1
|
Tarjányi O, Olasz K, Rátky F, Sétáló G, Boldizsár F. Proteasome Inhibitors: Potential in Rheumatoid Arthritis Therapy? Int J Mol Sci 2025; 26:2943. [PMID: 40243560 PMCID: PMC11988683 DOI: 10.3390/ijms26072943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to the destruction of peripheral joint cartilage and bone tissue. Despite the advent of biological therapies in the past decades, the complete remission of RA patients is still out of reach. Therefore, the search for novel therapeutic approaches is still open in the field of RA. Proteasome inhibitors (PIs) were originally designed to be used in hematological malignancies like multiple myeloma. However, evidence has shown that they are potent inhibitors of the NF-κB pathway, which plays a pivotal role in inflammatory processes and RA. Furthermore, inhibition of cell activation and induction of apoptosis was also reported about PIs. In the present review, we summarize the current knowledge about the potential effects of PIs in RA based on reports from animal and human studies. We believe that there is substantial potential in the use of PIs in RA therapy either alone or in combination with the medications already used.
Collapse
Affiliation(s)
- Oktávia Tarjányi
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - Katalin Olasz
- Department of Immunology and Biotechnology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| | - Fanni Rátky
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - György Sétáló
- Department of Medical Biology, Medical School, University of Pecs, H-7624 Pecs, Hungary; (O.T.); (F.R.); (G.S.)
| | - Ferenc Boldizsár
- Department of Immunology and Biotechnology, Medical School, University of Pecs, H-7624 Pecs, Hungary;
| |
Collapse
|
2
|
Plakoula E, Kalampounias G, Alexis S, Verigou E, Kourakli A, Zafeiropoulou K, Symeonidis A. Prognostic Value of PSMB5 and Correlations with LC3II and Reactive Oxygen Species Levels in the Bone Marrow Mononuclear Cells of Bortezomib-Resistant Multiple Myeloma Patients. Curr Issues Mol Biol 2025; 47:32. [PMID: 39852147 PMCID: PMC11763810 DOI: 10.3390/cimb47010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Proteasome inhibitors (PIs) constitute the most common type of induction treatment for multiple myeloma. Interactions between the proteasome, autophagy, and reactive oxygen species (ROS) have been shown in the past, thus emphasizing the need for a better understanding of the underlying pathophysiology. For this study, bone marrow mononuclear cells from 110 myeloma patients were collected at different disease stages. PSMB5 and LC3I/II protein levels were determined using Western blot, proteasome proteolytic activity (PPA) with spectrofluorometry, and ROS with flow cytometry. PSMB5 accumulation was found to diminish after PI treatment (p-value = 0.014), and the same pattern was observed in PPA (p-value < 0.001). Conversely, LC3II protein levels were elevated at both remission and relapse compared to baseline levels (p-value = 0.041). Patients with a baseline PSMB5 accumulation lower than 1.06 units had longer disease-free survival compared to those with values above 1.06 units (12.0 ± 6.7 vs. 36 ± 12.1 months; p-value < 0.001). Median ROS levels in plasma cells were significantly higher at relapse compared to both baseline and remission levels (p-value < 0.001), implying poor prognosis. Overall, post-treatment PSMB5 reduction could indicate a shift from proteasomal to autophagic degradation as a main proteostatic mechanism, thus explaining resistance. The elevated oxidative stress in PI-treated patients could possibly serve as an additional compensatory mechanism.
Collapse
Affiliation(s)
- Eva Plakoula
- Hematology Division, Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece; (E.P.); (S.A.); (E.V.); (K.Z.)
| | - Georgios Kalampounias
- Division of Genetics, Cell Biology and Development, Department of Biology, School of Natural Sciences, University of Patras, 26504 Patras, Greece;
| | - Spyridon Alexis
- Hematology Division, Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece; (E.P.); (S.A.); (E.V.); (K.Z.)
| | - Evgenia Verigou
- Hematology Division, Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece; (E.P.); (S.A.); (E.V.); (K.Z.)
| | - Alexandra Kourakli
- Department of Hematology, OLYMPION General Hospital, Volou & Meilichou Str., 26443 Patras, Greece;
| | - Kalliopi Zafeiropoulou
- Hematology Division, Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece; (E.P.); (S.A.); (E.V.); (K.Z.)
| | - Argiris Symeonidis
- Hematology Division, Department of Medicine, School of Health Sciences, University of Patras, 26504 Patras, Greece; (E.P.); (S.A.); (E.V.); (K.Z.)
- Department of Hematology, OLYMPION General Hospital, Volou & Meilichou Str., 26443 Patras, Greece;
| |
Collapse
|
3
|
Song X, Zhang M, Chen M, Shang X, Zhou F, Yu H, Song C, Tan Q. Transcriptomic Communication between Nucleus and Mitochondria during the Browning Process of Lentinula edodes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23592-23605. [PMID: 39382068 DOI: 10.1021/acs.jafc.4c03506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
To explore the reason for cytoplasmic replacement's significant effect on browning, transcriptomic data of nuclear (N) and mitochondrial (M) mRNAs and long noncoding RNAs (lncRNAs) in L808 and two cytoplasmic hybrids (cybrids) (L808-A2 and L808-B) of Lentinula edodes at three different culturing times (80, 100, and 120 days) were obtained. The results showed that the expression of N and M genes and lncRNAs changed with the culture time and cytoplasmic source. Cytoplasmic replacement significantly affected some M and N genes related to the internal mechanism and external morphological characteristics of L. edodes browning. The internal browning mechanism should be the nicotinamide adenine dinucleotide phosphate (NADPH)-mediated antioxidant machinery to protect mycelia against oxidative stress induced by the generation of reactive oxygen species under light irradiation. External morphological characteristics were the changing features of brown films by melanin (an antioxidant) aggregation on the surface of the mycelia of the bag or log. Especially, some genes were related to the remodeling of the plasma membrane, extracellular enzymes of celluloses and hemicellulases, small molecules, and NADPH metabolic processes. Additionally, communication between the nucleus and mitochondria mediated by M-rps3 was reported for the first time, and it is mainly appreciated in M structural assembly, functional implementation, and cooperation with other organelles.
Collapse
Affiliation(s)
- Xiaoxia Song
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Meiyan Zhang
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Mingjie Chen
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Xiaodong Shang
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Feng Zhou
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Hailong Yu
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Chunyan Song
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Qi Tan
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| |
Collapse
|
4
|
Shi S, Ou X, Liu C, Wen H, Jiang K. Immunoproteasome acted as immunotherapy 'coffee companion' in advanced carcinoma therapy. Front Immunol 2024; 15:1464267. [PMID: 39281672 PMCID: PMC11392738 DOI: 10.3389/fimmu.2024.1464267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Immunoproteasome is a specialized form of proteasome which plays a crucial role in antigen processing and presentation, and enhances immune responses against malignant cells. This review explores the role of immunoproteasome in the anti-tumor immune responses, including immune surveillance and modulation of the tumor microenvironment, as well as its potential as a target for cancer immunotherapy. Furthermore, we have also discussed the therapeutic potential of immunoproteasome inhibitors, strategies to enhance antigen presentation and combination therapies. The ongoing trials and case studies in urology, melanoma, lung, colorectal, and breast cancers have also been summarized. Finally, the challenges facing clinical translation of immunoproteasome-targeted therapies, such as toxicity and resistance mechanisms, and the future research directions have been addressed. This review underscores the significance of targeting the immunoproteasome in combination with other immunotherapies for solid tumors and its potential broader applications in other diseases.
Collapse
Affiliation(s)
- Shaoyan Shi
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chao Liu
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hao Wen
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ke Jiang
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
5
|
Tyrna P, Procyk G, Szeleszczuk Ł, Młynarczuk-Biały I. Different Strategies to Overcome Resistance to Proteasome Inhibitors-A Summary 20 Years after Their Introduction. Int J Mol Sci 2024; 25:8949. [PMID: 39201634 PMCID: PMC11354503 DOI: 10.3390/ijms25168949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Proteasome inhibitors (PIs), bortezomib, carfilzomib, and ixazomib, are the first-line treatment for multiple myeloma (MM). They inhibit cytosolic protein degradation in cells, which leads to the accumulation of misfolded and malfunctioned proteins in the cytosol and endoplasmic reticulum, resulting in cell death. Despite being a breakthrough in MM therapy, malignant cells develop resistance to PIs via different mechanisms. Understanding these mechanisms drives research toward new anticancer agents to overcome PI resistance. In this review, we summarize the mechanism of action of PIs and how MM cells adapt to these drugs to develop resistance. Finally, we explore these mechanisms to present strategies to interfere with PI resistance. The strategies include new inhibitors of the ubiquitin-proteasome system, drug efflux inhibitors, autophagy disruption, targeting stress response mechanisms, affecting survival and cell cycle regulators, bone marrow microenvironment modulation, and immunotherapy. We list potential pharmacological targets examined in in vitro, in vivo, and clinical studies. Some of these strategies have already provided clinicians with new anti-MM medications, such as panobinostat and selinexor. We hope that further exploration of the subject will broaden the range of therapeutic options and improve patient outcomes.
Collapse
Affiliation(s)
- Paweł Tyrna
- Histology and Embryology Students’ Science Association, Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland;
| | - Izabela Młynarczuk-Biały
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland
| |
Collapse
|
6
|
Park JE, Chaudhary CL, Bhattarai D, Kim KB. Brain-Permeable Immunoproteasome-Targeting Macrocyclic Peptide Epoxyketones for Alzheimer's Disease. J Med Chem 2024; 67:7146-7157. [PMID: 38636481 PMCID: PMC11733980 DOI: 10.1021/acs.jmedchem.3c02488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Previously, we demonstrated that linear peptide epoxyketones targeting the immunoproteasome (iP) could ameliorate cognitive deficits in mouse models of Alzheimer's disease (AD) independently of amyloid deposition. We also reported the first iP-targeting macrocyclic peptide epoxyketones, which exhibit improved metabolic stability compared with their linear counterparts. Here, we prepared additional macrocyclic peptide epoxyketones and compared them with existing macrocyclic iP inhibitors by assessing Caco2 cell-based permeability and microsomal stability, providing the four best macrocyclic iP inhibitors. We then evaluated the four compounds using the Ames test and the potency assays in BV2 cells, selecting compound 5 as our AD drug lead. When 5 was administered intravenously (40 mg/kg) or orally (150 mg/kg) into healthy BALB/c mice, we observed considerable iP inhibition in the mouse brain, indicating good blood-brain barrier permeability and target engagement. Combined results suggest that 5 is a promising AD drug lead that may need further investigation.
