsequent treatment regimens. The CBRs were similar during their 2nd, 3rd, and 4th treatments containing these agents. Many patients refractory to these agents showed responses to regimens containing these same drugs when used in different combinations. In addition, patients refractory to one PI often responded to the other PI. The results of this study demonstrate that novel agents can be used repeatedly in novel combinations with significant clinical benefit for patients with MM.
Collapse
Affiliation(s)
- Ariana Berenson
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - Suzie Vardanyan
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - Michael David
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - James Wang
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
- James R. Berenson, MD, Inc., 9201 W. Sunset Blvd., Suite 310, West Hollywood, CA, USA
| | - Nika Manik Harutyunyan
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - Jillian Gottlieb
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - Ran Halleluyan
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - Tanya M Spektor
- Oncotherapeutic, 9201 W. Sunset Blvd., Suite 317, West Hollywood, CA, USA
| | - Kyle A Udd
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
- James R. Berenson, MD, Inc., 9201 W. Sunset Blvd., Suite 310, West Hollywood, CA, USA
| | - Shahrooz Eshaghian
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA
| | - Youram Nassir
- James R. Berenson, MD, Inc., 9201 W. Sunset Blvd., Suite 310, West Hollywood, CA, USA
| | - Benjamin Eades
- James R. Berenson, MD, Inc., 9201 W. Sunset Blvd., Suite 310, West Hollywood, CA, USA
| | - Regina Swift
- James R. Berenson, MD, Inc., 9201 W. Sunset Blvd., Suite 310, West Hollywood, CA, USA
| | - James R Berenson
- Institute for Myeloma & Bone Cancer Research, 9201 W. Sunset Blvd., Suite 300, West Hollywood, CA, 90069, USA.
- James R. Berenson, MD, Inc., 9201 W. Sunset Blvd., Suite 310, West Hollywood, CA, USA.
- Oncotherapeutic, 9201 W. Sunset Blvd., Suite 317, West Hollywood, CA, USA.
| |
Collapse
|
62
|
Tew KD. Commentary on "Proteasome Inhibitors: A Novel Class of Potent and Effective Antitumor Agents". Cancer Res 2016; 76:4916-7. [PMID: 27587650 DOI: 10.1158/0008-5472.can-16-1974] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 11/16/2022]
Abstract
The relatively recent clinical success of bortezomib, particularly in multiple myeloma, has established the validity of the proteasome as a viable target for anticancer drug development. This highly cited 1999 Cancer Research article from Adams and colleagues was published during the period when this drug was transitioning from preclinical studies to phase I clinical trial status. Their results detail structure-activity analyses using a series of boronic acid proteasome inhibitors and correlate cytotoxicity with inhibition of proteasome activity. In and of itself, the recognition that interference with proteasome functions represented a novel therapeutic approach likely underlies the popularity of this article. In addition, the provision of in vitro (at that time using the NCI 60 cell line panel) and in vivo antitumor activity, toxicology, and mouse pharmacokinetic and pharmacodynamic data provided a solid basis for establishing the future credentials for bortezomib to gain initial FDA approval in 2003. Cancer Res; 76(17); 4916-7. ©2016 AACRSee related article by Adams et al., Cancer Res 1999;59:2615-22Visit the Cancer Research 75(th) Anniversary timeline.
Collapse
Affiliation(s)
- Kenneth D Tew
- Department of Cell & Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
63
|
Abstract
Chemotherapy-induced peripheral neuropathy is a common side effect of selected chemotherapeutic agents. Previous work has suggested that patients often under report the symptoms of chemotherapy-induced peripheral neuropathy and physicians fail to recognize the presence of such symptoms in a timely fashion. The precise pathophysiology that underlies chemotherapy-induced peripheral neuropathy, in both the acute and the chronic phase, remains complex and appears to be medication specific. Recent work has begun to demonstrate and further clarify potential pathophysiological processes that predispose and, ultimately, lead to the development of chemotherapy-induced peripheral neuropathy. There is increasing evidence that the pathway to neuropathy varies with each agent. With a clearer understanding of how these agents affect the peripheral nervous system, more targeted treatments can be developed in order to optimize treatment and prevent long-term side effects.
Collapse
Affiliation(s)
- James Addington
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Miriam Freimer
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
64
|
McCormack PL. Bortezomib: A Review in Mantle Cell Lymphoma in Previously Untreated Patients Unsuitable for Stem-Cell Transplantation. BioDrugs 2016; 29:207-14. [PMID: 26115634 DOI: 10.1007/s40259-015-0131-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bortezomib (Velcade(®)) is a proteasome inhibitor that is approved for the treatment of multiple myeloma and mantle cell lymphoma (MCL). This article reviews the efficacy and tolerability of bortezomib in combination with rituximab, cyclophosphamide, doxorubicin and prednisone (VR-CAP) in the treatment of previously untreated MCL unsuitable for stem-cell transplantation, and overviews the pharmacology of bortezomib. In the large, randomized, assessor-blinded, multinational LYM-3002 trial, induction therapy with VR-CAP improved progression-free survival significantly more than R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone) after a median follow-up of 40 months in patients with newly diagnosed MCL ineligible or not considered for stem-cell transplantation. Complete response and certain other secondary endpoints were improved significantly more with VR-CAP than R-CHOP. Overall survival data were not mature at the time of assessment. The improved efficacy with VR-CAP was accompanied by an increased incidence of grade 3 or higher adverse events, particularly haematological adverse events.
Collapse
Affiliation(s)
- Paul L McCormack
- Springer, Private Bag 65901, Mairangi Bay 0754, Auckland, New Zealand,
| |
Collapse
|
65
|
Upregulation of CCL2 via ATF3/c-Jun interaction mediated the Bortezomib-induced peripheral neuropathy. Brain Behav Immun 2016; 53:96-104. [PMID: 26554515 DOI: 10.1016/j.bbi.2015.11.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/27/2015] [Accepted: 11/07/2015] [Indexed: 11/22/2022] Open
Abstract
Bortezomib (BTZ) is a frequently used chemotherapeutic drug for the treatment of refractory multiple myeloma and hematological neoplasms. The mechanism by which the administration of BTZ leads to painful peripheral neuropathy remains unclear. In present study, we found that application of BTZ at 0.4 mg/kg for consecutive 5 days significantly increased the expression of CCL2 in DRG, and intrathecal administration of neutralizing antibody against CCL2 inhibited the mechanical allodynia induced by BTZ. We also found an increased expression of c-Jun in DRG, and that inhibition of c-Jun signaling prevented the CCL2 upregulation and mechanical allodynia in the rats treated with BTZ. Furthermore, the results with luciferase assay in vitro and ChIP assay in vivo showed that c-Jun might be essential for BTZ-induced CCL2 upregulation via binding directly to the specific position of the ccl2 promoter. In addition, the present results showed that an upregulated expression of ATF3 was co-expressed with c-Jun in the DRG neurons, and the enhanced interaction between c-Jun and ATF3 was observed in DRG in the rats treated with BTZ. Importantly, pretreatment with ATF3 siRNA significantly inhibited the recruitment of c-Jun to the ccl2 promoter in the rats treated with BTZ. Taken together, these findings suggested that upregulation of CCL2 resulting from the enhanced interaction between c-Jun and ATF3 in DRG contributed to BTZ-induced mechanical allodynia.
