1
|
Alpha-lipoic acid alters the antitumor effect of bortezomib in melanoma cells in vitro. Sci Rep 2020; 10:14287. [PMID: 32868799 PMCID: PMC7459291 DOI: 10.1038/s41598-020-71138-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 08/04/2020] [Indexed: 12/27/2022] Open
Abstract
Bortezomib (BOZ) is a proteasome inhibitor chemotherapeutic agent utilized to treat multiple myeloma and recently offered to cure melanoma. Bortezomib-induced neuropathy is one of the dose-limiting side-effects, which can be treated with antioxidants (e.g. alpha-lipoic acid-ALA and Vitamin B1-vit B1). We hypothesized that these antioxidants may counteract the antitumor activity by disrupting the BOZ-induced pathways (e.g. proteasome inhibition or reactive oxygen species generation). The objectives were: (i) to verify the anti-proliferative effect of BOZ; (ii) to compare the influence of the antioxidants on the antitumor effect of BOZ in melanoma (A2058) and myeloma (U266) cells. At first, the reduction in the anti-proliferative effect of BOZ by ALA was proved in melanoma cells. Analysis of p53 phosphorylation and the cell cycle progression revealed that ALA failed to counteract these effects of BOZ. Nevertheless, a good correlation was found between the inhibition of the anti-proliferative effect, the anti-proteasome activity and the oxidative stress level after the co-treatment with 20 ng/mL BOZ + 100 μg/mL ALA. Downregulation of apoptotic proteins such as HO-1 and Claspin along with the inhibition of the cleavage of Caspase-3 indicated the proteomic background of the altered responsiveness of the melanoma cells exposed to BOZ + ALA. This phenomenon draws attention to the proper application of cancer supportive care to avoid possible interactions.
Collapse
|
2
|
Kim Y, Lee J, Lee D, Othmer HG. Synergistic Effects of Bortezomib-OV Therapy and Anti-Invasive Strategies in Glioblastoma: A Mathematical Model. Cancers (Basel) 2019; 11:E215. [PMID: 30781871 PMCID: PMC6406513 DOI: 10.3390/cancers11020215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
It is well-known that the tumor microenvironment (TME) plays an important role in the regulation of tumor growth and the efficacy of anti-tumor therapies. Recent studies have demonstrated the potential of combination therapies, using oncolytic viruses (OVs) in conjunction with proteosome inhibitors for the treatment of glioblastoma, but the role of the TME in such therapies has not been studied. In this paper, we develop a mathematical model for combination therapies based on the proteosome inhibitor bortezomib and the oncolytic herpes simplex virus (oHSV), with the goal of understanding their roles in bortezomib-induced endoplasmic reticulum (ER) stress, and how the balance between apoptosis and necroptosis is affected by the treatment protocol. We show that the TME plays a significant role in anti-tumor efficacy in OV combination therapy, and illustrate the effect of different spatial patterns of OV injection. The results illustrate a possible phenotypic switch within tumor populations in a given microenvironment, and suggest new anti-invasion therapies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Junho Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
3
|
Salama Y, Lin SY, Dhahri D, Hattori K, Heissig B. The fibrinolytic factor tPA drives LRP1-mediated melanoma growth and metastasis. FASEB J 2018; 33:3465-3480. [PMID: 30458112 DOI: 10.1096/fj.201801339rrr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The multifunctional endocytic receptor low-density lipoprotein receptor-related protein (LRP)1 has recently been identified as a hub within a biomarker network for multicancer clinical outcome prediction. The mechanism how LRP1 modulates cancer progression is poorly understood. In this study we found that LRP1 and one of its ligands, tissue plasminogen activator (tPA), are expressed in melanoma cells and control melanoma growth and lung metastasis in vivo. Mechanistic studies were performed on 2 melanoma cancer cell lines, B16F10 and the B16F1 cells, both of which form primary melanoma tumors, but only B16F10 cells metastasize to the lungs. Tumor-, but not niche cell-derived tPA, enhanced melanoma cell proliferation in tPA-/- mice. Gain-of-function experiments revealed that melanoma LRP1 is critical for tumor growth, recruitment of mesenchymal stem cells into the tumor bed, and metastasis. Melanoma LRP1 was found to enhance ERK activation, resulting in increased matrix metalloproteinase (MMP)-9 RNA, protein, and secreted activity, a well-known modulator of melanoma metastasis. Restoration of LRP1 and tPA in the less aggressive, poorly metastatic B16F1 tumor cells enhanced tumor cell proliferation and led to massive lung metastasis in murine tumor models. Antimelanoma drug treatment induced tPA and LRP1 expression. tPA or LRP1 knockdown enhanced chemosensitivity in melanoma cells. Our results identify the tPA-LRP1 pathway as a key switch that drives melanoma progression, in part by modulating the cellular composition and proteolytic makeup of the tumor niche. Targeting this pathway may be a novel treatment strategy in combination treatments for melanoma.-Salama, Y., Lin, S.-Y., Dhahri, D., Hattori, K., Heissig, B. The fibrinolytic factor tPA drives LRP1-mediated melanoma growth and metastasis.