Collapse
Affiliation(s)
- Ji Eun Park
- Center for Translational Science, Florida International University, Port St. Lucie, Florida 34987, United States
| | - Chhabi L. Chaudhary
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Deepak Bhattarai
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Kyung Bo Kim
- Center for Translational Science, Florida International University, Port St. Lucie, Florida 34987, United States; Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, United States
| |
Collapse
|
7
|
Mitsiades CS. Proteasome Inhibitors in Multiple Myeloma: Biological Insights on Mechanisms of Action or Resistance Informed by Functional Genomics. Hematol Oncol Clin North Am 2024; 38:321-336. [PMID: 38278626 DOI: 10.1016/j.hoc.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
During the last 20 years, proteasome inhibitors have been a cornerstone for the therapeutic management of multiple myeloma (MM). This review highlights how MM research has evolved over time in terms of our understanding of the mechanistic basis for the pronounced clinical activity of proteasome inhibitors in MM, compared with the limited clinical applications of this drug class outside the setting of plasma cell dyscrasias.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA.
| |
Collapse
|
8
|
Fernandes PMP, Guedes RA, Victor BL, Salvador JAR, Guedes RC. Decoding the secrets: how conformational and structural regulators inhibit the human 20S proteasome. Front Chem 2024; 11:1322628. [PMID: 38260042 PMCID: PMC10801056 DOI: 10.3389/fchem.2023.1322628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Acquired resistance to drugs that modulate specific protein functions, such as the human proteasome, presents a significant challenge in targeted therapies. This underscores the importance of devising new methodologies to predict drug binding and potential resistance due to specific protein mutations. In this work, we conducted an extensive computational analysis to ascertain the effects of selected mutations (Ala49Thr, Ala50Val, and Cys52Phe) within the active site of the human proteasome. Specifically, we sought to understand how these mutations might disrupt protein function either by altering protein stability or by impeding interactions with a clinical administered drug. Leveraging molecular dynamics simulations and molecular docking calculations, we assessed the effect of these mutations on protein stability and ligand affinity. Notably, our results indicate that the Cys52Phe mutation critically impacts protein-ligand binding, providing valuable insights into potential proteasome inhibitor resistance.
Collapse
Affiliation(s)
- Pedro M. P. Fernandes
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Romina A. Guedes
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Bruno L. Victor
- BioISI─Biosystems & Integrative Sciences Institute, Faculty of Sciences, Universidade de Lisboa, Lisboa, Portugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Rita C. Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
9
|
Jansen G, Al M, Assaraf YG, Kammerer S, van Meerloo J, Ossenkoppele GJ, Cloos J, Peters GJ. Statins markedly potentiate aminopeptidase inhibitor activity against (drug-resistant) human acute myeloid leukemia cells. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:430-446. [PMID: 37842233 PMCID: PMC10571057 DOI: 10.20517/cdr.2023.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/22/2023] [Accepted: 06/25/2023] [Indexed: 09/15/2023]
Abstract
Aim: This study aimed to decipher the molecular mechanism underlying the synergistic effect of inhibitors of the mevalonate-cholesterol pathway (i.e., statins) and aminopeptidase inhibitors (APis) on APi-sensitive and -resistant acute myeloid leukemia (AML) cells. Methods: U937 cells and their sublines with low and high levels of acquired resistance to (6S)-[(R)-2-((S)-Hydroxy-hydroxycarbamoyl-methoxy-methyl)-4-methyl-pentanoylamino]-3,3 dimethyl-butyric acid cyclopentyl ester (CHR2863), an APi prodrug, served as main AML cell line models. Drug combination effects were assessed with CHR2863 and in vitro non-toxic concentrations of various statins upon cell growth inhibition, cell cycle effects, and apoptosis induction. Mechanistic studies involved analysis of Rheb prenylation required for mTOR activation. Results: A strong synergy of CHR2863 with the statins simvastatin, fluvastatin, lovastatin, and pravastatin was demonstrated in U937 cells and two CHR2863-resistant sublines. This potent synergy between simvastatin and CHR2863 was also observed with a series of other human AML cell lines (e.g., THP1, MV4-11, and KG1), but not with acute lymphocytic leukemia or multiple solid tumor cell lines. This synergistic activity was: (i) specific for APis (e.g., CHR2863 and Bestatin), rather than for other cytotoxic agents; and (ii) corroborated by enhanced induction of apoptosis and cell cycle arrest which increased the sub-G1 fraction. Consistently, statin potentiation of CHR2863 activity was abrogated by co-administration of mevalonate and/or farnesyl pyrophosphate, suggesting the involvement of protein prenylation; this was experimentally confirmed by impaired Rheb prenylation by simvastatin. Conclusion: These novel findings suggest that the combined inhibitory effect of impaired Rheb prenylation and CHR2863-dependent mTOR inhibition instigates a potent synergistic inhibition of statins and APis on human AML cells.
Collapse
Affiliation(s)
- Gerrit Jansen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
| | - Marjon Al
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
| | - Yehuda G. Assaraf
- The Fred Wyszkowsky Cancer Research Laboratory, Faculty of Biology, The Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Sarah Kammerer
- Department of Medical Oncology, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
| | - Johan van Meerloo
- Department of Hematology, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
| | - Gert J. Ossenkoppele
- Department of Hematology, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
| | - Godefridus J. Peters
- Department of Medical Oncology, Amsterdam University Medical Center, location VUmc, Amsterdam 1081 HV, The Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdansk 80-210, Poland
| |
Collapse
|
10
|
Review on Bortezomib Resistance in Multiple Myeloma and Potential Role of Emerging Technologies. Pharmaceuticals (Basel) 2023; 16:ph16010111. [PMID: 36678608 PMCID: PMC9864669 DOI: 10.3390/ph16010111] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Multiple myeloma is a hematological cancer type. For its treatment, Bortezomib has been widely used. However, drug resistance to this effective chemotherapeutic has been developed for various reasons. 2D cell cultures and animal models have failed to understand the MM disease and Bortezomib resistance. It is therefore essential to utilize new technologies to reveal a complete molecular profile of the disease. In this review, we in-depth examined the possible molecular mechanisms that cause Bortezomib resistance and specifically addressed MM and Bortezomib resistance. Moreover, we also included the use of nanoparticles, 3D culture methods, microfluidics, and organ-on-chip devices in multiple myeloma. We also discussed whether the emerging technology offers the necessary tools to understand and prevent Bortezomib resistance in multiple myeloma. Despite the ongoing research activities on MM, the related studies cannot provide a complete summary of MM. Nanoparticle and 3D culturing have been frequently used to understand MM disease and Bortezomib resistance. However, the number of microfluidic devices for this application is insufficient. By combining siRNA/miRNA technologies with microfluidic devices, a complete molecular genetic profile of MM disease could be revealed. Microfluidic chips should be used clinically in personal therapy and point-of-care applications. At least with Bortezomib microneedles, it could be ensured that MM patients can go through the treatment process more painlessly. This way, MM can be switched to the curable cancer type list, and Bortezomib can be targeted for its treatment with fewer side effects.
Collapse
|
11
|
Haertle L, Barrio S, Munawar U, Han S, Zhou X, Simicek M, Vogt C, Truger M, Fernandez RA, Steinhardt M, Weingart J, Snaurova R, Nerreter S, Teufel E, Garitano-Trojaola A, Da Viá M, Ruiz-Heredia Y, Rosenwald A, Bolli N, Hajek R, Raab P, Raab MS, Weinhold N, Haferlach C, Haaf T, Martinez-Lopez J, Einsele H, Rasche L, Kortüm KM. Single-Nucleotide Variants and Epimutations Induce Proteasome Inhibitor Resistance in Multiple Myeloma. Clin Cancer Res 2023; 29:279-288. [PMID: 36282272 DOI: 10.1158/1078-0432.ccr-22-1161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/08/2022] [Accepted: 10/21/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Proteasome inhibitors (PI) are the backbone of various treatment regimens in multiple myeloma. We recently described the first in-patient point mutations affecting the 20S subunit PSMB5 underlying PI resistance. Notably, in vivo, the incidence of mutations in PSMB5 and other proteasome encoding genes is too low to explain the development of resistance in most of the affected patients. Thus, additional genetic and epigenetic alterations need to be explored. EXPERIMENTAL DESIGN We performed DNA methylation profiling by Deep Bisulfite Sequencing in PSMB5, PSMC2, PSMC5, PSMC6, PSMD1, and PSMD5, a subset of proteasome subunits that have hitherto been associated with PI resistance, recruited from our own previous research, the literature, or a meta-analysis on the frequency of somatic mutations. Methylation was followed up on gene expression level and by dual-luciferase reporter assay. The KMS11 cell line served as a model to functionally test the impact of demethylating agents. RESULTS We identified PSMD5 promoter hypermethylation and subsequent epigenetic gene silencing in 24% of PI refractory patients. Hypermethylation correlated with decreased expression and the regulatory impact of this region was functionally confirmed. In contrast, patients with newly diagnosed multiple myeloma, along with peripheral blood mononuclear cells and CD138+ plasma cells from healthy donors, generally show unmethylated profiles. CONCLUSIONS Under the selective pressure of PI treatment, multiple myeloma cells acquire methylation of the PSMD5 promoter silencing the PSMD5 gene expression. PSMD5 acts as a key orchestrator of proteasome assembly and its downregulation was described to increase the cell's proteolytic capacity. PSMD5 hypermethylation, therefore, represents a novel mechanism of PI tolerance in multiple myeloma.
Collapse
Affiliation(s)
- Larissa Haertle
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Department of Hematology, Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Complutense University Madrid, Madrid, Spain
| | - Santiago Barrio
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Department of Hematology, Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Complutense University Madrid, Madrid, Spain.,Altum Sequencing Co., Madrid, Spain
| | - Umair Munawar
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Seungbin Han
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Xiang Zhou
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Michal Simicek
- Haematology, Ostrava University Hospital, Ostrava, Czech Republic.,Faculty of Medicine, Ostrava University, Ostrava, Czech Republic
| | - Cornelia Vogt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Rafael Alonso Fernandez
- Department of Hematology, Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Complutense University Madrid, Madrid, Spain
| | | | - Julia Weingart
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Renata Snaurova
- Haematology, Ostrava University Hospital, Ostrava, Czech Republic.,Faculty of Medicine, Ostrava University, Ostrava, Czech Republic
| | - Silvia Nerreter
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Eva Teufel
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Matteo Da Viá
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Yanira Ruiz-Heredia
- Department of Hematology, Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Complutense University Madrid, Madrid, Spain.,Altum Sequencing Co., Madrid, Spain
| | | | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roman Hajek
- Haematology, Ostrava University Hospital, Ostrava, Czech Republic.,Faculty of Medicine, Ostrava University, Ostrava, Czech Republic
| | - Peter Raab
- Department of Orthopaedic Surgery, König Ludwig Haus, University of Würzburg, Würzburg, Germany
| | - Marc S Raab
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Molecular Hematology/Oncology, Department of Internal Medicine V, Heidelberg University Hospital, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Joaquin Martinez-Lopez
- Department of Hematology, Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Complutense University Madrid, Madrid, Spain.,Altum Sequencing Co., Madrid, Spain
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
Sellin M, Berg S, Hagen P, Zhang J. The molecular mechanism and challenge of targeting XPO1 in treatment of relapsed and refractory myeloma. Transl Oncol 2022; 22:101448. [PMID: 35660848 PMCID: PMC9166471 DOI: 10.1016/j.tranon.2022.101448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/14/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022] Open
Abstract
Significant progress has been made on the treatment of MM during past two decades. Acquired drug-resistance continues to drive early relapse in primary refractory MM. XPO1 over-expression and cargo mislocalization are associated with drug-resistance. XPO1 inhibitor selinexor restores drug sensitivity to subsets of RR-MM cells.