Collapse
|
66
|
Rui YN, Xu Z, Chen Z, Zhang S. The GST-BHMT assay reveals a distinct mechanism underlying proteasome inhibition-induced macroautophagy in mammalian cells. Autophagy 2016; 11:812-32. [PMID: 25984893 DOI: 10.1080/15548627.2015.1034402] [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: 12/18/2022] Open
Abstract
By monitoring the fragmentation of a GST-BHMT (a protein fusion of glutathionine S-transferase N-terminal to betaine-homocysteine S-methyltransferase) reporter in lysosomes, the GST-BHMT assay has previously been established as an endpoint, cargo-based assay for starvation-induced autophagy that is largely nonselective. Here, we demonstrate that under nutrient-rich conditions, proteasome inhibition by either pharmaceutical or genetic manipulations induces similar autophagy-dependent GST-BHMT processing. However, mechanistically this proteasome inhibition-induced autophagy is different from that induced by starvation as it does not rely on regulation by MTOR (mechanistic target of rapamycin [serine/threonine kinase]) and PRKAA/AMPK (protein kinase, AMP-activated, α catalytic subunit), the upstream central sensors of cellular nutrition and energy status, but requires the presence of the cargo receptors SQSTM1/p62 (sequestosome 1) and NBR1 (neighbor of BRCA1 gene 1) that are normally involved in the selective autophagy pathway. Further, it depends on ER (endoplasmic reticulum) stress signaling, in particular ERN1/IRE1 (endoplasmic reticulum to nucleus signaling 1) and its main downstream effector MAPK8/JNK1 (mitogen-activated protein kinase 8), but not XBP1 (X-box binding protein 1), by regulating the phosphorylation-dependent disassociation of BCL2 (B-cell CLL/lymphoma 2) from BECN1 (Beclin 1, autophagy related). Moreover, the multimerization domain of GST-BHMT is required for its processing in response to proteasome inhibition, in contrast to its dispensable role in starvation-induced processing. Together, these findings support a model in which under nutrient-rich conditions, proteasome inactivation induces autophagy-dependent processing of the GST-BHMT reporter through a distinct mechanism that bears notable similarity with the yeast Cvt (cytoplasm-to-vacuole targeting) pathway, and suggest the GST-BHMT reporter might be employed as a convenient assay to study selective macroautophagy in mammalian cells.
Collapse
Key Words
- ACACA/B, acetyl-CoA carboxylase α/β
- ACTB, actin, β
- ATF4, activating transcription factor 4
- ATF6, activating transcription factor 6
- ATG7, autophagy-related 7
- BCL2, B-cell CLL/lymphoma 2
- BECN1, Beclin 1, autophagy-related
- BHMT
- BHMT, betaine-homocysteine S-methyltransferase
- Baf A1, bafilomycin A1
- CTNNB1, catenin (cadherin-associated protein), β 1, 88kDa
- Cvt, cytoplasm-to-vacuole-targeting
- DDIT3, DNA-damage-inducible transcript 3
- EBSS, Earle's Balanced Salt Solution
- EIF2AK3, eukaryotic translation initiation factor 2-α, kinase 3
- EIF4EBP1, eukaryotic translation initiation factor 4E binding protein 1
- ER, endoplasmic reticulum
- ERN1, endoplasmic reticulum to nucleus signaling 1
- GST, glutathionine S-transferase
- GST-BHMT(FRAG), an autophagy-mediated cleavage product of the GST-BHMT reporter
- GST-BHMT, a fusion protein of glutathionine S-transferase N-terminal to betaine-homocysteine S-methyltransferase
- HA, hemagglutinin
- HSPA5, heat shock 70kDa protein 5 (glucose-regulated protein, 78kDa)
- LSCS, linker-specific cleavage site
- MAP1LC3, microtubule-associated protein 1 light chain 3
- MAP2K7, mitogen-activated protein kinase kinase 7
- MAPK8, mitogen-activated protein kinase 8
- MTOR
- MTOR, mechanistic target of rapamycin (serine/threonine kinase)
- MTORC1, MTOR complex 1
- NBR1, neighbor of BRCA1 gene 1
- P4HB, prolyl 4-hydroxylase, β polypeptide
- PRKAA, protein kinase, AMP-activated, α catalytic subunit
- PRKAA/AMPK
- RHEB, Ras homolog enriched in brain
- RM, rich medium
- RPS6KB1, ribosomal protein S6 kinase, 70kDa, polypeptide 1
- SQSTM1, sequestosome 1
- TSC1/2, tuberous sclerosis 1/2
- ULK1, unc-51 like autophagy activating kinase 1
- UPR, unfolded protein response
- UPS, ubiquitin proteasome system
- XBP1, X-box binding protein 1
- cargo receptors SQSTM1/p62 and NBR1
- proteasome inhibition
- selective macroautophagy
Collapse
Affiliation(s)
- Yan-Ning Rui
- a The Brown Foundation Institute of Molecular Medicine
| | | | | | | |
Collapse
|
67
|
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
|
68
|
Zhang X, Cai J, Zheng Z, Polin L, Lin Z, Dandekar A, Li L, Sun F, Finley RL, Fang D, Yang ZQ, Zhang K. A novel ER-microtubule-binding protein, ERLIN2, stabilizes Cyclin B1 and regulates cell cycle progression. Cell Discov 2015; 1:15024. [PMID: 27462423 PMCID: PMC4860859 DOI: 10.1038/celldisc.2015.24] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/10/2015] [Indexed: 12/11/2022] Open
Abstract
The gene encoding endoplasmic reticulum (ER) lipid raft-associated protein 2 (ERLIN2) is amplified in human breast cancers. ERLIN2 gene mutations were also found to be associated with human childhood progressive motor neuron diseases. Yet, an understanding of the physiological function and mechanism for ERLIN2 remains elusive. In this study, we reveal that ERLIN2 is a spatially and temporally regulated ER–microtubule-binding protein that has an important role in cell cycle progression by interacting with and stabilizing the mitosis-promoting factors. Whereas ERLIN2 is highly expressed in aggressive human breast cancers, during normal development ERLIN2 is expressed at the postnatal stage and becomes undetectable in adulthood. ERLIN2 interacts with the microtubule component α-tubulin, and this interaction is maximal during the cell cycle G2/M phase where ERLIN2 simultaneously interacts with the mitosis-promoting complex Cyclin B1/Cdk1. ERLIN2 facilitates K63-linked ubiquitination and stabilization of Cyclin B1 protein in G2/M phase. Downregulation of ERLIN2 results in cell cycle arrest, represses breast cancer proliferation and malignancy and increases sensitivity of breast cancer cells to anticancer drugs. In summary, our study revealed a novel ER–microtubule-binding protein, ERLIN2, which interacts with and stabilizes mitosis-promoting factors to regulate cell cycle progression associated with human breast cancer malignancy.