Collapse
Affiliation(s)
- Yousef Salama
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Shiou-Yuh Lin
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Douaa Dhahri
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Hattori
- Center for Genome and Regenerative Medicine Juntendo University School of Medicine, Tokyo, Japan; and
| | - Beate Heissig
- Division of Stem Cell Dynamics, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Atopy (Allergy) Center, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Suarez-Kelly LP, Kemper GM, Duggan MC, Stiff A, Noel TC, Markowitz J, Luedke EA, Yildiz VO, Yu L, Jaime-Ramirez AC, Karpa V, Zhang X, Carson WE. The combination of MLN2238 (ixazomib) with interferon-alpha results in enhanced cell death in melanoma. Oncotarget 2018; 7:81172-81186. [PMID: 27783987 PMCID: PMC5348384 DOI: 10.18632/oncotarget.12791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 10/10/2016] [Indexed: 11/25/2022] Open
Abstract
The ubiquitin-proteasome signaling pathway is critical for cell cycle regulation and neoplastic growth. Proteasome inhibition can activate apoptotic pathways. Bortezomib, a selective proteasome inhibitor, has anti-melanoma activity. MLN2238 (ixazomib), an oral proteasome inhibitor, has improved pharmacotherapeutic parameters compared to bortezomib. Interferon-alpha (IFN-α), an immune boosting agent, is FDA-approved for treatment of melanoma. In this study in vitro and in vivo evaluation of the antitumor potential of ixazomib and combination treatments with ixazomib and IFN-α were performed. Apoptosis induced by ixazomib was first observed at 12 hours and was maximal at 48 hours with similar levels of cell death compared to bortezomib. IFN-α alone had little effect on cell viability in vitro. However, the combination of ixazomib with IFN-α significantly enhanced ixazomib's ability to induce apoptotic cell death in BRAF V600E mutant and BRAF wild-type human melanoma tumor cells. The combination of ixazomib and IFN-α also enhanced inhibition of cell proliferation in BRAF V600E mutant melanoma tumor cells; however, this was not seen in BRAF wild-type cells. Ixazomib-induced apoptosis was associated with processing of the pro-apoptotic proteins procaspase-3, -7, -8, and -9, and cleavage of poly-ADP-ribose polymerase (PARP). In an in vivo xenograft model of human melanoma, combination treatment with IFN-α-2b and ixazomib demonstrated a significant reduction in tumor volume when compared to vehicle (p = 0.005) and single therapy ixazomib (p = 0.017) and IFN-α-2b (p = 0.036). These pre-clinical results support further evaluation of combination treatment with ixazomib and IFN-α for the treatment of advanced BRAF V600E mutant melanoma.
Collapse
Affiliation(s)
- Lorena P Suarez-Kelly
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Gregory M Kemper
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Megan C Duggan
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Andrew Stiff
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Tiffany C Noel
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Joseph Markowitz
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus OH, USA
| | - Eric A Luedke
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA.,Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus OH, USA
| | - Vedat O Yildiz
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Alena Cristina Jaime-Ramirez
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Volodymyr Karpa
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Xiaoli Zhang
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - William E Carson
- The Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, USA.,Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus OH, USA
| |
Collapse
|
5
|
Perez M, Peinado-Serrano J, Garcia-Heredia JM, Felipe-Abrio I, Tous C, Ferrer I, Martin-Broto J, Saez C, Carnero A. Efficacy of bortezomib in sarcomas with high levels of MAP17 (PDZK1IP1). Oncotarget 2018; 7:67033-67046. [PMID: 27563810 PMCID: PMC5341855 DOI: 10.18632/oncotarget.11475] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/09/2016] [Indexed: 11/25/2022] Open
Abstract
Sarcomas are malignant tumors accounting for a high percentage of cancer morbidity and mortality in children and young adults. Surgery and radiation therapy are the accepted treatments for most sarcomas; however, patients with metastatic disease are treated with systemic chemotherapy. Many tumors display marginal levels of chemoresponsiveness, and new treatment approaches are needed. MAP17 is a small non-glycosylated membrane protein overexpressed in carcinomas. The levels of MAP17 could be used as a prognostic marker to predict the response to bortezomib in hematological malignancies and in breast tumors. Therefore, we analyzed the expression of this oncogene in sarcomas and its relationship with clinico-pathological features, as well as tested whether it can be used as a new biomarker to predict the therapeutic response to bortezomib and new therapies for sarcomas. We found that the levels of MAP17 were related to clinical features and poor survival in a cohort of 69 patients with different sarcoma types, not being restricted to any special subtype of tumor. MAP17 expression is associated with poor overall survival (p<0.001) and worse disease-free survival (p=0.002). Cell lines with high levels of MAP17 show a better response to bortezomib in vitro. Furthermore, patient-derived xenografts (PDX) with high levels of MAP17 respond to bortezomib in vivo. Our results showed that this response is due to the lower levels of NFκB and autophagy activation. Therefore, we suggest that MAP17 is a new biomarker to predict the efficacy of bortezomib as a new therapy for sarcomas.