Multiple myeloma (MM) treatment regimens have vastly improved since the introduction of immunomodulators, proteasome inhibitors, and anti-CD38 monoclonal antibodies; however, MM is considered an incurable disease due to inevitable relapse and acquired drug resistance. Understanding the molecular mechanism by which drug resistance is acquired will help create novel strategies to prevent relapse and help develop novel therapeutics to treat relapsed/refractory (RR)-MM patients. Currently, only homozygous deletion/mutation of TP53 gene due to “double-hits” on Chromosome 17p region is consistently associated with a poor prognosis. The exciting discovery of XPO1 overexpression and mislocalization of its cargos in the RR-MM cells has led to a novel treatment options. Clinical studies have demonstrated that the XPO1 inhibitor selinexor can restore sensitivity of RR-MM to PIs and dexamethasone. We will elaborate on the problems of MM treatment strategies and discuss the mechanism and challenges of using XPO1 inhibitors in RR-MM therapies while deliberating potential solutions.
Collapse
Affiliation(s)
- Mark Sellin
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Loyola University Chicago, USA
| | - Stephanie Berg
- Loyola University Chicago, Department of Cancer Biology and Internal Medicine, Cardinal Bernardin Cancer Center, Stritch School of Medicine, Maywood, IL, USA.
| | - Patrick Hagen
- Department of Medicine, Division of Hematology/Oncology, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, USA
| |
Collapse
|
13
|
Revisiting Proteasome Inhibitors: Molecular Underpinnings of Their Development, Mechanisms of Resistance and Strategies to Overcome Anti-Cancer Drug Resistance. Molecules 2022; 27:molecules27072201. [PMID: 35408601 PMCID: PMC9000344 DOI: 10.3390/molecules27072201] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Proteasome inhibitors have shown relevant clinical activity in several hematological malignancies, namely in multiple myeloma and mantle cell lymphoma, improving patient outcomes such as survival and quality of life, when compared with other therapies. However, initial response to the therapy is a challenge as most patients show an innate resistance to proteasome inhibitors, and those that respond to the therapy usually develop late relapses suggesting the development of acquired resistance. The mechanisms of resistance to proteasome inhibition are still controversial and scarce in the literature. In this review, we discuss the development of proteasome inhibitors and the mechanisms of innate and acquired resistance to their activity—a major challenge in preclinical and clinical therapeutics. An improved understanding of these mechanisms is crucial to guiding the design of new and more effective drugs to tackle these devastating diseases. In addition, we provide a comprehensive overview of proteasome inhibitors used in combination with other chemotherapeutic agents, as this is a key strategy to combat resistance.
Collapse
|
14
|
Das S, Juliana N, Yazit NAA, Azmani S, Abu IF. Multiple Myeloma: Challenges Encountered and Future Options for Better Treatment. Int J Mol Sci 2022; 23:1649. [PMID: 35163567 PMCID: PMC8836148 DOI: 10.3390/ijms23031649] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is a malignant hematological disease. The disease is characterized by the clonal proliferation of malignant plasma cells in the bone marrow. MM accounts for 1.3% of all malignancies and has been increasing in incidence all over the world. Various genetic abnormalities, mutations, and translocation, including epigenetic modifications, are known to contribute to the disease's pathophysiology. The prognosis is good if detected early, or else the outcome is very bad if distant metastasis has already occurred. Conventional treatment with drugs poses a challenge when there is drug resistance. In the present review, we discuss multiple myeloma and its treatment, drug resistance, the molecular basis of epigenetic regulation, the role of natural products in epigenetic regulators, diet, physical activity, addiction, and environmental pollutants, which may be beneficial for clinicians and researchers.
Collapse
Affiliation(s)
- Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Al-Khoud, Muscat 123, Oman;
| | - Norsham Juliana
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Persiaran Ilmu, Putra Nilai, Nilai 71800, Negeri Sembilan, Malaysia; (N.A.A.Y.); (S.A.)
| | - Noor Anisah Abu Yazit
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Persiaran Ilmu, Putra Nilai, Nilai 71800, Negeri Sembilan, Malaysia; (N.A.A.Y.); (S.A.)
| | - Sahar Azmani
- Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, Persiaran Ilmu, Putra Nilai, Nilai 71800, Negeri Sembilan, Malaysia; (N.A.A.Y.); (S.A.)
| | - Izuddin Fahmy Abu
- Institute of Medical Science Technology, Universiti Kuala Lumpur, Kuala Lumpur 50250, Selangor, Malaysia;
| |
Collapse
|
15
|
Zaal EA, de Grooth HJ, Oudaert I, Langerhorst P, Levantovsky S, van Slobbe GJJ, Jansen JWA, Menu E, Wu W, Berkers CR. Targeting coenzyme Q10 synthesis overcomes bortezomib resistance in multiple myeloma. Mol Omics 2021; 18:19-30. [PMID: 34879122 DOI: 10.1039/d1mo00106j] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
During the development of drug resistance, multiple myeloma (MM) cells undergo changes to their metabolism. However, how these metabolic changes can be exploited to improve treatment efficacy is not known. Here we demonstrate that targeting coenzyme Q10 (CoQ) biosynthesis through the mevalonate pathway works in synergy with the proteasome inhibitor bortezomib (BTZ) in MM. We show that gene expression signatures relating to the mitochondrial tricarboxylic acid (TCA) cycle and electron transport chain (ETC) predispose to clinical BTZ resistance and poor prognosis in MM patients. Mechanistically, BTZ-resistant cells show increased activity of glutamine-driven TCA cycle and oxidative phosphorylation, together with an increased vulnerability towards ETC inhibition. Moreover, BTZ resistance is accompanied by high levels of the mitochondrial electron carrier CoQ, while the mevalonate pathway inhibitor simvastatin increases cell death and decreases CoQ levels, specifically in BTZ-resistant cells. Both in vitro and in vivo, simvastatin enhances the effect of bortezomib treatment. Our study links CoQ synthesis to drug resistance in MM and provides a novel avenue for improving BTZ responses through statin-induced inhibition of mitochondrial metabolism.
Collapse
Affiliation(s)
- Esther A Zaal
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Division of Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Harm-Jan de Grooth
- Department of Intensive Care & Department of Anesthesiology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Inge Oudaert
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pieter Langerhorst
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Sophie Levantovsky
- Division of Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Gijs J J van Slobbe
- Division of Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Jeroen W A Jansen
- Division of Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Celia R Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Division of Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
16
|
Lee MJ, Bhattarai D, Jang H, Baek A, Yeo IJ, Lee S, Miller Z, Lee S, Hong JT, Kim DE, Lee W, Kim KB. Macrocyclic Immunoproteasome Inhibitors as a Potential Therapy for Alzheimer's Disease. J Med Chem 2021; 64:10934-10950. [PMID: 34309393 PMCID: PMC10913540 DOI: 10.1021/acs.jmedchem.1c00291] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previously, we reported that immunoproteasome (iP)-targeting linear peptide epoxyketones improve cognitive function in mouse models of Alzheimer's disease (AD) in a manner independent of amyloid β. However, these compounds' clinical prospect for AD is limited due to potential issues, such as poor brain penetration and metabolic instability. Here, we report the development of iP-selective macrocyclic peptide epoxyketones prepared by a ring-closing metathesis reaction between two terminal alkenes attached at the P2 and P3/P4 positions of linear counterparts. We show that a lead macrocyclic compound DB-60 (20) effectively inhibits the catalytic activity of iP in ABCB1-overexpressing cells (IC50: 105 nM) and has metabolic stability superior to its linear counterpart. DB-60 (20) also lowered the serum levels of IL-1α and ameliorated cognitive deficits in Tg2576 mice. The results collectively suggest that macrocyclic peptide epoxyketones have improved CNS drug properties than their linear counterparts and offer promising potential as an AD drug candidate.
Collapse
Affiliation(s)
- Min Jae Lee
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| | - Deepak Bhattarai
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| | - Hyeryung Jang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ahreum Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Seongsoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Zachary Miller
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| | - Sukyeong Lee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| |
Collapse
|
17
|
Bo Kim K. Proteasomal adaptations to FDA-approved proteasome inhibitors: a potential mechanism for drug resistance? CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:634-645. [PMID: 34308274 PMCID: PMC8297691 DOI: 10.20517/cdr.2021.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With proteasome inhibitors (PIs) becoming clinically available since 2003, outcomes for patients with multiple myeloma (MM) have dramatically changed, improving quality of life and survival. Despite the impressive treatment success, however, almost all MM patients who initially respond to these PIs eventually develop resistance. Furthermore, a portion of MM patients is inherently unresponsive to the PIs. Extensive mechanistic investigations identified several non-proteasomal signaling pathways suspected to be linked to the PI resistance, for which several excellent reviews are currently available. On the other hand, it is still unclear how cancer cells under high PI environments adapt to spare proteasome activity essential for survival and proliferation regardless of cancer evolution stages. This review outlines current progress towards understanding the proteasomal adaptations of cells in response to PI treatment to maintain necessary proteasome activity. A better understanding of cellular proteasomal changes in response to the PIs could provide a rationale to develop new therapeutics that could be used to overcome resistance to existing PI drugs.
Collapse
Affiliation(s)
- Kyung Bo Kim
- Department of Pharmaceutics, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA
| |
Collapse
|
18
|
Mynott RL, Wallington-Beddoe CT. Drug and Solute Transporters in Mediating Resistance to Novel Therapeutics in Multiple Myeloma. ACS Pharmacol Transl Sci 2021; 4:1050-1065. [PMID: 34151200 DOI: 10.1021/acsptsci.1c00074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 02/06/2023]
Abstract
Multiple myeloma remains an incurable malignancy of plasma cells. Novel therapies, notably proteasome inhibitors and immunomodulatory drugs, have improved the survival of multiple myeloma patients; however, patients either present with, or develop resistance to, these therapies. Resistance to traditional chemotherapeutic agents can be caused by cellular drug efflux via adenosine triphosphate (ATP)-binding cassette (ABC) transporters, but it is still not clear whether these transporters mediate resistance to proteasome inhibitors and immunomodulatory drugs in multiple myeloma. Solute carrier (SLC) transporters also play a role in cancer drug resistance due to changes in cell homeostasis caused by their abnormal expression and changes in the solutes they transport. In this review, we evaluate resistance to novel therapies used to treat multiple myeloma, as mediated by drug and solute transporters.