Collapse
Affiliation(s)
- Xuebao Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine , Detroit, MI, USA
| | - Juan Cai
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine , Detroit, MI, USA
| | - Ze Zheng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine , Detroit, MI, USA
| | - Lisa Polin
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhenghong Lin
- Department of Pathology, Northwestern University Feinberg School of Medicine , Chicago, IL, USA
| | - Aditya Dandekar
- Department of Immunology and Microbiology, Wayne State University School of Medicine , Detroit, MI, USA
| | - Li Li
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA; Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Fei Sun
- Department of Physiology, Wayne State University School of Medicine , Chicago, IL, USA
| | - Russell L Finley
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA; Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zeng-Quan Yang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA; Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
69
|
Novel roles of the unfolded protein response in the control of tumor development and aggressiveness. Semin Cancer Biol 2015; 33:67-73. [DOI: 10.1016/j.semcancer.2015.04.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 11/20/2022]
|
70
|
Cormier K, Curry RD, Betsch MP, Goguen JA, Vogels CM, Decken A, Turcotte S, Westcott SA. Synthesis, Characterization, and Anticancer Activities of Pyrogallol-Based Arylspiroborates. J Heterocycl Chem 2015. [DOI: 10.1002/jhet.2490] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kevin Cormier
- Département de chimie et biochimie; Université de Moncton; Campus de Moncton Moncton NB E1A 3E9 Canada
- Atlantic Cancer Research Institute; Moncton NB E1C 8X3 Canada
| | - R. Daniel Curry
- Department of Chemistry and Biochemistry; Mount Allison University; Sackville NB E4L 1G8 Canada
| | - Mitchel P. Betsch
- Department of Chemistry and Biochemistry; Mount Allison University; Sackville NB E4L 1G8 Canada
| | - Jeremy A. Goguen
- Department of Chemistry and Biochemistry; Mount Allison University; Sackville NB E4L 1G8 Canada
| | - Christopher M. Vogels
- Department of Chemistry and Biochemistry; Mount Allison University; Sackville NB E4L 1G8 Canada
| | - Andreas Decken
- Department of Chemistry; University of New Brunswick; Fredericton NB E3B 5A3 Canada
| | - Sandra Turcotte
- Département de chimie et biochimie; Université de Moncton; Campus de Moncton Moncton NB E1A 3E9 Canada
- Atlantic Cancer Research Institute; Moncton NB E1C 8X3 Canada
| | - Stephen A. Westcott
- Department of Chemistry and Biochemistry; Mount Allison University; Sackville NB E4L 1G8 Canada
| |
Collapse
|
71
|
Stanley M, Han C, Knebel A, Murphy P, Shpiro N, Virdee S. Orthogonal thiol functionalization at a single atomic center for profiling transthiolation activity of E1 activating enzymes. ACS Chem Biol 2015; 10:1542-54. [PMID: 25845023 DOI: 10.1021/acschembio.5b00118] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Transthiolation is a fundamental biological reaction and is utilized by many enzymes involved in the conjugation of ubiquitin and ubiquitin-like proteins. However, tools that enable selective profiling of this activity are lacking. Transthiolation requires cysteine-cysteine juxtaposition; therefore a method that enables irreversible "stapling" of proximal thiols would facilitate the development of novel probes that could be used to profile this activity. Herein, we characterize biocompatible chemistry that enables sequential functionalization of cysteines within proteins at a single atomic center. We use our method to develop a new class of activity-based probe that profiles transthiolation activity of human E1 activating enzymes. We demonstrate use in vitro and in situ and compatibility with competitive activity-based protein profiling. We also use the probe to gain insight into the determinants of transthiolation between E2 and a RING-in-between-RING (RBR) E3 ligase. Furthermore, we anticipate that this method of thiol functionalization will have broad utility by enabling simple redox-stable cross-linking of proximal cysteines in general.
Collapse
Affiliation(s)
- Mathew Stanley
- MRC Protein
Phosphorylation
and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Cong Han
- MRC Protein
Phosphorylation
and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Axel Knebel
- MRC Protein
Phosphorylation
and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Paul Murphy
- MRC Protein
Phosphorylation
and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Natalia Shpiro
- MRC Protein
Phosphorylation
and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Satpal Virdee
- MRC Protein
Phosphorylation
and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| |
Collapse
|
72
|
Mitochondrial Dysfunction in Chemotherapy-Induced Peripheral Neuropathy (CIPN). TOXICS 2015; 3:198-223. [PMID: 29056658 PMCID: PMC5634687 DOI: 10.3390/toxics3020198] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 12/18/2022]
Abstract
The mitochondrial dysfunction has a critical role in several disorders including chemotherapy-induced peripheral neuropathies (CIPN). This is due to a related dysregulation of pathways involving calcium signalling, reactive oxygen species and apoptosis. Vincristine is able to affect calcium movement through the Dorsal Root Ganglia (DRG) neuronal mitochondrial membrane, altering its homeostasis and leading to abnormal neuronal excitability. Paclitaxel induces the opening of the mitochondrial permeability transition pore in axons followed by mitochondrial membrane potential loss, increased reactive oxygen species generation, ATP level reduction, calcium release and mitochondrial swelling. Cisplatin and oxaliplatin form adducts with mitochondrial DNA producing inhibition of replication, disruption of transcription and morphological abnormalities within mitochondria in DRG neurons, leading to a gradual energy failure. Bortezomib is able to modify mitochondrial calcium homeostasis and mitochondrial respiratory chain. Moreover, the expression of a certain number of genes, including those controlling mitochondrial functions, was altered in patients with bortezomib-induced peripheral neuropathy.
Collapse
|
73
|
The ubiquitin proteasome system plays a role in venezuelan equine encephalitis virus infection. PLoS One 2015; 10:e0124792. [PMID: 25927990 PMCID: PMC4415917 DOI: 10.1371/journal.pone.0124792] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 03/11/2015] [Indexed: 01/20/2023] Open
Abstract
Many viruses have been implicated in utilizing or modulating the Ubiquitin Proteasome System (UPS) to enhance viral multiplication and/or to sustain a persistent infection. The mosquito-borne Venezuelan equine encephalitis virus (VEEV) belongs to the Togaviridae family and is an important biodefense pathogen and select agent. There are currently no approved vaccines or therapies for VEEV infections; therefore, it is imperative to identify novel targets for therapeutic development. We hypothesized that a functional UPS is required for efficient VEEV multiplication. We have shown that at non-toxic concentrations Bortezomib, a FDA-approved inhibitor of the proteasome, proved to be a potent inhibitor of VEEV multiplication in the human astrocytoma cell line U87MG. Bortezomib inhibited the virulent Trinidad donkey (TrD) strain and the attenuated TC-83 strain of VEEV. Additional studies with virulent strains of Eastern equine encephalitis virus (EEEV) and Western equine encephalitis virus (WEEV) demonstrated that Bortezomib is a broad spectrum inhibitor of the New World alphaviruses. Time-of-addition assays showed that Bortezomib was an effective inhibitor of viral multiplication even when the drug was introduced many hours post exposure to the virus. Mass spectrometry analyses indicated that the VEEV capsid protein is ubiquitinated in infected cells, which was validated by confocal microscopy and immunoprecipitation assays. Subsequent studies revealed that capsid is ubiquitinated on K48 during early stages of infection which was affected by Bortezomib treatment. This study will aid future investigations in identifying host proteins as potential broad spectrum therapeutic targets for treating alphavirus infections.