Collapse
Affiliation(s)
- Marco Perez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Javier Peinado-Serrano
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Jose Manuel Garcia-Heredia
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain
| | - Irene Felipe-Abrio
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Cristina Tous
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Irene Ferrer
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Javier Martin-Broto
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain.,Department of Medical Oncology, Virgen del Rocío University Hospital, Seville, Spain
| | - Carmen Saez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain.,Department of Pathology, Virgen del Rocío University Hospital, Seville, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| |
Collapse
|
6
|
Friedman AA, Xia Y, Trippa L, Le LP, Igras V, Frederick DT, Wargo JA, Tanabe KK, Lawrence DP, Neuberg DS, Flaherty KT, Fisher DE. Feasibility of Ultra-High-Throughput Functional Screening of Melanoma Biopsies for Discovery of Novel Cancer Drug Combinations. Clin Cancer Res 2017; 23:4680-4692. [PMID: 28446504 DOI: 10.1158/1078-0432.ccr-16-3029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/22/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Purpose: Successful development of targeted therapy combinations for cancer patients depends on first discovering such combinations in predictive preclinical models. Stable cell lines and mouse xenograft models can have genetic and phenotypic drift and may take too long to generate to be useful as a personalized medicine tool.Experimental Design: To overcome these limitations, we have used a platform of ultra-high-throughput functional screening of primary biopsies preserving both cancer and stroma cell populations from melanoma patients to nominate such novel combinations from a library of thousands of drug combinations in a patient-specific manner within days of biopsy. In parallel, patient-derived xenograft (PDX) mouse models were created and novel combinations tested for their ability to shrink matched PDXs.Results: The screening method identifies specific drug combinations in tumor cells with patterns that are distinct from those obtained from stable cell lines. Screening results were highly specific to individual patients. For patients with matched PDX models, we confirmed that individualized novel targeted therapy combinations could inhibit tumor growth. In particular, a combination of multi-kinase and PI3K/Akt inhibitors was effective in some BRAF-wild-type melanomas, and the addition of cediranib to the BRAF inhibitor PLX4720 was effective in a PDX model with BRAF mutation.Conclusions: This proof-of-concept study demonstrates the feasibility of using primary biopsies directly for combinatorial drug discovery, complementing stable cell lines and xenografts, but with much greater speed and efficiency. This process could potentially be used in a clinical setting to rapidly identify therapeutic strategies for individual patients. Clin Cancer Res; 23(16); 4680-92. ©2017 AACR.