Collapse
Affiliation(s)
- Rachel L Mynott
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Craig T Wallington-Beddoe
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia.,Flinders Medical Centre, Bedford Park, South Australia 5042, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, South Australia 5000, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
19
|
Roeten MS, van Meerloo J, Kwidama ZJ, ter Huizen G, Segerink WH, Zweegman S, Kaspers GJ, Jansen G, Cloos J. Pre-Clinical Evaluation of the Proteasome Inhibitor Ixazomib against Bortezomib-Resistant Leukemia Cells and Primary Acute Leukemia Cells. Cells 2021; 10:665. [PMID: 33802801 PMCID: PMC8002577 DOI: 10.3390/cells10030665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
At present, 20-30% of children with acute leukemia still relapse from current chemotherapy protocols, underscoring the unmet need for new treatment options, such as proteasome inhibition. Ixazomib (IXA) is an orally available proteasome inhibitor, with an improved safety profile compared to Bortezomib (BTZ). The mechanism of action (proteasome subunit inhibition, apoptosis induction) and growth inhibitory potential of IXA vs. BTZ were tested in vitro in human (BTZ-resistant) leukemia cell lines. Ex vivo activity of IXA vs. BTZ was analyzed in 15 acute lymphoblastic leukemia (ALL) and 9 acute myeloid leukemia (AML) primary pediatric patient samples. BTZ demonstrated more potent inhibitory effects on constitutive β5 and immunoproteasome β5i proteasome subunit activity; however, IXA more potently inhibited β1i subunit than BTZ (70% vs. 29% at 2.5 nM). In ALL/AML cell lines, IXA conveyed 50% growth inhibition at low nanomolar concentrations, but was ~10-fold less potent than BTZ. BTZ-resistant cells (150-160 fold) displayed similar (100-fold) cross-resistance to IXA. Finally, IXA and BTZ exhibited anti-leukemic effects for primary ex vivo ALL and AML cells; mean LC50 (nM) for IXA: 24 ± 11 and 30 ± 8, respectively, and mean LC50 for BTZ: 4.5 ± 1 and 11 ± 4, respectively. IXA has overlapping mechanisms of action with BTZ and showed anti-leukemic activity in primary leukemic cells, encouraging further pre-clinical in vivo evaluation.
Collapse
Affiliation(s)
- Margot S.F. Roeten
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Johan van Meerloo
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Zinia J. Kwidama
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Giovanna ter Huizen
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Wouter H. Segerink
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Sonja Zweegman
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Gertjan J.L. Kaspers
- Princess Maxima Center of Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1105 AZ Amsterdam, The Netherlands
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands;
| | - Jacqueline Cloos
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| |
Collapse
|
20
|
Robak P, Jarych D, Mikulski D, Dróżdż I, Węgłowska E, Kotkowska A, Misiewicz M, Smolewski P, Stawiski K, Fendler W, Szemraj J, Robak T. The Prognostic Value of Whole-Blood PSMB5, CXCR4, POMP, and RPL5 mRNA Expression in Patients with Multiple Myeloma Treated with Bortezomib. Cancers (Basel) 2021; 13:951. [PMID: 33668794 PMCID: PMC7956525 DOI: 10.3390/cancers13050951] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Proteasome inhibitors, like bortezomib, play a key role in the treatment of multiple myeloma (MM); however, most patients eventually relapse and eventually show multiple drug resistance, and the molecular mechanisms of this resistance remain unclear. The aim of our study is to assess the expression of previously described genes that may influence the resistance to bortezomib treatment at the mRNA level (ABCB1, CXCR4, MAF, MARCKS, POMP, PSMB5, RPL5, TXN, and XBP1) and prognosis of MM patients. mRNA expression was determined in 73 MM patients treated with bortezomib-based regimens (30 bortzomib-sensitive and 43 bortezomib-refractory patients) and 11 healthy controls. RPL5 was significantly down-regulated in multiple myeloma patients as compared with healthy controls. Moreover, POMP was significantly up-regulated in MM patients refractory to bortezomib-based treatment. In multivariate analysis, high expression of PSMB5 and CXCR and autologous stem cell transplantation were independent predictors of progression-free survival, and high expression of POMP and RPL5 was associated with shorter overall survival.
Collapse
Affiliation(s)
- Pawel Robak
- Department of Experimental Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (P.R.); (P.S.)
| | - Dariusz Jarych
- Laboratory of Personalized Medicine, Bionanopark, 93-465 Lodz, Poland; (D.J.); (E.W.)
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 93–232 Lodz, Poland
| | - Damian Mikulski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (D.M.); (K.S.); (W.F.)
| | - Izabela Dróżdż
- Department of Clinical Genetics, Medical University of Lodz, 92-213 Lodz, Poland;
| | - Edyta Węgłowska
- Laboratory of Personalized Medicine, Bionanopark, 93-465 Lodz, Poland; (D.J.); (E.W.)
| | - Aleksandra Kotkowska
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (A.K.); (M.M.)
| | - Małgorzata Misiewicz
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (A.K.); (M.M.)
| | - Piotr Smolewski
- Department of Experimental Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (P.R.); (P.S.)
| | - Konrad Stawiski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (D.M.); (K.S.); (W.F.)
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (D.M.); (K.S.); (W.F.)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (A.K.); (M.M.)
| |
Collapse
|
21
|
Mynott RL, Wallington-Beddoe CT. Inhibition of P-Glycoprotein Does Not Increase the Efficacy of Proteasome Inhibitors in Multiple Myeloma Cells. ACS Pharmacol Transl Sci 2021; 4:713-729. [PMID: 33860196 DOI: 10.1021/acsptsci.0c00200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Indexed: 12/14/2022]
Abstract
P-Glycoprotein is a well-known drug transporter associated with chemotherapy resistance in a number of cancers, but its role in modulating proteasome inhibitor efficacy in multiple myeloma is not well understood. The second-generation proteasome inhibitor carfilzomib is thought to be a substrate of P-glycoprotein whose efficacy may correlate with P-glycoprotein activity; however, research concerning the first-in-class proteasome inhibitor bortezomib is inconsistent. We show that while P-glycoprotein gene expression increases with the disease stages leading to multiple myeloma it does not affect the survival of newly diagnosed patients treated with bortezomib. Moreover, RNA-seq on LP-1 cells demonstrated minimal basal P-glycoprotein expression which did not increase after exposure to bortezomib or carfilzomib. Only one (KMS-18) of nine multiple myeloma cell lines expressed P-glycoprotein, including RPMI-8226 cells that are resistant to bortezomib or carfilzomib. We hypothesized that by inhibiting P-glycoprotein multiple myeloma cell sensitivity to proteasome inhibitors would increase; however, the sensitivity of multiple myeloma cells lines to proteasome inhibition was not enhanced by the specific P-glycoprotein inhibitor tariquidar. In addition, targeting glucosylceramide synthase with eliglustat did not inhibit P-glycoprotein activity nor improve proteasome inhibitor efficacy except at a high concentration. To confirm these negative findings, tariquidar did not substantially increase the cytotoxicity of bortezomib or carfilzomib in P-glycoprotein-expressing K562/ADM cells. We conclude the following: P-glycoprotein expression may not correlate with the survival of newly diagnosed multiple myeloma patients treated with proteasome inhibitors. P-glycoprotein is poorly expressed in many multiple myeloma cell lines, and its inhibition does not appreciably enhance the efficacy of proteasome inhibitors.
Collapse
Affiliation(s)
- Rachel L Mynott
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Craig T Wallington-Beddoe
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia.,Flinders Medical Centre, Bedford Park, South Australia 5042, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, South Australia 5000, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
22
|
Agbana P, Lee MJ, Rychahou P, Kim KB, Bae Y. Ternary Polypeptide Nanoparticles with Improved Encapsulation, Sustained Release, and Enhanced In Vitro Efficacy of Carfilzomib. Pharm Res 2020; 37:213. [PMID: 33025286 DOI: 10.1007/s11095-020-02922-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/01/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To develop a new nanoparticle formulation for a proteasome inhibitor Carfilzomib (CFZ) to improve its stability and efficacy for future in vivo applications. METHODS CFZ-loaded ternary polypeptide nanoparticles (CFZ/tPNPs) were prepared by using heptakis(6-amino-6-deoxy)-β-cyclodextrin(hepta-hydrochloride) (HaβCD) and azido-poly(ethylene glycol)-block-poly(L-glutamic acid sodium salt) (N3-PEG-PLE). The process involved ternary (hydrophobic/ionic/supramolecular) interactions in three steps: 1) CFZ was entrapped in the cavity of HaβCD by hydrophobic interaction, 2) the drug-cyclodextrin inclusion complexes were mixed with N3-PEG-PLE to form polyion complex nanoparticles, and 3) the nanoparticles were modified with fluorescent dyes (AFDye 647) for imaging and/or epithelial cell adhesion molecule (EpCAM) antibodies for cancer cell targeting. CFZ/tPNPs were characterized for particle size, surface charge, drug release, stability, intracellular uptake, proteasome inhibition, and in vitro cytotoxicity. RESULTS tPNPs maintained an average particle size of 50 nm after CFZ entrapment, EpCAM conjugation, and freeze drying. tPNPs achieved high aqueous solubility of CFZ (>1 mg/mL), sustained drug release (t1/2 = 6.46 h), and EpCAM-mediated cell targeting, which resulted in increased intracellular drug accumulation, prolonged proteasome inhibition, and enhanced cytotoxicity of CFZ in drug-resistant DLD-1 colorectal cancer cells. CONCLUSIONS tPNPs improved stability and efficacy of CFZ in vitro, and these results potentiate effective cancer treatment using CFZ/tPNPs in future vivo studies.
Collapse
Affiliation(s)
- Preye Agbana
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, Kentucky, 40536-0596, USA
| | - Min Jae Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, Kentucky, 40536-0596, USA
| | - Piotr Rychahou
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, 40536, USA
| | - Kyung-Bo Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, Kentucky, 40536-0596, USA
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone, Lexington, Kentucky, 40536-0596, USA.
| |
Collapse
|
23
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
24
|
Hashimoto E, Okuno S, Hirayama S, Arata Y, Goto T, Kosako H, Hamazaki J, Murata S. Enhanced O-GlcNAcylation Mediates Cytoprotection under Proteasome Impairment by Promoting Proteasome Turnover in Cancer Cells. iScience 2020; 23:101299. [PMID: 32634741 PMCID: PMC7338785 DOI: 10.1016/j.isci.2020.101299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 01/27/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
The proteasome is a therapeutic target in cancer, but resistance to proteasome inhibitors often develops owing to the induction of compensatory pathways. Through a genome-wide siRNA screen combined with RNA sequencing analysis, we identified hexokinase and downstream O-GlcNAcylation as cell survival factors under proteasome impairment. The inhibition of O-GlcNAcylation synergistically induced massive cell death in combination with proteasome inhibition. We further demonstrated that O-GlcNAcylation was indispensable for maintaining proteasome activity by enhancing biogenesis as well as proteasome degradation in a manner independent of Nrf1, a well-known compensatory transcription factor that upregulates proteasome gene expression. Our results identify a pathway that maintains proteasome function under proteasome impairment, providing potential targets for cancer therapy. O-GlcNAcylation suppresses cell death under proteasome impairment Combined inhibition of O-GlcNAcylation and proteasome induces massive tumor cell death O-GlcNAcylation maintains proteasome activity independently of Nrf1 O-GlcNAcylation enhances proteasome turnover under the proteasome impairment
Collapse
Affiliation(s)
- Eiichi Hashimoto
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shota Okuno
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shoshiro Hirayama
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Yoshiyuki Arata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Tsuyoshi Goto
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Kuramoto-cho, Tokushima 7708503, Japan
| | - Jun Hamazaki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan.
| |
Collapse
|
25
|
Pinto V, Bergantim R, Caires HR, Seca H, Guimarães JE, Vasconcelos MH. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers (Basel) 2020; 12:E407. [PMID: 32050631 PMCID: PMC7072128 DOI: 10.3390/cancers12020407] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is the second most common blood cancer. Treatments for MM include corticosteroids, alkylating agents, anthracyclines, proteasome inhibitors, immunomodulatory drugs, histone deacetylase inhibitors and monoclonal antibodies. Survival outcomes have improved substantially due to the introduction of many of these drugs allied with their rational use. Nonetheless, MM patients successively relapse after one or more treatment regimens or become refractory, mostly due to drug resistance. This review focuses on the main drugs used in MM treatment and on causes of drug resistance, including cytogenetic, genetic and epigenetic alterations, abnormal drug transport and metabolism, dysregulation of apoptosis, autophagy activation and other intracellular signaling pathways, the presence of cancer stem cells, and the tumor microenvironment. Furthermore, we highlight the areas that need to be further clarified in an attempt to identify novel therapeutic targets to counteract drug resistance in MM patients.