Collapse
|
74
|
Glynn SJ, Gaffney KJ, Sainz MA, Louie SG, Petasis NA. Molecular characterization of the boron adducts of the proteasome inhibitor bortezomib with epigallocatechin-3-gallate and related polyphenols. Org Biomol Chem 2015; 13:3887-99. [PMID: 25669488 PMCID: PMC4366333 DOI: 10.1039/c4ob02512a] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The green tea polyphenol epigallocatechin-3-gallate (EGCG) was reported to effectively antagonize the ability of Bortezomib (BZM) to induce apoptosis in cancer cells. This interaction was attributed to the formation of a covalent adduct between a phenolic moiety of EGCG with the boronic acid group of Bortezomib. However, the structural details of this boron adduct and the molecular factors that contribute to its formation and its ability to inhibit Bortezomib's activity remain unclear. This paper describes the use of NMR spectroscopy and cell assays to characterize the structures and properties of the boron adducts of EGCG and related polyphenols. The observed boron adducts included both boronate and borate derivatives, and their structural characteristics were correlated with cell-based evaluation of the ability of EGCG and other phenols to antagonize the anticancer activity of Bortezomib. The enhanced stability of the BZM/EGCG adduct was attributed to electronic and steric reasons, and a newly identified intramolecular interaction of the boron atom of BZM with the adjacent amide bond. The reported approach provides a useful method for determining the potential ability of polyphenols to form undesired adducts with boron-based drugs and interfere with their actions.
Collapse
Affiliation(s)
- Stephen J Glynn
- Department of Chemistry and Loker Hydrocarbon Research Institute, University of Southern California, Los Angeles, California 90089, USA.
| | | | | | | | | |
Collapse
|
75
|
LeBeau AM, Denmeade SR. Protease-activated pore-forming peptides for the treatment and imaging of prostate cancer. Mol Cancer Ther 2014; 14:659-68. [PMID: 25537662 DOI: 10.1158/1535-7163.mct-14-0744] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023]
Abstract
A common hallmark of cancers with highly aggressive phenotypes is increased proteolysis in the tumor and the surrounding microenvironment. Prostate cancer has a number of proteases uniquely associated with it that may play various important roles in disease progression. In this report, we utilize the peritumoral proteolytic activity of prostate cancer to activate engineered peptide constructs for the treatment and noninvasive imaging of prostate cancer. Using a modular "propeptide" approach, a cationic diastereomeric pore-forming peptide domain was linked to an inactivating acidic peptide domain. The inactivating acidic peptide domain was engineered to be a cleavable substrate for the secreted serine protease prostate-specific antigen (PSA) or the transmembrane metalloprotease prostate-specific membrane antigen (PSMA). The propeptides were then evaluated in a direct comparison study. Both the PSA and PSMA activated propeptides were found to be cytotoxic to prostate cancer cells in vitro. In vivo, however, treatment of LNCaP and CWR22Rv1 xenografts with the PSMA propeptide resulted in a pronounced cytostatic effect when compared with xenografts treated with the PSA propeptide or the cationic diastereomeric peptide alone. The PSMA activated propeptide also proved to be an effective optical imaging probe in vivo when labeled with a near-infrared fluorophore. These data suggest that protease-activated pore-forming peptides could potentially be used for both imaging and treating prostate cancer.
Collapse
Affiliation(s)
- Aaron M LeBeau
- Department of Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota.
| | - Samuel R Denmeade
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
76
|
Bin BH, Hojyo S, Ryong Lee T, Fukada T. Spondylocheirodysplastic Ehlers-Danlos syndrome (SCD-EDS) and the mutant zinc transporter ZIP13. Rare Dis 2014; 2:e974982. [PMID: 26942106 PMCID: PMC4755239 DOI: 10.4161/21675511.2014.974982] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/28/2014] [Accepted: 10/06/2014] [Indexed: 12/31/2022] Open
Abstract
The zinc transporter protein ZIP13 plays crucial roles in bone, tooth, and connective tissue development, and its dysfunction is responsible for the spondylocheirodysplastic form of Ehlers-Danlos syndrome (SCD-EDS, OMIM 612350). We recently reported that the pathogenic mutations in ZIP13 reduce its functional protein level by accelerating the protein degradation via the VCP-linked ubiquitin proteasome pathway, resulting in the disturbance of intracellular zinc homeostasis that appears to contribute to SCD-EDS pathogenesis. Finally, we implicate that possible therapeutic approaches for SCD-EDS would be based on regulating the degradation of the pathogenic mutant ZIP13 proteins.
Collapse
Affiliation(s)
- Bum-Ho Bin
- Bioscience Research Institute; Amorepacific Corporation R&D Center; Yongin, Republic of Korea; Division of Pathology; Department of Oral Diagnostic Sciences; School of Dentistry; Showa University, Shinagawa, Japan
| | - Shintaro Hojyo
- Deutsches Rheuma-Forschungszentrum, Berlin; Osteoimmunology; Berlin, Germany; RIKEN Center for Integrative Medical Sciences; Yokohama, Japan
| | - Tae Ryong Lee
- Bioscience Research Institute; Amorepacific Corporation R&D Center ; Yongin, Republic of Korea
| | - Toshiyuki Fukada
- Division of Pathology; Department of Oral Diagnostic Sciences; School of Dentistry; Showa University, Shinagawa, Japan; RIKEN Center for Integrative Medical Sciences; Yokohama, Japan
| |
Collapse
|
77
|
Carozzi VA, Canta A, Chiorazzi A. Chemotherapy-induced peripheral neuropathy: What do we know about mechanisms? Neurosci Lett 2014; 596:90-107. [PMID: 25459280 DOI: 10.1016/j.neulet.2014.10.014] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/09/2014] [Indexed: 12/14/2022]
Abstract
Cisplatin, oxaliplatin, paclitaxel, vincristine and bortezomib are some of the most effective drugs successfully employed (alone or in combinations) as first-line treatment for common cancers. However they often caused severe peripheral neurotoxicity and neuropathic pain. Structural deficits in Dorsal Root Ganglia and sensory nerves caused symptoms as sensory loss, paresthesia, dysaesthesia and numbness that result in patient' suffering and also limit the life-saving therapy. Several scientists have explored the various mechanisms involved in the onset of chemotherapy-related peripheral neurotoxicity identifying molecular targets useful for the development of selected neuroprotective strategies. Dorsal Root Ganglia sensory neurons, satellite cells, Schwann cells, as well as neuronal and glial cells in the spinal cord, are the preferential sites in which chemotherapy neurotoxicity occurs. DNA damage, alterations in cellular system repairs, mitochondria changes, increased intracellular reactive oxygen species, alterations in ion channels, glutamate signalling, MAP-kinases and nociceptors ectopic activation are among the events that trigger the onset of peripheral neurotoxicity and neuropathic pain. In the present work we review the role of the main players in determining the pathogenesis of anticancer drugs-induced peripheral neuropathy.