Collapse
Affiliation(s)
- Adam A Friedman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Yun Xia
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts.,Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lorenzo Trippa
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Long Phi Le
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Vivien Igras
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Dennie T Frederick
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jennifer A Wargo
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Division of Surgical Oncology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Kenneth K Tanabe
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.,Division of Surgical Oncology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Donald P Lawrence
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Donna S Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - David E Fisher
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts. .,Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts
| |
Collapse
|
7
|
The Proteasome Inhibitor Bortezomib Affects Chondrosarcoma Cells via the Mitochondria-Caspase Dependent Pathway and Enhances Death Receptor Expression and Autophagy. PLoS One 2016; 11:e0168193. [PMID: 27978543 PMCID: PMC5158315 DOI: 10.1371/journal.pone.0168193] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/28/2016] [Indexed: 01/19/2023] Open
Abstract
High grade chondrosarcoma is characterized by its lack of response to conventional cytotoxic chemotherapy, the tendency to develop lung metastases, and low survival rates. Research within the field prioritizes the development and expansion of new treatment options for dealing with unresectable or metastatic diseases. Numerous clinical trials using the proteasome inhibitor bortezomib have shown specific efficacy as an active antitumor agent for treating a variety of solid tumors. However, as of yet the effect of bortezomib on chondrosarcoma has not been investigated. In our study, bortezomib decreased cell viability and proliferation in two different chondrosarcoma cell lines in a time- and dose dependent manner. FACS analysis, mRNA- and protein expression studies illustrated that induction of apoptosis developed through the intrinsic mitochondria-caspase dependent pathway. Furthermore, bortezomib treatment significantly increased expression of the death receptors TRAILR-1 and TRAILR-2 in chondrosarcoma cells. An increased expression of the autophagy markers Atg5/12, Beclin, and LC3BI-II supports the interpretation that bortezomib functions as a trigger for autophagy. Our results demonstrated for the first time that bortezomib reduced viability and proliferation of chondrosarcoma cells, induced apoptosis via the mitochondria-caspase dependent pathway and enhanced death receptor expression and autophagy.
Collapse
|
8
|
Hassan M, Selimovic D, Hannig M, Haikel Y, Brodell RT, Megahed M. Endoplasmic reticulum stress-mediated pathways to both apoptosis and autophagy: Significance for melanoma treatment. World J Exp Med 2015; 5:206-217. [PMID: 26618107 PMCID: PMC4655250 DOI: 10.5493/wjem.v5.i4.206] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/29/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive form of skin cancer. Disrupted intracellular signaling pathways are responsible for melanoma's extraordinary resistance to current chemotherapeutic modalities. The pathophysiologic basis for resistance to both chemo- and radiation therapy is rooted in altered genetic and epigenetic mechanisms that, in turn, result in the impairing of cell death machinery and/or excessive activation of cell growth and survival-dependent pathways. Although most current melanoma therapies target mitochondrial dysregulation, there is increasing evidence that endoplasmic reticulum (ER) stress-associated pathways play a role in the potentiation, initiation and maintenance of cell death machinery and autophagy. This review focuses on the reliability of ER-associated pathways as therapeutic targets for melanoma treatment.
Collapse
|
9
|
Kardos GR, Robertson GP. Therapeutic interventions to disrupt the protein synthetic machinery in melanoma. Pigment Cell Melanoma Res 2015; 28:501-19. [PMID: 26139519 PMCID: PMC4716672 DOI: 10.1111/pcmr.12391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/30/2015] [Indexed: 01/23/2023]
Abstract
Control of the protein synthetic machinery is deregulated in many cancers, including melanoma, to increase the protein production. Tumor suppressors and oncogenes play key roles in protein synthesis from the transcription of rRNA and ribosome biogenesis to mRNA translation initiation and protein synthesis. Major signaling pathways are altered in melanoma to modulate the protein synthetic machinery, thereby promoting tumor development. However, despite the importance of this process in melanoma development, involvement of the protein synthetic machinery in this cancer type is an underdeveloped area of study. Here, we review the coupling of melanoma development to deregulation of the protein synthetic machinery. We examine existing knowledge regarding RNA polymerase I inhibition and mRNA translation focusing on their inhibition for therapeutic applications in melanoma. Furthermore, the contribution of amino acid biosynthesis and involvement of ribosomal proteins are also reviewed as future therapeutic strategies to target deregulated protein production in melanoma.
Collapse
Affiliation(s)
- Gregory R. Kardos
- Department of Pharmacology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
| | - Gavin P. Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- Department of Pathology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- Department of Dermatology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- Department of Surgery, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
- The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA, 17033
| |
Collapse
|
10
|
Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2015; 2:e974463. [PMID: 27308423 PMCID: PMC4904962 DOI: 10.4161/23723556.2014.974463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/12/2023]
Abstract
The so-called "ubiquitin-proteasome system" (UPS) is a multicomponent molecular apparatus that catalyzes the covalent attachment of several copies of the small protein ubiquitin to other proteins that are generally (but not always) destined to proteasomal degradation. This enzymatic cascade is crucial for the maintenance of intracellular protein homeostasis (both in physiological conditions and in the course of adaptive stress responses), and regulates a wide array of signaling pathways. In line with this notion, defects in the UPS have been associated with aging as well as with several pathological conditions including cardiac, neurodegenerative, and neoplastic disorders. As transformed cells often experience a constant state of stress (as a result of the hyperactivation of oncogenic signaling pathways and/or adverse microenvironmental conditions), their survival and proliferation are highly dependent on the integrity of the UPS. This rationale has driven an intense wave of preclinical and clinical investigation culminating in 2003 with the approval of the proteasomal inhibitor bortezomib by the US Food and Drug Administration for use in multiple myeloma patients. Another proteasomal inhibitor, carfilzomib, is now licensed by international regulatory agencies for use in multiple myeloma patients, and the approved indications for bortezomib have been extended to mantle cell lymphoma. This said, the clinical activity of bortezomib and carfilzomib is often limited by off-target effects, innate/acquired resistance, and the absence of validated predictive biomarkers. Moreover, the antineoplastic activity of proteasome inhibitors against solid tumors is poor. In this Trial Watch we discuss the contribution of the UPS to oncogenesis and tumor progression and summarize the design and/or results of recent clinical studies evaluating the therapeutic profile of proteasome inhibitors in cancer patients.