Collapse
Affiliation(s)
- Vanessa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- FCTUC–Faculty of Science and Technology of the University of Coimbra, 3030-790 Coimbra, Portugal
| | - Rui Bergantim
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hugo R. Caires
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Hugo Seca
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - M. Helena Vasconcelos
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP-Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
26
|
Targeting the ubiquitin-proteasome pathway to overcome anti-cancer drug resistance. Drug Resist Updat 2020; 48:100663. [DOI: 10.1016/j.drup.2019.100663] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/01/2019] [Accepted: 11/03/2019] [Indexed: 02/07/2023]
|
27
|
Mogollón P, Díaz-Tejedor A, Algarín EM, Paíno T, Garayoa M, Ocio EM. Biological Background of Resistance to Current Standards of Care in Multiple Myeloma. Cells 2019; 8:cells8111432. [PMID: 31766279 PMCID: PMC6912619 DOI: 10.3390/cells8111432] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/03/2019] [Accepted: 11/09/2019] [Indexed: 12/16/2022] Open
Abstract
A high priority problem in multiple myeloma (MM) management is the development of resistance to administered therapies, with most myeloma patients facing successively shorter periods of response and relapse. Herewith, we review the current knowledge on the mechanisms of resistance to the standard backbones in MM treatment: proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies (mAbs). In some cases, strategies to overcome resistance have been discerned, and an effort should be made to evaluate whether resensitization to these agents is feasible in the clinical setting. Additionally, at a time in which we are moving towards precision medicine in MM, it is equally important to identify reliable and accurate biomarkers of sensitivity/refractoriness to these main therapeutic agents with the goal of having more efficacious treatments and, if possible, prevent the development of relapse.
Collapse
Affiliation(s)
- Pedro Mogollón
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Andrea Díaz-Tejedor
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Esperanza M. Algarín
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Teresa Paíno
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Mercedes Garayoa
- Hospital Universitario de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), 37007 Salamanca, Spain; (P.M.); (A.D.-T.); (E.M.A.); (T.P.); (M.G.)
| | - Enrique M. Ocio
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39008 Santander, Spain
- Correspondence: ; Tel.: +34-942202520
| |
Collapse
|
28
|
Mofers A, Perego P, Selvaraju K, Gatti L, Gullbo J, Linder S, D'Arcy P. Analysis of determinants for in vitro resistance to the small molecule deubiquitinase inhibitor b-AP15. PLoS One 2019; 14:e0223807. [PMID: 31639138 PMCID: PMC6804958 DOI: 10.1371/journal.pone.0223807] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/27/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND b-AP15/VLX1570 are small molecule inhibitors of the ubiquitin specific peptidase 14 (USP14) and ubiquitin carboxyl-terminal hydrolase 5 (UCHL5) deubiquitinases (DUBs) of the 19S proteasome. b-AP15/VLX1570 have been shown to be cytotoxic to cells resistant to bortezomib, raising the possibility that this class of drugs can be used as a second-line therapy for treatment-resistant multiple myeloma. Limited information is available with regard to potential resistance mechanisms to b-AP15/VLX1570. RESULTS We found that b-AP15-induced cell death is cell-cycle dependent and that non-cycling tumor cells may evade b-AP15-induced cell death. Such non-cycling cells may re-enter the proliferative state to form colonies of drug-sensitive cells. Long-term selection of cells with b-AP15 resulted in limited drug resistance (~2-fold) that could be reversed by buthionine sulphoximine, implying altered glutathione (GSH) metabolism as a resistance mechanism. In contrast, drug uptake and overexpression of drug efflux transporters were found not to be associated with b-AP15 resistance. CONCLUSIONS The proteasome DUB inhibitors b-AP15/VLX1570 are cell cycle-active. The slow and incomplete development of resistance towards these compounds is an attractive feature in view of future clinical use.
Collapse
Affiliation(s)
- Arjan Mofers
- Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - Paola Perego
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Karthik Selvaraju
- Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - Laura Gatti
- Cerebrovascular Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Joachim Gullbo
- Department of Radiology, Oncology and Radiation Science, Section of Oncology, Uppsala University, Uppsala, Sweden
| | - Stig Linder
- Department of Medicine and Health, Linköping University, Linköping, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Padraig D'Arcy
- Department of Medicine and Health, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
29
|
Sarmento-Ribeiro AB, Scorilas A, Gonçalves AC, Efferth T, Trougakos IP. The emergence of drug resistance to targeted cancer therapies: Clinical evidence. Drug Resist Updat 2019; 47:100646. [PMID: 31733611 DOI: 10.1016/j.drup.2019.100646] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022]
Abstract
For many decades classical anti-tumor therapies included chemotherapy, radiation and surgery; however, in the last two decades, following the identification of the genomic drivers and main hallmarks of cancer, the introduction of therapies that target specific tumor-promoting oncogenic or non-oncogenic pathways, has revolutionized cancer therapeutics. Despite the significant progress in cancer therapy, clinical oncologists are often facing the primary impediment of anticancer drug resistance, as many cancer patients display either intrinsic chemoresistance from the very beginning of the therapy or after initial responses and upon repeated drug treatment cycles, acquired drug resistance develops and thus relapse emerges, resulting in increased mortality. Our attempts to understand the molecular basis underlying these drug resistance phenotypes in pre-clinical models and patient specimens revealed the extreme plasticity and adaptive pathways employed by tumor cells, being under sustained stress and extensive genomic/proteomic instability due to the applied therapeutic regimens. Subsequent efforts have yielded more effective inhibitors and combinatorial approaches (e.g. the use of specific pharmacologic inhibitors with immunotherapy) that exhibit synergistic effects against tumor cells, hence enhancing therapeutic indices. Furthermore, new advanced methodologies that allow for the early detection of genetic/epigenetic alterations that lead to drug chemoresistance and prospective validation of biomarkers which identify patients that will benefit from certain drug classes, have started to improve the clinical outcome. This review discusses emerging principles of drug resistance to cancer therapies targeting a wide array of oncogenic kinases, along with hedgehog pathway and the proteasome and apoptotic inducers, as well as epigenetic and metabolic modulators. We further discuss mechanisms of resistance to monoclonal antibodies, immunomodulators and immune checkpoint inhibitors, potential biomarkers of drug response/drug resistance, along with possible new therapeutic avenues for the clinicians to combat devastating drug resistant malignancies. It is foreseen that these topics will be major areas of focused multidisciplinary translational research in the years to come.
Collapse
Affiliation(s)
- Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Department, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
30
|
What sustains the multidrug resistance phenotype beyond ABC efflux transporters? Looking beyond the tip of the iceberg. Drug Resist Updat 2019; 46:100643. [PMID: 31493711 DOI: 10.1016/j.drup.2019.100643] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022]
Abstract
Identification of multidrug (MDR) efflux transporters that belong to the ATP-Binding Cassette (ABC) superfamily, represented an important breakthrough for understanding cancer multidrug resistance (MDR) and its possible overcoming. However, recent data indicate that drug resistant cells have a complex intracellular physiology that involves constant changes in energetic and oxidative-reductive metabolic pathways, as well as in the molecular circuitries connecting mitochondria, endoplasmic reticulum (ER) and lysosomes. The aim of this review is to discuss the key molecular mechanisms of cellular reprogramming that induce and maintain MDR, beyond the presence of MDR efflux transporters. We specifically highlight how cancer cells characterized by high metabolic plasticity - i.e. cells able to shift the energy metabolism between glycolysis and oxidative phosphorylation, to survive both the normoxic and hypoxic conditions, to modify the cytosolic and mitochondrial oxidative-reductive metabolism, are more prone to adapt to exogenous stressors such as anti-cancer drugs and acquire a MDR phenotype. Similarly, we discuss how changes in mitochondria dynamics and mitophagy rates, changes in proteome stability ensuring non-oncogenic proteostatic mechanisms, changes in ubiquitin/proteasome- and autophagy/lysosome-related pathways, promote the cellular survival under stress conditions, along with the acquisition or maintenance of MDR. After dissecting the complex intracellular crosstalk that takes place during the development of MDR, we suggest that mapping the specific adaptation pathways underlying cell survival in response to stress and targeting these pathways with potent pharmacologic agents may be a new approach to enhance therapeutic efficacy against MDR tumors.
Collapse
|
31
|
Abstract
Bortezomib is a dipeptidyl boronic acid that selectively inhibits the ubiquitin proteasome pathway, which plays a role in the degradation of many intracellular proteins. It is the first-in-class selective and reversible inhibitor of the 26S proteasome, with antiproliferative and antitumor activity. It exerts its anti-neoplastic action mainly via the inhibition of the nuclear factor-κB pathway components associated with cell proliferation, apoptosis, and angiogenesis. The drug has revolutionized the treatment of multiple myeloma and, more recently, mantle cell lymphoma. In 2003, bortezomib received accelerated approval from the US Food and Drug Administration for the treatment of relapsed/refractory multiple myeloma and in 2008 for patients with previously untreated multiple myeloma. In 2006, bortezomib was approved for the treatment of refractory/relapsed mantle cell lymphoma and, in 2014, for previously untreated mantle cell lymphoma. Bortezomib has also demonstrated clinical efficacy both as a single drug and in combination with other agents in light chain amyloidosis, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, and peripheral T-cell lymphomas. Furthermore, continued clinical studies are required to confirm its value for patients with indolent and aggressive B-cell non-Hodgkin lymphomas and acute leukemias.
Collapse
Affiliation(s)
- Pawel Robak
- Department of Experimental Hematology, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Ul. Ciolkowskiego 2, 93-510, Lodz, Poland.
| |
Collapse
|
32
|
Hu Y, Chen W, Wang J. Progress in the identification of gene mutations involved in multiple myeloma. Onco Targets Ther 2019; 12:4075-4080. [PMID: 31213829 PMCID: PMC6538831 DOI: 10.2147/ott.s205922] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
Sequencing studies have been used to determine a spectrum of multiple myeloma (MM) mutations. Mutation of certain genes, including KRAS, NRAS, TP53, FAM46C, DIS3 and BRAF, have a high recurrence rate and may play important roles in the pathogenesis, progression and prognosis of MM. Mutations in DIS3, which encodes a highly conserved RNA exonuclease, lead to loss of function. The expression of FAM46C is highly correlated with the expression of ribosomal protein, but the exact function of FAM46C mutation is unclear. There are mutants of IRF4, which is considered an MM survival factor. Mutations in the gene coding for the DNA damage-binding protein (DDB1) may affect interactions with CUL4A, which is part of the cereblon (CRBN) ubiquitin ligase complex. IRF4is part of the complex, which binds to DNA. These findings might explain the resistance to immunomodulatory. TP53 deletion or mutation is often present in B-cell malignancies and is associated with low response rates. Myeloma pathogenic mutations in ATM have been found in adult lymphatic tumors. XBP1 and PSMB5 mutations may be related to bortezomib resistance. Multiple gene mutations (KRAS, NRAS and BRAF) involved in the same pathway were found a single patient. Identification of driver gene mutations has brought great hope to the field of individualized, targeted medicine for MM.