Collapse
Affiliation(s)
- V A Carozzi
- Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, Italy.
| | - A Canta
- Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, Italy
| | - A Chiorazzi
- Department of Surgery and Translational Medicine, University of Milan-Bicocca, Monza, Italy
| |
Collapse
|
78
|
Osawa T, Naito T, Kaneko T, Mino Y, Ohnishi K, Yamada H, Kawakami J. Blood distribution of bortezomib and its kinetics in multiple myeloma patients. Clin Biochem 2014; 47:54-9. [DOI: 10.1016/j.clinbiochem.2014.06.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 06/18/2014] [Accepted: 06/28/2014] [Indexed: 11/25/2022]
|
79
|
Nrf2- and ATF4-dependent upregulation of xCT modulates the sensitivity of T24 bladder carcinoma cells to proteasome inhibition. Mol Cell Biol 2014; 34:3421-34. [PMID: 25002527 DOI: 10.1128/mcb.00221-14] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The ubiquitin-proteasome pathway degrades ubiquitinated proteins to remove damaged or misfolded protein and thus plays an important role in the maintenance of many important cellular processes. Because the pathway is also crucial for tumor cell growth and survival, proteasome inhibition by specific inhibitors exhibits potent antitumor effects in many cancer cells. xCT, a subunit of the cystine antiporter system xc (-), plays an important role in cellular cysteine and glutathione homeostasis. Several recent reports have revealed that xCT is involved in cancer cell survival; however, it was unknown whether xCT affects the cytotoxic effects of proteasome inhibitors. In this study, we found that two stress-inducible transcription factors, Nrf2 and ATF4, were upregulated by proteasome inhibition and cooperatively enhance human xCT gene expression upon proteasome inhibition. In addition, we demonstrated that the knockdown of xCT by small interfering RNA (siRNA) or pharmacological inhibition of xCT by sulfasalazine (SASP) or (S)-4-carboxyphenylglycine (CPG) significantly increased the sensitivity of T24 cells to proteasome inhibition. These results suggest that the simultaneous inhibition of both the proteasome and xCT could have therapeutic benefits in the treatment of bladder tumors.
Collapse
|
80
|
Hong M, Kim H, Kim I. Ribosomal protein L19 overexpression activates the unfolded protein response and sensitizes MCF7 breast cancer cells to endoplasmic reticulum stress-induced cell death. Biochem Biophys Res Commun 2014; 450:673-8. [PMID: 24950402 DOI: 10.1016/j.bbrc.2014.06.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/07/2014] [Indexed: 02/02/2023]
Abstract
Although first identified for their roles in protein synthesis, certain ribosomal proteins exert pleiotropic physiological functions in the cell. Ribosomal protein L19 is overexpressed in breast cancer cells by amplification and copy number variation. In this study, we examined the novel pro-apoptotic role of ribosomal protein L19 in the breast cancer cell line MCF7. Overexpression of RPL19 sensitized MCF7 cells to endoplasmic reticulum stress-induced cell death. RPL19 overexpression itself was not cytotoxic; however, cell death induction was enhanced when RPL19 overexpressing cells were incubated with endoplasmic reticulum stress-inducing agents, and this sensitizing effect was specific to MCF7 cells. Examination of the cell signaling pathways that mediate the unfolded protein response (UPR) revealed that overexpression of RPL19 induced pre-activation of the UPR, including phosphorylation of pERK-like ER kinase (PERK), phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α), and activation of p38 MAPK-associated stress signaling. Our findings suggest that upregulation of RPL19 induces ER stress, resulting in increased sensitivity to ER stress and enhanced cell death in MCF7 breast cancer cells.
Collapse
Affiliation(s)
- Mina Hong
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
| | - HyungRyong Kim
- Department of Dental Pharmacology, School of Dentistry, Wonkwang University, Iksan, Chonbuk, Republic of Korea.
| | - Inki Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea; Department of Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
81
|
Ashley JD, Stefanick JF, Schroeder VA, Suckow MA, Kiziltepe T, Bilgicer B. Liposomal Bortezomib Nanoparticles via Boronic Ester Prodrug Formulation for Improved Therapeutic Efficacy in Vivo. J Med Chem 2014; 57:5282-92. [DOI: 10.1021/jm500352v] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jonathan D. Ashley
- Department
of Chemical and Biomolecular Engineering, University of Notre Dame, Notre
Dame, 182 Fitzpatrick Hall, Indiana 46556, United States
| | - Jared F. Stefanick
- Department
of Chemical and Biomolecular Engineering, University of Notre Dame, Notre
Dame, 182 Fitzpatrick Hall, Indiana 46556, United States
| | - Valerie A. Schroeder
- Department
of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
- Freimann
Life Science Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Mark A. Suckow
- Department
of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, United States
- Freimann
Life Science Center, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Tanyel Kiziltepe
- Department
of Chemical and Biomolecular Engineering, University of Notre Dame, Notre
Dame, 182 Fitzpatrick Hall, Indiana 46556, United States
- Advanced
Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Basar Bilgicer
- Department
of Chemical and Biomolecular Engineering, University of Notre Dame, Notre
Dame, 182 Fitzpatrick Hall, Indiana 46556, United States
- Advanced
Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, Indiana 46556, United States
- Department
of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
82
|
Claes DJ, Yin H, Goebel J. Protective immunity and use of bortezomib for antibody-mediated rejection in a pediatric kidney transplant recipient. Pediatr Transplant 2014; 18:E100-5. [PMID: 24725066 DOI: 10.1111/petr.12256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/24/2014] [Indexed: 12/27/2022]
Abstract
Standard treatments for AMR-rituximab, intravenous immunoglobulin, and/or plasmapheresis-aim to suppress the production and modulate the effect of donor-specific antibodies and remove them, respectively. Proteasome inhibitors such as bortezomib are potent therapeutic agents that target plasma cells more effectively than rituximab to reduce measurable donor-specific antibody production. Little is known in adults, and no data exist in children about effects of proteasome inhibition to treat AMR on protective antibody titers. We present a pediatric renal transplant recipient who received bortezomib for relatively early AMR and whose antibody titers to measles and tetanus were tracked. The AMR was treated successfully, and we noted no clinical decrease in the overall level of protective immunity from pretransplant baseline levels at almost one yr after AMR treatment cessation. Larger studies will elucidate more clearly how proteasome inhibition to treat AMR affects protective immunity in pediatric transplant recipients.