Collapse
Affiliation(s)
- Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “Tor Vergata”
| | - Lorenzo Galluzzi
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
11
|
Mairinger FD, Walter RFH, Theegarten D, Hager T, Vollbrecht C, Christoph DC, Worm K, Ting S, Werner R, Stamatis G, Mairinger T, Baba H, Zarogoulidis K, Huang H, Li Q, Tsakiridis K, Zarogoulidis P, Schmid KW, Wohlschlaeger J. Gene Expression Analysis of the 26S Proteasome Subunit PSMB4 Reveals Significant Upregulation, Different Expression and Association with Proliferation in Human Pulmonary Neuroendocrine Tumours. J Cancer 2014; 5:646-54. [PMID: 25157275 PMCID: PMC4142326 DOI: 10.7150/jca.9955] [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: 05/17/2014] [Accepted: 06/20/2014] [Indexed: 12/23/2022] Open
Abstract
Background: Proteasomal subunit PSMB4 was suggested to be a survival gene in an animal model of hepatocellular carcinoma and in glioblastoma cell lines. In pulmonary adenocarcinoma, a high expression of these genes was found to be associated with poor differentiation and survival. This study investigates the gene expression levels of 26S proteasome subunits in human pulmonary neuroendocrine tumours including typical (TC) and atypical (AC) carcinoid tumours as well as small cell (SCLC) and large cell (LCNEC) neuroendocrine carcinomas. Material and methods: Gene expression levels of proteasomal subunits (PSMA1, PSMA5, PSMB4, PSMB5 and PSMD1) were investigated in 80 neuroendocrine pulmonary tumours (each 20 TC, AC, LCNLC and SCLC) and compared to controls. mRNA levels were determined by using TaqMan assays. Immunohistochemistry on tissue microarrays (TMA) was performed to determine the expression of ki67, cleaved caspase 3 and PSMB4. Results: All proteasomal subunit gene expressions were significantly upregulated in TC, AC, SCLC and LCNEC compared to controls. PSMB4 mRNA is differently expressed between all neuroendocrine tumour subtypes demonstrating the highest expression and greatest range in LCNEC (p=0.043), and is significantly associated with proliferative activity (p=0.039). Conclusion: In line with other 26S proteasomal subunits PSMB4 is significantly increased, but differently expressed between pulmonary neuroendocrine tumours and is associated with the proliferative activity. Unlike in pulmonary adenocarcinomas, no association with biological behaviour was observed, suggesting that increased proteasomal subunit gene expression is a common and probably early event in the tumorigenesis of pulmonary neuroendocrine tumours regardless of their differentiation.
Collapse
Affiliation(s)
| | - Robert Fred Henry Walter
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany. ; 2. Ruhrlandklinik, West German Lung Centre, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Dirk Theegarten
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Thomas Hager
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Claudia Vollbrecht
- 3. Institute of Pathology, University Hospital Cologne, University of Cologne, Germany
| | | | - Karl Worm
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Saskia Ting
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Robert Werner
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Georgios Stamatis
- 5. Department of Thoracic Surgery, Ruhrlandklinik, West German Lung Centre, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Thomas Mairinger
- 6. Department of Pathology, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Hideo Baba
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Konstantinos Zarogoulidis
- 7. Pulmonary-Oncology, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haidong Huang
- 8. Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, People's Republic of China, China
| | - Qiang Li
- 8. Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, People's Republic of China, China
| | - Kosmas Tsakiridis
- 9. Thoracic Surgery Department, ``Saint Luke`` Private Clinic, Thessaloniki, Panorama, Greece
| | - Paul Zarogoulidis
- 7. Pulmonary-Oncology, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kurt Werner Schmid
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Jeremias Wohlschlaeger
- 1. Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany
| |
Collapse
|