Collapse
Affiliation(s)
- Ying Hu
- Department of Hematology, Aerospace Central Hospital of Peking University, Beijing, People's Republic of China
| | - Wenming Chen
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jingbo Wang
- Department of Hematology, Aerospace Central Hospital of Peking University, Beijing, People's Republic of China
| |
Collapse
|
33
|
Lee MJ, Bhattarai D, Yoo J, Miller Z, Park JE, Lee S, Lee W, Driscoll JJ, Kim KB. Development of Novel Epoxyketone-Based Proteasome Inhibitors as a Strategy To Overcome Cancer Resistance to Carfilzomib and Bortezomib. J Med Chem 2019; 62:4444-4455. [PMID: 30964987 DOI: 10.1021/acs.jmedchem.8b01943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the past 15 years, proteasome inhibitors (PIs), namely bortezomib, carfilzomib (Cfz) and ixazomib, have significantly improved the overall survival and quality-of-life for multiple myeloma (MM) patients. However, a significant portion of MM patients do not respond to PI therapies. Drug resistance is present either de novo or acquired after prolonged therapy through mechanisms that remain poorly defined. The lack of a clear understanding of clinical PI resistance has hampered the development of next-generation PI drugs to treat MM patients who no longer respond to currently available therapies. Here, we designed and synthesized novel epoxyketone-based PIs by structural modifications at the P1' site. We show that a Cfz analog, 9, harboring a hydroxyl substituent at its P1' position was highly cytotoxic against cancer cell lines displaying de novo or acquired resistance to Cfz. These results suggest that peptide epoxyketones incorporating P1'-targeting moieties may have the potential to bypass resistance mechanisms associated with Cfz and to provide additional clinical options for patients resistant to Cfz.
Collapse
Affiliation(s)
- Min Jae Lee
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Deepak Bhattarai
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Jisu Yoo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences , Seoul National University , Seoul 08826 , Korea
| | - Zach Miller
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences , Seoul National University , Seoul 08826 , Korea
| | - Sukyeong Lee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology , Baylor College of Medicine , Houston , Texas 77030 , United States
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences , Seoul National University , Seoul 08826 , Korea
| | - James J Driscoll
- Department of Internal Medicine , Division of Hematology and Oncology and University of Cincinnati Cancer Institute , Cincinnati , Ohio 45267 , United States
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| |
Collapse
|
34
|
Park JE, Park J, Jun Y, Oh Y, Ryoo G, Jeong YS, Gadalla HH, Min JS, Jo JH, Song MG, Kang KW, Bae SK, Yeo Y, Lee W. Expanding therapeutic utility of carfilzomib for breast cancer therapy by novel albumin-coated nanocrystal formulation. J Control Release 2019; 302:148-159. [PMID: 30954620 DOI: 10.1016/j.jconrel.2019.04.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023]
Abstract
Carfilzomib (CFZ) is the second-in-class proteasome inhibitor with much improved efficacy and safety profiles over bortezomib in multiple myeloma patients. In expanding the utility of CFZ to solid cancer therapy, the poor aqueous solubility and in vivo instability of CFZ are considered major drawbacks. We investigated whether a nanocrystal (NC) formulation can address these issues and enhance anticancer efficacy of CFZ against breast cancer. The surface of NC was coated with albumin in order to enhance the formulation stability and drug delivery to tumors via interactions with albumin-binding proteins located in and near cancer cells. The novel albumin-coated NC formulation of CFZ (CFZ-alb NC) displayed improved metabolic stability and enhanced cellular interactions, uptake and cytotoxic effects in breast cancer cells in vitro. Consistently, CFZ-alb NC showed greater anticancer efficacy in a murine 4T1 orthotopic breast cancer model than the currently used cyclodextrin-based formulation. Overall, our results demonstrate the potential of CFZ-alb NC as a viable formulation for breast cancer therapy.
Collapse
Affiliation(s)
- Ji Eun Park
- College of Pharmacy and Research, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Joonyoung Park
- Department of Industrial and Physical Pharmacy, Purdue University, Heine Pharmacy Bldg, 575 W Stadium Ave, West Lafayette, IN 47907, USA
| | - Yearin Jun
- College of Pharmacy and Research, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Yunseok Oh
- College of Pharmacy and Research, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Gongmi Ryoo
- College of Pharmacy and Research, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Yoo-Seong Jeong
- College of Pharmacy and Research, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Hytham H Gadalla
- Department of Industrial and Physical Pharmacy, Purdue University, Heine Pharmacy Bldg, 575 W Stadium Ave, West Lafayette, IN 47907, USA
| | - Jee Sun Min
- College of Pharmacy and Integrated Research, Institute of Pharmaceutical Sciences, Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 42083, South Korea
| | - Jung Hwan Jo
- Department of Nuclear Medicine & Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea
| | - Myung Geun Song
- Biomedical Research Institute, Seoul National University Hospital & Department of Nuclear Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine & Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea
| | - Soo Kyung Bae
- College of Pharmacy and Integrated Research, Institute of Pharmaceutical Sciences, Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 42083, South Korea
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, Heine Pharmacy Bldg, 575 W Stadium Ave, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN 47907, USA
| | - Wooin Lee
- College of Pharmacy and Research, Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea.
| |
Collapse
|
35
|
Lee MJ, Miller Z, Park JE, Bhattarai D, Lee W, Kim KB. H727 cells are inherently resistant to the proteasome inhibitor carfilzomib, yet require proteasome activity for cell survival and growth. Sci Rep 2019; 9:4089. [PMID: 30858500 PMCID: PMC6411724 DOI: 10.1038/s41598-019-40635-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/20/2019] [Indexed: 01/06/2023] Open
Abstract
The second-in-class proteasome inhibitor (PI) carfilzomib (Kyprolis, Cfz) has contributed to a substantial advancement in multiple myeloma treatment by improving patient survival and quality of life. A considerable portion of patients however display intrinsic resistance to Cfz. Our mechanistic understanding of intrinsic Cfz resistance is limited due to a lack of suitable cell-based models. We report that H727 human bronchial carcinoid cells are inherently resistant to Cfz, yet susceptible to other PIs and inhibitors targeting upstream components of the ubiquitin-proteasome system (UPS). These results indicate that H727 cells remain dependent on the UPS for cell survival and growth despite harboring intrinsic resistance to Cfz. Alterations in the composition of proteasome catalytic subunits via interferon-γ treatment or siRNA knockdown results in sensitization of H727 cells to Cfz. We postulate that a potential link may exist between the composition of proteasome catalytic subunits and the cellular response to Cfz. Overall, H727 cells may serve as a useful cell-based model for de novo Cfz resistance and our results suggest previously unexplored mechanisms of de novo PI resistance.
Collapse
Affiliation(s)
- Min Jae Lee
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Zachary Miller
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Deepak Bhattarai
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
36
|
Covalent docking modelling-based discovery of tripeptidyl epoxyketone proteasome inhibitors composed of aliphatic-heterocycles. Eur J Med Chem 2019; 164:602-614. [DOI: 10.1016/j.ejmech.2018.12.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 02/08/2023]
|
37
|
Robak P, Drozdz I, Szemraj J, Robak T. Drug resistance in multiple myeloma. Cancer Treat Rev 2018; 70:199-208. [DOI: 10.1016/j.ctrv.2018.09.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/05/2018] [Accepted: 09/01/2018] [Indexed: 02/07/2023]
|
38
|
Oerlemans R, Berkers CR, Assaraf YG, Scheffer GL, Peters GJ, Verbrugge SE, Cloos J, Slootstra J, Meloen RH, Shoemaker RH, Dijkmans BAC, Scheper RJ, Ovaa H, Jansen G. Proteasome inhibition and mechanism of resistance to a synthetic, library-based hexapeptide. Invest New Drugs 2018; 36:797-809. [PMID: 29442210 PMCID: PMC6153520 DOI: 10.1007/s10637-018-0569-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/31/2018] [Indexed: 12/14/2022]
Abstract
Background The hexapeptide 4A6 (Ac-Thr(tBu)-His(Bzl)-Thr(Bzl)-Nle-Glu(OtBu)-Gly-Bza) was isolated from a peptide library constructed to identify peptide-based transport inhibitors of multidrug resistance (MDR) efflux pumps including P-glycoprotein and Multidrug Resistance-associated Protein 1. 4A6 proved to be a substrate but not an inhibitor of these MDR efflux transporters. In fact, 4A6 and related peptides displayed potent cytotoxic activity via an unknown mechanism. Objective To decipher the mode of cytotoxic activity of 4A6. Methods Screening of 4A6 activity was performed against the NCI60 panel of cancer cell lines. Possible interactions of 4A6 with the 26S proteasome were assessed via proteasome activity and affinity labeling, and cell growth inhibition studies with leukemic cells resistant to the proteasome inhibitor bortezomib (BTZ). Results The NCI60 panel COMPARE analysis revealed that 4A6 had an activity profile overlapping with BTZ. Consistently, 4A6 proved to be a selective and reversible inhibitor of β5 subunit (PSMB5)-associated chymotrypsin-like activity of the 26S proteasome. This conclusion is supported by several lines of evidence: (i) inhibition of chymotrypsin-like proteasome activity by 4A6 and related peptides correlated with their cell growth inhibition potencies; (ii) 4A6 reversibly inhibited functional β5 active site labeling with the affinity probe BodipyFL-Ahx3L3VS; and (iii) human myeloid THP1 cells with acquired BTZ resistance due to mutated PSMB5 were highly (up to 287-fold) cross-resistant to 4A6 and its related peptides. Conclusion 4A6 is a novel specific inhibitor of the β5 subunit-associated chymotrypsin-like proteasome activity. Further exploration of 4A6 as a lead compound for development as a novel proteasome-targeted drug is warranted.
Collapse
Affiliation(s)
- Ruud Oerlemans
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Celia R Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - George L Scheffer
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sue Ellen Verbrugge
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | | | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ben A C Dijkmans
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Rik J Scheper
- Department of Pathology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Huib Ovaa
- Division of Cell Biology II, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Chemical Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerrit Jansen
- Departments of Rheumatology, Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Rm 2.46, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Park JE, Miller Z, Jun Y, Lee W, Kim KB. Next-generation proteasome inhibitors for cancer therapy. Transl Res 2018; 198:1-16. [PMID: 29654740 PMCID: PMC6151281 DOI: 10.1016/j.trsl.2018.03.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/16/2018] [Accepted: 03/18/2018] [Indexed: 02/06/2023]
Abstract
Over 2 decades ago, the proteasome was considered a risky or even untenable therapeutic target. Today, proteasome inhibitors are a mainstay in the treatment of multiple myeloma (MM) and have sales in excess of 3 billion US dollars annually. More importantly, the availability of proteasome inhibitors has greatly improved the survival and quality of life for patients with MM. Despite the remarkable success of proteasome inhibitor therapies to date, the potential for improvement remains, and the development and optimal use of proteasome inhibitors as anticancer agents continues to be an active area of research. In this review, we briefly discuss the features and limitations of the 3 proteasome inhibitor drugs currently used in the clinic and provide an update on current efforts to develop next-generation proteasome inhibitors with the potential to overcome the limitations of existing proteasome inhibitor drugs.