Collapse
Affiliation(s)
- Donna J Claes
- Department of Pediatric Nephrology & Hypertension, Cincinnati Children's Hospital & Medical Center, Cincinnati, OH, USA
| | | | | |
Collapse
|
83
|
Zhang J, Su YM, Li D, Cui Y, Huang ZZ, Wei JY, Xue Z, Pang RP, Liu XG, Xin WJ. TNF-α-mediated JNK activation in the dorsal root ganglion neurons contributes to Bortezomib-induced peripheral neuropathy. Brain Behav Immun 2014; 38:185-91. [PMID: 24530998 DOI: 10.1016/j.bbi.2014.01.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/29/2014] [Accepted: 01/31/2014] [Indexed: 01/30/2023] Open
Abstract
Bortezomib (BTZ) is a frequently used chemotherapeutic drug for the treatment of refractory multiple myeloma and hematological neoplasms. The mechanism by which the administration of BTZ leads to painful peripheral neuropathy remains unclear. In the present study, we first determined that the administration of BTZ upregulated the expression of TNF-α and phosphorylated JNK1/2 in the dorsal root ganglion (DRG) of rat. Furthermore, the TNF-α synthesis inhibitor thalidomide significantly blocked the activation of both isoforms JNK1 and JNK2 in the DRG and attenuated mechanical allodynia following BTZ treatment. Knockout of the expression of TNF-α receptor TNFR1 (TNFR1 KO mice) or TNFR2 (TNFR2 KO mice) inhibited JNK1 and JNK2 activation and decreased mechanical allodynia induced by BTZ. These results suggest that upregulated TNF-α expression may activate JNK signaling via TNFR1 or TNFR2 to mediate mechanical allodynia following BTZ treatment.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Yi-Min Su
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Dai Li
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Yu Cui
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Zhen-Zhen Huang
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Jia-You Wei
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Zi Xue
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Rui-Ping Pang
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Xian-Guo Liu
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China
| | - Wen-Jun Xin
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yet-Sen University, 74 Zhongshan Rd. 2, Guangzhou 510080, China.
| |
Collapse
|
84
|
Kitamura R, Andoh T, Mizoguchi S, Saito Y, Takahata H, Kuraishi Y. Gabapentin inhibits bortezomib-induced mechanical allodynia through supraspinal action in mice. J Pharmacol Sci 2014; 124:502-10. [PMID: 24681698 DOI: 10.1254/jphs.13274fp] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Bortezomib, an inhibitor of proteasome holoenzyme, is used to treat relapsed and refractory multiple myeloma. Peripheral neuropathy is a treatment-limiting adverse effect of bortezomib and is very difficult to control. In this study, we examined the efficacy of gabapentin in inhibiting bortezomib-induced peripheral neuropathy. Single intravenous injections of bortezomib (0.03 - 0.3 mg/kg) dose-dependently induced mechanical allodynia with a peak effect 12 days after injection. Bortezomib (0.3 mg/kg) also caused mechanical hyperalgesia, but neither affected thermal nociception nor induced cold allodynia. Bortezomib increased the response of the saphenous nerve to weak punctate stimulation but not response to cool stimulation of the skin. When administered 12 days after bortezomib injection, oral and intracisternal gabapentin markedly inhibited mechanical allodynia. Intrathecal, but not intraplantar, gabapentin had a tendency to reduce mechanical allodynia. The antiallodynic activity of orally administered gabapentin was suppressed by noradrenaline, but not serotonin, depletion in the spinal cord. Bortezomib did not affect the expression levels of the calcium channel α₂δ-1 subunit, a high-affinity binding site of gabapentin, in the plantar skin, spinal cord, medulla oblongata, and pons. These results suggest that gabapentin inhibits bortezomib-induced mechanical allodynia, most likely through the activation of the descending noradrenergic system.
Collapse
Affiliation(s)
- Ryo Kitamura
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Japan
| | | | | | | | | | | |
Collapse
|
85
|
Schorn M, Zettler J, Noel JP, Dorrestein PC, Moore BS, Kaysser L. Genetic basis for the biosynthesis of the pharmaceutically important class of epoxyketone proteasome inhibitors. ACS Chem Biol 2014; 9:301-9. [PMID: 24168704 DOI: 10.1021/cb400699p] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The epoxyketone proteasome inhibitors are an established class of therapeutic agents for the treatment of cancer. Their unique α',β'-epoxyketone pharmacophore allows binding to the catalytic β-subunits of the proteasome with extraordinary specificity. Here, we report the characterization of the first gene clusters for the biosynthesis of natural peptidyl-epoxyketones. The clusters for epoxomicin, the lead compound for the anticancer drug Kyprolis, and for eponemycin were identified in the actinobacterial producer strains ATCC 53904 and Streptomyces hygroscopicus ATCC 53709, respectively, using a modified protocol for Ion Torrent PGM genome sequencing. Both gene clusters code for a hybrid nonribosomal peptide synthetase/polyketide synthase multifunctional enzyme complex and homologous redox enzymes. Epoxomicin and eponemycin were heterologously produced in Streptomyces albus J1046 via whole pathway expression. Moreover, we employed mass spectral molecular networking for a new comparative metabolomics approach in a heterologous system and discovered a number of putative epoxyketone derivatives. With this study, we have definitively linked epoxyketone proteasome inhibitors and their biosynthesis genes for the first time in any organism, which will now allow for their detailed biochemical investigation.
Collapse
Affiliation(s)
- Michelle Schorn
- Scripps
Institution of Oceanography, University of California, San Diego, California 92093, United States of America
| | - Judith Zettler
- Pharmaceutical
Biology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- German Center
for Infection Research (DZIF), partner site Tübingen, 72076 Tübingen, Germany
| | - Joseph P. Noel
- Jack
H. Skirball Center for Chemical Biology and Proteomics, Salk Institute for Biological Studies, La Jolla, California 92037, United States of America
| | - Pieter C. Dorrestein
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San
Diego, California 92093, United States of America
| | - Bradley S. Moore
- Scripps
Institution of Oceanography, University of California, San Diego, California 92093, United States of America
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San
Diego, California 92093, United States of America
| | - Leonard Kaysser
- Scripps
Institution of Oceanography, University of California, San Diego, California 92093, United States of America
- Pharmaceutical
Biology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- German Center
for Infection Research (DZIF), partner site Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
86
|
Nanduri B, Suvarnapunya AE, Venkatesan M, Edelmann MJ. Deubiquitinating enzymes as promising drug targets for infectious diseases. Curr Pharm Des 2013; 19:3234-47. [PMID: 23151130 DOI: 10.2174/1381612811319180008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/01/2012] [Indexed: 01/25/2023]
Abstract
Deubiquitinating enzymes (DUBs) remove ubiquitin and ubiquitin-like modifications from proteins and they have been known to contribute to processes relevant in microbial infection, such as immune responses pathways. Numerous viral and bacterial DUBs have been identified, and activities of several host DUBs are known to be modulated during the infection process, either by a pathogen or by a host. Recently there have been attempts to take advantage of this feature and design therapeutic inhibitors of DUBs that can be used to limit the spread of infection. This review is focused on exploring the potential of DUBs in the treatment of infectious diseases.
Collapse
Affiliation(s)
- Bindu Nanduri
- Institute for Genomics, Biocomputing and Biotechnology, Mississippi State University, Mississippi State, MS 39762, USA
| | | | | | | |
Collapse
|
87
|
Waight CC, Cain R. Authorising bortezomib treatment prior to reviewing haematology results: a step toward home administration. J Oncol Pharm Pract 2013; 20:351-5. [PMID: 24154652 DOI: 10.1177/1078155213508438] [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] [Indexed: 11/17/2022]
Abstract
Bortezomib treatment requires four visits to a chemotherapy unit in each 21-day cycle. Analysis of the Day 1 full blood count could allow clinicians to predict the risk of Grade 4 thrombocytopenia, thus negating the need to review the full blood count prior to each dose. The freedom to administer bortezomib without reviewing full blood count results on each treatment day could minimise appointment times and be a step toward home administration. A prospective study of treatment authorisation following a full toxicity assessment and full blood count results from the previous treatment day was undertaken. The full blood count results from 27 patients, receiving 381 doses revealed 12 treatment episodes where bortezomib was administered in the presence of Grade 4 thrombocytopenia. One instance of bleeding and two episodes of neutropenic sepsis were detected during toxicity assessments and treatment was not administered. Only one instance of Grade 4 thrombocytopenia was reported on any other treatment day when the Day 1 platelet count was greater than 75 × 10(9) units/l. From this data, Day 1 full blood count parameters were derived, which minimise the risk of Grade 4 haematological toxicity on subsequent treatment days, allowing clinicians to identify suitable patients for administration of bortezomib prior to reviewing full blood count results. When platelet counts on Day 1 are greater than 75 × 10(9) units/l and neutrophil counts are greater than 1.0 × 10(9) units/l, the administration of bortezomib can be authorised without the need for review of the full blood count on subsequent days of that cycle.
Collapse
Affiliation(s)
| | - Rebecca Cain
- Pharmacy Department, Victoria Hospital Kirkcaldy, NHS Fife, UK
| |
Collapse
|
88
|
Carozzi VA, Renn CL, Bardini M, Fazio G, Chiorazzi A, Meregalli C, Oggioni N, Shanks K, Quartu M, Serra MP, Sala B, Cavaletti G, Dorsey SG. Bortezomib-induced painful peripheral neuropathy: an electrophysiological, behavioral, morphological and mechanistic study in the mouse. PLoS One 2013; 8:e72995. [PMID: 24069168 PMCID: PMC3772181 DOI: 10.1371/journal.pone.0072995] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 07/23/2013] [Indexed: 11/18/2022] Open
Abstract
Bortezomib is the first proteasome inhibitor with significant antineoplastic activity for the treatment of relapsed/refractory multiple myeloma as well as other hematological and solid neoplasms. Peripheral neurological complications manifesting with paresthesias, burning sensations, dysesthesias, numbness, sensory loss, reduced proprioception and vibratory sensitivity are among the major limiting side effects associated with bortezomib therapy. Although bortezomib-induced painful peripheral neuropathy is clinically easy to diagnose and reliable models are available, its pathophysiology remains partly unclear. In this study we used well-characterized immune-competent and immune-compromised mouse models of bortezomib-induced painful peripheral neuropathy. To characterize the drug-induced pathological changes in the peripheral nervous system, we examined the involvement of spinal cord neuronal function in the development of neuropathic pain and investigated the relevance of the immune response in painful peripheral neuropathy induced by bortezomib. We found that bortezomib treatment induced morphological changes in the spinal cord, dorsal roots, dorsal root ganglia (DRG) and peripheral nerves. Neurophysiological abnormalities and specific functional alterations in Aδ and C fibers were also observed in peripheral nerve fibers. Mice developed mechanical allodynia and functional abnormalities of wide dynamic range neurons in the dorsal horn of spinal cord. Bortezomib induced increased expression of the neuronal stress marker activating transcription factor-3 in most DRG. Moreover, the immunodeficient animals treated with bortezomib developed a painful peripheral neuropathy with the same features observed in the immunocompetent mice. In conclusion, this study extends the knowledge of the sites of damage induced in the nervous system by bortezomib administration. Moreover, a selective functional vulnerability of peripheral nerve fiber subpopulations was found as well as a change in the electrical activity of wide dynamic range neurons of dorsal horn of spinal cord. Finally, the immune response is not a key factor in the development of morphological and functional damage induced by bortezomib in the peripheral nervous system.
Collapse
Affiliation(s)
- Valentina A. Carozzi
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
- * E-mail: (VAC)
| | - Cynthia L. Renn
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| | - Michela Bardini
- “M. Tettamanti” Research Center, Department of Health Sciences, University of Milan Bicocca, Monza, Italy
| | - Grazia Fazio
- “M. Tettamanti” Research Center, Department of Health Sciences, University of Milan Bicocca, Monza, Italy
| | - Alessia Chiorazzi
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Cristina Meregalli
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Norberto Oggioni
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Kathleen Shanks
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| | - Marina Quartu
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Italy
| | - Maria Pina Serra
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Italy
| | - Barbara Sala
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Guido Cavaletti
- Department of Surgery and Translational Medicine, University of Milan Bicocca, Monza, Italy
| | - Susan G. Dorsey
- School of Nursing, Center for Pain Studies, University of Maryland, Baltimore, Maryland, United States of America
| |
Collapse
|
89
|
Abstract
A subcutaneous formulation of bortezomib is now indicated in the EU and the US for the treatment of patients with multiple myeloma. This article reviews pharmacological, therapeutic efficacy and tolerability data relevant to the utilization of subcutaneous bortezomib (Velcade(®)) in the treatment of patients with multiple myeloma. In a randomized, nonblind, phase III study, subcutaneous bortezomib was noninferior to intravenous bortezomib in the treatment of adults with relapsed multiple myeloma, as determined by the overall response rate after four cycles of therapy (primary endpoint). No significant differences between the subcutaneous and intravenous bortezomib formulations were observed in the median time to first response, median progression-free survival, median time to progression and 1-year overall survival. Compared with intravenous bortezomib, subcutaneous bortezomib confers a significant advantage with respect to the incidence of peripheral neuropathy (all grades, grade ≥2 and grade ≥3). As a consequence, it provides a new treatment option for patients with multiple myeloma, particularly those with pre-existing neuropathy or at a high risk of developing peripheral neuropathy.
Collapse
|
90
|
|
91
|
Bortezomib for patients with previously untreated multiple myeloma: a systematic review and meta-analysis of randomized controlled trials. Ann Hematol 2013; 92:935-43. [DOI: 10.1007/s00277-013-1711-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/15/2013] [Indexed: 10/27/2022]
|
92
|
Du XL, Chen Q. Recent advancements of bortezomib in acute lymphocytic leukemia treatment. Acta Haematol 2013; 129:207-14. [PMID: 23295437 DOI: 10.1159/000345260] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 10/03/2012] [Indexed: 12/21/2022]
Abstract
Although survival rates for acute lymphocytic leukemia (ALL), especially in children, have shown dramatic improvement over time, poor outcomes are still observed in patients who have refractory or relapsed disease after conventional chemotherapy. New therapeutic options are urgently needed. Bortezomib (Velcade, formerly PS-341) is the first proteasome inhibitor approved by the US FDA for the treatment of newly diagnosed multiple myeloma and relapsed/refractory multiple myeloma and mantle cell lymphoma. Although the mechanisms of bortezomib anticancer activity are still not completely understood, it is a new treatment option for patients with refractory or relapsed ALL, particularly when used in combination with conventional chemotherapy or targeted agents. This review summarizes recent advancements in the understanding of the bortezomib molecular mechanism of action in ALL. Understanding of the molecular approaches might help customize cancer chemotherapy for each individual patient, directing the field towards rational therapeutics.