Collapse
Affiliation(s)
- Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Zachary Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Yearin Jun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
40
|
Barrio S, Stühmer T, Da-Viá M, Barrio-Garcia C, Lehners N, Besse A, Cuenca I, Garitano-Trojaola A, Fink S, Leich E, Chatterjee M, Driessen C, Martinez-Lopez J, Rosenwald A, Beckmann R, Bargou RC, Braggio E, Stewart AK, Raab MS, Einsele H, Kortüm KM. Spectrum and functional validation of PSMB5 mutations in multiple myeloma. Leukemia 2018; 33:447-456. [PMID: 30026573 DOI: 10.1038/s41375-018-0216-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Despite an increasing number of approved therapies, multiple myeloma (MM) remains an incurable disease and only a small number of patients achieve prolonged disease control. Some genes have been linked with response to commonly used anti-MM compounds, including immunomodulators (IMiDs) and proteasome inhibitors (PIs). In this manuscript, we demonstrate an increased incidence of acquired proteasomal subunit mutations in relapsed MM compared to newly diagnosed disease, underpinning a potential role of point mutations in the clonal evolution of MM. Furthermore, we are first to present and functionally characterize four somatic PSMB5 mutations from primary MM cells identified in a patient under prolonged proteasome inhibition, with three of them affecting the PI-binding pocket S1. We confirm resistance induction through missense mutations not only to Bortezomib, but also, in variable extent, to the next-generation PIs Carfilzomib and Ixazomib. In addition, a negative impact on the proteasome activity is assessed, providing a potential explanation for later therapy-induced eradication of the affected tumor subclones in this patient.
Collapse
Affiliation(s)
- Santiago Barrio
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Stühmer
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Matteo Da-Viá
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Nicola Lehners
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrej Besse
- Departement of Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Isabel Cuenca
- Department of Hematology, Hospital Universitario 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Andoni Garitano-Trojaola
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Severin Fink
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ellen Leich
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Manik Chatterjee
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Driessen
- Departement of Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Joaquin Martinez-Lopez
- Department of Hematology, Hospital Universitario 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | | | - Ralf C Bargou
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Esteban Braggio
- Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - A Keith Stewart
- Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Marc S Raab
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hermann Einsele
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - K Martin Kortüm
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
41
|
Hanke NT, Imler E, Marron MT, Seligmann BE, Garland LL, Baker AF. Characterization of carfilzomib-resistant non-small cell lung cancer cell lines. J Cancer Res Clin Oncol 2018; 144:1317-1327. [PMID: 29766327 DOI: 10.1007/s00432-018-2662-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/08/2018] [Indexed: 01/12/2023]
Abstract
PURPOSE We previously showed that carfilzomib (CFZ) has potent anti-proliferative and cytotoxic activity in a broad range of lung cancer cell lines. Here we investigate possible mechanisms of CFZ acquired resistance in lung cancer cell lines. METHODS CFZ-resistant non-small cell lung cancer (NSCLC) cell lines were developed by exposing A549 and H520 cells to stepwise increasing concentrations of CFZ. Resistance to CFZ and cross-resistance to bortezomib and other chemotherapy drugs was measured using the MTT assay. Cytotoxicity to CFZ was determined using a CytoTox assay. Western blot was used to measure apoptosis, autophagy, and drug efflux transporter-related proteins. Quantitative targeted whole transcriptome sequencing and quantitative RT-PCR was used to measure gene expression. Flow cytometry was used to analyze intracellular accumulation of doxorubicin. RESULTS The CFZ IC50 value of the resistant cells increased versus parental lines (2.5-fold for A549, 122-fold for H520). Resistant lines showed reduced expression of apoptosis and autophagy markers and reduced death versus parental lines following CFZ treatment. Both resistant lines exhibited higher P-glycoprotein (Pgp) gene (TempO-Seq® analysis, increased 1.2-fold in A549, > 9000-fold in H520) and protein expression levels versus parental lines. TempO-Seq® analysis indicated other drug resistance pathways were upregulated. The resistant cell lines demonstrated less accumulation of intracellular doxorubicin, and were cross-resistant to other Pgp client drugs: bortezomib, doxorubicin, and paclitaxel, but not cisplatin. CONCLUSIONS Upregulation of Pgp appears to be an important, but not the only, mechanism of CFZ resistance in NSCLC cell lines.
Collapse
Affiliation(s)
- Neale T Hanke
- College of Medicine, University of Arizona Cancer Center, Tucson, AZ, USA
| | | | | | | | - Linda L Garland
- College of Medicine, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Amanda F Baker
- College of Medicine, University of Arizona Cancer Center, Tucson, AZ, USA.
- Ventana Medical Systems, Inc., A Member of the Roche Group, 1910 East Innovation Park Drive, Tucson, AZ, 85755, USA.
| |
Collapse
|
42
|
Wallington-Beddoe CT, Sobieraj-Teague M, Kuss BJ, Pitson SM. Resistance to proteasome inhibitors and other targeted therapies in myeloma. Br J Haematol 2018; 182:11-28. [PMID: 29676460 DOI: 10.1111/bjh.15210] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The number of novel therapies for the treatment of myeloma is rapidly increasing, as are the clinical trials evaluating them in combination with other novel and established therapies. Proteasome inhibitors, immunomodulatory agents and monoclonal antibodies are the most well known and studied classes of novel agents targeting myeloma, with histone deacetylase inhibitors, nuclear export inhibitors and several other approaches also being actively investigated. However, in parallel with the development and clinical use of these novel myeloma therapies is the emergence of novel mechanisms of resistance, many of which remain elusive, particularly for more recently developed agents. Whilst resistance mechanisms have been best studied for proteasome inhibitors, particularly bortezomib, class effects do not universally apply to all class members, and within-class differences in efficacy, toxicity and resistance mechanisms have been observed. Although immunomodulatory agents share the common cellular target cereblon and thus resistance patterns relate to cereblon expression, the unique cell surface antigens to which monoclonal antibodies are directed means these agents frequently exhibit unique within-class differences in clinical efficacy and resistance patterns. This review describes the major classes of novel therapies for myeloma, highlights the major clinical trials within each class and discusses known resistance mechanisms.
Collapse
Affiliation(s)
- Craig T Wallington-Beddoe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, South Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Magdalena Sobieraj-Teague
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia.,SA Pathology, Adelaide, Australia
| | - Bryone J Kuss
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia.,SA Pathology, Adelaide, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|
43
|
Cloos J, Roeten MS, Franke NE, van Meerloo J, Zweegman S, Kaspers GJ, Jansen G. (Immuno)proteasomes as therapeutic target in acute leukemia. Cancer Metastasis Rev 2018; 36:599-615. [PMID: 29071527 PMCID: PMC5721123 DOI: 10.1007/s10555-017-9699-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The clinical efficacy of proteasome inhibitors in the treatment of multiple myeloma has encouraged application of proteasome inhibitor containing therapeutic interventions in (pediatric) acute leukemia. Here, we summarize the positioning of bortezomib, as first-generation proteasome inhibitor, and second-generation proteasome inhibitors in leukemia treatment from a preclinical and clinical perspective. Potential markers for proteasome inhibitor sensitivity and/or resistance emerging from leukemia cell line models and clinical sample studies will be discussed focusing on the role of immunoproteasome and constitutive proteasome (subunit) expression, PSMB5 mutations, and alternative mechanisms of overcoming proteolytic stress.
Collapse
Affiliation(s)
- Jacqueline Cloos
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Margot Sf Roeten
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Niels E Franke
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Johan van Meerloo
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Departments of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gertjan Jl Kaspers
- Departments of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
- Princess Màxima Center, Utrecht, The Netherlands
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Franke NE, Kaspers GL, Assaraf YG, van Meerloo J, Niewerth D, Kessler FL, Poddighe PJ, Kole J, Smeets SJ, Ylstra B, Bi C, Chng WJ, Horton TM, Menezes RX, Musters RJP, Zweegman S, Jansen G, Cloos J. Exocytosis of polyubiquitinated proteins in bortezomib-resistant leukemia cells: a role for MARCKS in acquired resistance to proteasome inhibitors. Oncotarget 2018; 7:74779-74796. [PMID: 27542283 PMCID: PMC5342701 DOI: 10.18632/oncotarget.11340] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 07/26/2016] [Indexed: 12/11/2022] Open
Abstract
PSMB5 mutations and upregulation of the β5 subunit of the proteasome represent key determinants of acquired resistance to the proteasome inhibitor bortezomib (BTZ) in leukemic cells in vitro. We here undertook a multi-modality (DNA, mRNA, miRNA) array-based analysis of human CCRF-CEM leukemia cells and BTZ-resistant subclones to determine whether or not complementary mechanisms contribute to BTZ resistance. These studies revealed signatures of markedly reduced expression of proteolytic stress related genes in drug resistant cells over a broad range of BTZ concentrations along with a high upregulation of myristoylated alanine-rich C-kinase substrate (MARCKS) gene expression. MARCKS upregulation was confirmed on protein level and also observed in other BTZ-resistant tumor cell lines as well as in leukemia cells with acquired resistance to other proteasome inhibitors. Moreover, when MARCKS protein expression was demonstrated in specimens derived from therapy-refractory pediatric leukemia patients (n = 44), higher MARCKS protein expression trended (p = 0.073) towards a dismal response to BTZ-containing chemotherapy. Mechanistically, we show a BTZ concentration-dependent association of MARCKS protein levels with the emergence of ubiquitin-containing vesicles in BTZ-resistant CEM cells. These vesicles were found to be extruded and taken up in co-cultures with proteasome-proficient acceptor cells. Consistent with these observations, MARCKS protein associated with ubiquitin-containing vesicles was also more prominent in clinical leukemic specimen with ex vivo BTZ resistance compared to BTZ-sensitive leukemia cells. Collectively, we propose a role for MARCKS in a novel mechanism of BTZ resistance via exocytosis of ubiquitinated proteins in BTZ-resistant cells leading to quenching of proteolytic stress.