Collapse
Affiliation(s)
- Xiao-Li Du
- Department of Hematology, The Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, China
| | | |
Collapse
|
93
|
Gilbar PJ, Seger AC. Fatalities Resulting From Accidental Intrathecal Administration of Bortezomib: Strategies for Prevention. J Clin Oncol 2012; 30:3427-8. [DOI: 10.1200/jco.2012.44.5866] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Andrew C. Seger
- Brigham and Women's Hospital; Center for Patient Safety, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
94
|
Abstract
Medication errors due to the inadvertent intrathecal administration of vincristine and other antineoplastic agents continue to occur despite the development of preventative strategies. Three fatalities due to bortezomib being accidentally given intrathecally instead of by the intended intravenous route have recently been reported by the European Medicines Agency. The most effective method for preventing accidental intrathecal administration is to eliminate the syringe as a means of administrating neurotoxic agents and prepare them in a small volume minibag. However due to a lack of stability data for bortezomib in a minibag and the increasing use of bortezomib via the subcutaneous route necessitates the continued preparation of bortezomib in a syringe. A number of recommendations aimed at preventing the possibility of accidental intrathecal administration of bortezomib are made. These need to be incorporated into standard practice internationally and pharmacists must take the lead to ensure this occurs as a matter of urgency.
Collapse
|
95
|
Meregalli C, Ceresa C, Canta A, Carozzi VA, Chiorazzi A, Sala B, Oggioni N, Lanza M, Letari O, Ferrari F, Avezza F, Marmiroli P, Caselli G, Cavaletti G. CR4056, a new analgesic I2 ligand, is highly effective against bortezomib-induced painful neuropathy in rats. J Pain Res 2012; 5:151-67. [PMID: 22792002 PMCID: PMC3392714 DOI: 10.2147/jpr.s32122] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although bortezomib (BTZ) is the frontline treatment for multiple myeloma, its clinical use is limited by the occurrence of painful peripheral neuropathy, whose treatment is still an unmet clinical need. Previous studies have shown chronic BTZ administration (0.20 mg/kg intravenously three times a week for 8 weeks) to female Wistar rats induced a peripheral neuropathy similar to that observed in humans. In this animal model of BTZ-induced neurotoxicity, the present authors evaluated the efficacy of CR4056, a novel I2 ligand endowed with a remarkable efficacy in several animal pain models. CR4056 was administered in a wide range of doses (0.6-60 mg/kg by gavage every day for 2-3 weeks) in comparison with buprenorphine (Bupre) (28.8 μg/kg subcutaneously every day for 2 weeks) and gabapentin (Gaba) (100 mg/kg by gavage every day for 3 weeks). Chronic administration of BTZ reduced nerve conduction velocity and induced allodynia. CR4056, Bupre, or Gaba did not affect the impaired nerve conduction velocity. Conversely, CR4056 dose-dependently reversed BTZ-induced allodynia (minimum effective dose 0.6 mg/kg). The optimal dose found, 6 mg/kg, provided a constant pain relief throughout the treatment period and without rebound after suspension, being effective when coadministered with BTZ, starting before or after allodynia was established, or when administered alone after BTZ cessation. A certain degree of tolerance was seen after 7 days of administration, but only at the highest doses (20 and 60 mg/kg). Bupre was effective only acutely, since tolerance was evident from the fourth day onwards. Gaba showed a significant activity only at the fourth day of treatment. CR4056, over the range of concentrations of 3-30 μM, was unable to hinder BTZ cytotoxicity on several tumor cell lines, which could indicate that this substance does not directly interfere with BTZ antitumor activity. Therefore, CR4056 could represent a new treatment option for BTZ-induced neuropathic pain.
Collapse
Affiliation(s)
- Cristina Meregalli
- Department of Neuroscience and Biomedical Technologies, University of Milan-Bicocca
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Smoum R, Rubinstein A, Dembitsky VM, Srebnik M. Boron containing compounds as protease inhibitors. Chem Rev 2012; 112:4156-220. [PMID: 22519511 DOI: 10.1021/cr608202m] [Citation(s) in RCA: 320] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Reem Smoum
- The School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel.
| | | | | | | |
Collapse
|
97
|
Karman J, Gumlaw NK, Zhang J, Jiang JL, Cheng SH, Zhu Y. Proteasome inhibition is partially effective in attenuating pre-existing immunity against recombinant adeno-associated viral vectors. PLoS One 2012; 7:e34684. [PMID: 22514654 PMCID: PMC3326043 DOI: 10.1371/journal.pone.0034684] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/08/2012] [Indexed: 12/12/2022] Open
Abstract
Pre-existing immunity against adeno-associated virus (AAV) remains a major challenge facing the clinical use of systemic administration of recombinant AAV vectors for the treatment of genetic and acquired diseases using gene therapy. In this study, we evaluated the potential of bortezomib (marketed under trade name Velcade) to abrogate a pre-existing immunity to AAV in mice, thereby allowing subsequent transduction by a recombinant AAV vector of the same serotype. We demonstrate that bortezomib efficiently reduces AAV-specific IgG titres and moderates the cytotoxic T cell response in mice that have a pre-existing immunity to AAV2/8. Significant depletion of AAV2/8-specific IgG-producing plasma cells in secondary lymphoid organs and bone marrow was observed. However, this inhibition of the immune response by bortezomib was insufficient to allow subsequent re-infection with a recombinant AAV vector of a similar serotype. We show that this shortcoming is probably due to the combination of residual antibody levels and the inability of bortezomib to completely deplete the memory B cells that are re-activated in response to a repeated infection with a recombinant AAV vector. Taken together, the results of this study argue for the use of immunosuppressive therapies that target both plasma and memory B cells for the efficient elimination of pre-existing immunity against AAV2/8 vectors.
Collapse
Affiliation(s)
- Jozsef Karman
- Genetic Disease Science, Genzyme Corporation, Framingham, Massachusetts, United States of America.
| | | | | | | | | | | |
Collapse
|
98
|
Gazić Smilović I, Casas-Arcé E, Roseblade SJ, Nettekoven U, Zanotti-Gerosa A, Kovačevič M, Časar Z. Iridium-Catalyzed Chemoselective and Enantioselective Hydrogenation of (1-Chloro-1-Alkenyl) Boronic Esters. Angew Chem Int Ed Engl 2011; 51:1014-8. [DOI: 10.1002/anie.201106262] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Indexed: 02/04/2023]
|
99
|
Gazić Smilović I, Casas-Arcé E, Roseblade SJ, Nettekoven U, Zanotti-Gerosa A, Kovačevič M, Časar Z. Iridium-Catalyzed Chemoselective and Enantioselective Hydrogenation of (1-Chloro-1-Alkenyl) Boronic Esters. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201106262] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
100
|
Kihm LP, Zeier M, Morath C. Emerging drugs for the treatment of transplant rejection. Expert Opin Emerg Drugs 2011; 16:683-95. [DOI: 10.1517/14728214.2011.641012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|