Collapse
Affiliation(s)
- Niels E Franke
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gertjan L Kaspers
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Technion-Israel Institute of Technology, Haifa, Israel
| | - Johan van Meerloo
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Denise Niewerth
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Floortje L Kessler
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Pino J Poddighe
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen Kole
- Department of Physiology, VU University, Amsterdam, The Netherlands
| | - Serge J Smeets
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Chonglei Bi
- Department of Experimental Therapeutics, Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Current address: BGI-Shenzhen, Shenzhen, China
| | - Wee Joo Chng
- Department of Experimental Therapeutics, Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Terzah M Horton
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Rene X Menezes
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gerrit Jansen
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
45
|
Roeten MSF, Cloos J, Jansen G. Positioning of proteasome inhibitors in therapy of solid malignancies. Cancer Chemother Pharmacol 2018; 81:227-243. [PMID: 29184971 PMCID: PMC5778165 DOI: 10.1007/s00280-017-3489-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/19/2017] [Indexed: 12/13/2022]
Abstract
Targeting of the protein degradation pathway, in particular, the ubiquitin-proteasome system, has emerged as an attractive novel cancer chemotherapeutic modality. Although proteasome inhibitors have been most successfully applied in the treatment of hematological malignancies, they also received continuing interest for the treatment of solid tumors. In this review, we summarize the current positioning of proteasome inhibitors in the treatment of common solid malignancies (e.g., lung, colon, pancreas, breast, and head and neck cancer), addressing topics of their mechanism(s) of action, predictive factors and molecular mechanisms of resistance.
Collapse
Affiliation(s)
- Margot S F Roeten
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands.
- Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Location VUmc, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Basler M, Maurits E, de Bruin G, Koerner J, Overkleeft HS, Groettrup M. Amelioration of autoimmunity with an inhibitor selectively targeting all active centres of the immunoproteasome. Br J Pharmacol 2017; 175:38-52. [PMID: 29034459 DOI: 10.1111/bph.14069] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Multicatalytic endopeptidase complex-like-1 (β2i), low molecular mass polypeptide (LMP) 2 (β1i) and LMP7 (β5i) are the proteolytically active subunits of the immunoproteasome, a special type of proteasome mainly expressed in haematopoietic cells. Targeting LMP7 has been shown to be therapeutically effective in preclinical models of autoimmune diseases. In this study, we investigated the selectivity and biological activity of LU-005i, a recently described inhibitor of the immunoproteasome. EXPERIMENTAL APPROACH The specificity of LU-005i and other immunoproteasome-selective inhibitors was characterized using fluorogenic peptide substrates. The effect of proteasome inhibition on cytokine release was investigated in endotoxin-stimulated mouse splenocytes or human peripheral blood mononuclear cells (PBMCs). The effect of proteasome inhibition on inflammatory bowel disease in the dextran sulfate sodium (DSS)-induced colitis model was assessed by measuring weight loss and colon length. KEY RESULTS LU-005i is the first human and mouse immunoproteasome-selective inhibitor that targets all three proteolytically active immunoproteasome subunits. LU-005i inhibited cytokine secretion from endotoxin-stimulated mouse splenocytes or human PBMCs. Furthermore, differentiation of naïve T helper cells to T helper 17 cells was impaired in the presence of LU-005i. Additionally, LU-005i ameliorated DSS-induced colitis. CONCLUSION AND IMPLICATIONS This study with a novel pan-immunoproteasome inhibitor substantiates that the immunoproteasome is a promising drug target for the treatment of inflammatory diseases and that exclusive inhibition of LMP7 is not necessary for therapeutic effectiveness. Our results will promote the design of new generations of immunoproteasome inhibitors with optimal therapeutic efficacy for clinical use in the treatment of autoimmunity and cancer.
Collapse
Affiliation(s)
- Michael Basler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, 8280, Switzerland.,Department of Biology, Division of Immunology, University of Konstanz, Konstanz, 78457, Germany
| | - Elmer Maurits
- Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Gerjan de Bruin
- Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Julia Koerner
- Department of Biology, Division of Immunology, University of Konstanz, Konstanz, 78457, Germany
| | - Herman S Overkleeft
- Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Marcus Groettrup
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, 8280, Switzerland.,Department of Biology, Division of Immunology, University of Konstanz, Konstanz, 78457, Germany
| |
Collapse
|
47
|
Zaal EA, Wu W, Jansen G, Zweegman S, Cloos J, Berkers CR. Bortezomib resistance in multiple myeloma is associated with increased serine synthesis. Cancer Metab 2017; 5:7. [PMID: 28855983 PMCID: PMC5575874 DOI: 10.1186/s40170-017-0169-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022] Open
Abstract
Background The proteasome inhibitor bortezomib (BTZ) is successfully applied in the treatment of multiple myeloma, but its efficacy is restricted by the wide-spread occurrence of resistance. Metabolic alterations play an important role in cancer development and aid in the cellular adaptation to pharmacologically changed environments. Metabolic changes could therefore play an essential role in the development of drug resistance. However, specific metabolic pathways that can be targeted to improve bortezomib therapy remain unidentified. Methods We elucidated the metabolic mechanisms underlying bortezomib resistance by using mass spectrometry-based metabolomics and proteomics on BTZ-sensitive and BTZ–resistant multiple myeloma cell lines as well as in a set of CD138+ cells obtained from multiple myeloma patients. Results Our findings demonstrate that a rewired glucose metabolism sustains bortezomib resistance. Mechanistically, this results in higher activity of both the pentose phosphate pathway and serine synthesis pathway, ultimately leading to an increased anti-oxidant capacity of BTZ-resistant cells. Moreover, our results link both serine synthesis pathway activity and expression of 3-phosphoglycerate dehydrogenase (PHGDH), which catalyzes the rate-limiting step of serine synthesis, to bortezomib resistance across different BTZ-resistant multiple myeloma cell lines. Consistently, serine starvation enhanced the cytotoxicity of bortezomib, underscoring the importance of serine metabolism in the response to BTZ. Importantly, in CD138+ cells of clinically bortezomib refractory multiple myeloma patients, PHGDH expression was also markedly increased. Conclusions Our findings indicate that interfering with serine metabolism may be a novel strategy to improve bortezomib therapy and identify PHGDH as a potential biomarker for BTZ resistance. Electronic supplementary material The online version of this article (doi:10.1186/s40170-017-0169-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Esther A Zaal
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center-Location VUMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Celia R Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
48
|
Ge Y, Li A, Wu J, Feng H, Wang L, Liu H, Xu Y, Xu Q, Zhao L, Li Y. Design, synthesis and biological evaluation of novel non-peptide boronic acid derivatives as proteasome inhibitors. Eur J Med Chem 2017; 128:180-191. [PMID: 28182990 DOI: 10.1016/j.ejmech.2017.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/03/2016] [Accepted: 01/21/2017] [Indexed: 10/20/2022]
Abstract
A novel series of non-peptide proteasome inhibitors bearing the 1, 4-naphthoquinone scaffold and boronic acid warhead was developed. In the biological evaluation on the chymotrypsin-like activity of human 20S proteasome, five compounds showed IC50 values in the nanomolar range. Docking experiments into the yeast 20S proteasome rationalized their biological activities and allowed further optimization of this interesting class of inhibitors. Within the cellular proliferation inhibition assay and western blot analysis, compound 3e demonstrated excellent anti-proliferative activity against solid tumor cells and clear accumulation of ubiquitinated cellular proteins. Furthermore, in the microsomal stability assay compound 3e demonstrated much improved metabolic stability compared to bortezomib, emerging as a promising lead compound for further design of non-peptide proteasome inhibitors.
Collapse
Affiliation(s)
- Ying Ge
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Aibo Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Jianwei Wu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Haiwei Feng
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Letian Wang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Hongwu Liu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yungen Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qingxiang Xu
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, Nanjing 210008, China.
| | - Li Zhao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| | - Yuyan Li
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
49
|
Deshmukh RR, Kim S, Elghoul Y, Dou QP. P-Glycoprotein Inhibition Sensitizes Human Breast Cancer Cells to Proteasome Inhibitors. J Cell Biochem 2017; 118:1239-1248. [PMID: 27813130 DOI: 10.1002/jcb.25783] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 11/09/2022]
Abstract
Although effective for the treatment of hematological malignancies, the FDA approved proteasome inhibitors bortezomib and carfilzomib have limited efficacy in solid tumors including triple negative breast cancer (TNBC). Chemotherapy is the only option for treating TNBC due to the absence of specific therapeutic targets. Therefore, development of new TNBC therapeutic strategies has been warranted. We studied whether P-glycoprotein (P-gp) inhibition could sensitize TNBC cells to proteasome inhibitors. When verapamil, a P-gp inhibitor, was combined with the proteasome inhibitor MG132, bortezomib, or carfilzomib, the cytotoxic effects and apoptosis in TNBC MDA-MB-231 cells were enhanced, compared to each treatment alone. Furthermore, addition of verapamil improved proteasome-inhibitory properties of MG132, bortezomib, or carfilzomib in MDA-MB-231 cells, as shown by the increased accumulation of ubiquitinated proteins and proteasome substrates such as IκBα and p27kip1 . Additionally, when nicardipine, another P-gp inhibitor, was combined with bortezomib or carfilzomib, enhanced inhibition of MDA-MB-231 cell proliferation was observed. These findings indicate that P-gp inhibitors could sensitize TNBC cells to structurally and functionally diverse proteasome inhibitors and might provide new treatment strategy for TNBC. J. Cell. Biochem. 118: 1239-1248, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rahul R Deshmukh
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.,School of Pharmacy, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, 34211
| | - Seongho Kim
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201-2013
| | - Yasmine Elghoul
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201-2013
| | - Q Ping Dou
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.,Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201-2013.,Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
50
|
Cree IA, Charlton P. Molecular chess? Hallmarks of anti-cancer drug resistance. BMC Cancer 2017; 17:10. [PMID: 28056859 PMCID: PMC5214767 DOI: 10.1186/s12885-016-2999-1] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022] Open
Abstract
Background The development of resistance is a problem shared by both classical chemotherapy and targeted therapy. Patients may respond well at first, but relapse is inevitable for many cancer patients, despite many improvements in drugs and their use over the last 40 years. Review Resistance to anti-cancer drugs can be acquired by several mechanisms within neoplastic cells, defined as (1) alteration of drug targets, (2) expression of drug pumps, (3) expression of detoxification mechanisms, (4) reduced susceptibility to apoptosis, (5) increased ability to repair DNA damage, and (6) altered proliferation. It is clear, however, that changes in stroma and tumour microenvironment, and local immunity can also contribute to the development of resistance. Cancer cells can and do use several of these mechanisms at one time, and there is considerable heterogeneity between tumours, necessitating an individualised approach to cancer treatment. As tumours are heterogeneous, positive selection of a drug-resistant population could help drive resistance, although acquired resistance cannot simply be viewed as overgrowth of a resistant cancer cell population. The development of such resistance mechanisms can be predicted from pre-existing genomic and proteomic profiles, and there are increasingly sophisticated methods to measure and then tackle these mechanisms in patients. Conclusion The oncologist is now required to be at least one step ahead of the cancer, a process that can be likened to ‘molecular chess’. Thus, as well as an increasing role for predictive biomarkers to clinically stratify patients, it is becoming clear that personalised strategies are required to obtain best results.
Collapse
Affiliation(s)
- Ian A Cree
- Department of Pathology, University Hospitals Coventry and Warwickshire, Coventry, CV2 2DX, UK. .,Faculty of Health and Life Sciences, Coventry University, Priory Street, Coventry, CV1 5FB, UK.
| | - Peter Charlton
- Imperial Innovations, 52 Princes Gate, Exhibition Road, London, SW7 2PG, UK
| |
Collapse
|