1
|
Shah A, Johnson E, Ponnusamy MP, Batra SK. Emerging pathways yielding opportunities for future treatments in pancreatic ductal adenocarcinoma. Expert Opin Ther Targets 2025:1-18. [PMID: 40382194 DOI: 10.1080/14728222.2025.2507035] [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: 12/31/2024] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy that is often diagnosed at a late stage, resulting in poor survival rates and limited treatment options. Several factors contribute to the dismal prognosis of PDAC, including the absence of reliable biomarkers and effective therapies, as well as the complex biology of the disease. AREAS COVERED The pathobiology of PDAC encompasses its unique mutational landscape, desmoplastic stroma, and immune suppressive tumor microenvironment (TME). These characteristics are influenced by an intricate network of signaling pathways activated by oncogenic KRAS, DNA damage and repair machinery, metabolic adaptations, and aberrant mucin expression. This review summarizes our current understanding of these pathways to explore their potential for therapeutic vulnerabilities in PDAC. We discuss how recent efforts to elucidate these pathways have identified novel targets and treatments for this dreadful disease. EXPERT OPINION The complex biology of PDAC complicates the effectiveness of single therapeutic agents. To achieve durable clinical responses in patients with PDAC, it is essential to simultaneously inhibit multiple parallel or unrelated pathways. Therefore, a combination therapeutic regimen is necessary to significantly improve treatment outcomes that rely solely on biologically driven concepts. These studies suggest ways to expand our understanding of the therapeutic vulnerabilities in PDAC.
Collapse
Affiliation(s)
- Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Esther Johnson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
2
|
Meng F, Li T, Singh AK, Wang Y, Attiyeh M, Kohram F, Feng Q, Li YR, Shen B, Williams T, Liu Y, Raoof M. Base-excision repair pathway regulates transcription-replication conflicts in pancreatic ductal adenocarcinoma. Cell Rep 2024; 43:114820. [PMID: 39368091 DOI: 10.1016/j.celrep.2024.114820] [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: 11/24/2023] [Revised: 07/19/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024] Open
Abstract
Oncogenic mutations (such as in KRAS) can dysregulate transcription and replication, leading to transcription-replication conflicts (TRCs). Here, we demonstrate that TRCs are enriched in human pancreatic ductal adenocarcinoma (PDAC) compared to other common solid tumors or normal cells. Several orthogonal approaches demonstrated that TRCs are oncogene dependent. A small interfering RNA (siRNA) screen identified several factors in the base-excision repair (BER) pathway as main regulators of TRCs in PDAC cells. Inhibitors of BER pathway (methoxyamine and CRT) enhanced TRCs. Mechanistically, BER pathway inhibition severely altered RNA polymerase II (RNAPII) and R-loop dynamics at nascent DNA, causing RNAPII trapping and contributing to enhanced TRCs. The ensuing DNA damage activated the ATR-Chk1 pathway. Co-treatment with ATR inhibitor (VX970) and BER inhibitor (methoxyamine) at clinically relevant doses synergistically enhanced DNA damage and reduced cell proliferation in PDAC cells. The study provides mechanistic insights into the regulation of TRCs in PDAC by the BER pathway, which has biologic and therapeutic implications.
Collapse
Affiliation(s)
- Fan Meng
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Tiane Li
- Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA, USA; Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Yingying Wang
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Marc Attiyeh
- Department of Surgery, Cedars Sinai, Los Angeles, CA, USA
| | - Fatemeh Kohram
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Qianhua Feng
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yun R Li
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Binghui Shen
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Terence Williams
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yilun Liu
- Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA; Department of Cancer Genetic & Epigenetics, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
3
|
Guo L, Ding Z, Hu J, Liu S. Efficient Encapsulation of β-Lapachone into Self-Immolative Polymer Nanoparticles for Cyclic Amplification of Intracellular Reactive Oxygen Species Stress. ACS NANO 2024. [PMID: 39263977 DOI: 10.1021/acsnano.4c09232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The selective upregulation of intracellular oxidative stress in cancer cells presents a promising approach for effective cancer treatment. In this study, we report the integration of enzyme catalytic amplification and chemical amplification reactions in β-lapachone (Lap)-loaded micellar nanoparticles (NPs), which are self-assembled from reactive oxygen species (ROS)-responsive self-immolative polymers (SIPs). This integration enables cyclic amplification of intracellular oxidative stress in cancer cells. Specifically, we have developed ROS-responsive SIPs with phenylboronic ester triggering motifs and hexafluoroisopropanol moieties in the side chains, significantly enhancing Lap loading efficiency (98%) and loading capacity (33%) through multiple noncovalent interactions. Upon ROS activation in tumor cells, the Lap-loaded micellar NPs disassemble, releasing Lap and generating additional ROS via enzyme catalytic amplification. This process elevates intracellular oxidative stress and triggers polymer depolymerization in a positive feedback loop. Furthermore, the degradation of SIPs via chemical amplification produces azaquinone methide intermediates, which consume intracellular thiol-related substrates, disrupt intracellular redox hemostasis, further intensify oxidative stress, and promote cancer cell apoptosis. This work introduces a strategy to enhance intracellular oxidative stress by combining enzymatic and chemical amplification reactions, providing a potential pathway for the development of highly efficient anticancer agents.
Collapse
Affiliation(s)
- Lingxiao Guo
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Zexuan Ding
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| |
Collapse
|
4
|
Runnebohm AM, Wijeratne HRS, Justice SAP, Wijeratne AB, Roy G, Singh N, Hergenrother P, Boothman DA, Motea EA, Mosley AL. IB-DNQ and Rucaparib dual treatment alters cell cycle regulation and DNA repair in triple negative breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594427. [PMID: 38798459 PMCID: PMC11118307 DOI: 10.1101/2024.05.15.594427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Triple negative breast cancer (TNBC), characterized by the lack of three canonical receptors, is unresponsive to commonly used hormonal therapies. One potential TNBC-specific therapeutic target is NQO1, as it is highly expressed in many TNBC patients and lowly expressed in non-cancer tissues. DNA damage induced by NQO1 bioactivatable drugs in combination with Rucaparib-mediated inhibition of PARP1-dependent DNA repair synergistically induces cell death. Methods To gain a better understanding of the mechanisms behind this synergistic effect, we used global proteomics, phosphoproteomics, and thermal proteome profiling to analyze changes in protein abundance, phosphorylation and protein thermal stability. Results Very few protein abundance changes resulted from single or dual agent treatment; however, protein phosphorylation and thermal stability were impacted. Histone H2AX was among several proteins identified to have increased phosphorylation when cells were treated with the combination of IB-DNQ and Rucaparib, validating that the drugs induced persistent DNA damage. Thermal proteome profiling revealed destabilization of H2AX following combination treatment, potentially a result of the increase in phosphorylation. Kinase substrate enrichment analysis predicted altered activity for kinases involved in DNA repair and cell cycle following dual agent treatment. Further biophysical analysis of these two processes revealed alterations in SWI/SNF complex association and tubulin / p53 interactions. Conclusions Our findings that the drugs target DNA repair and cell cycle regulation, canonical cancer treatment targets, in a way that is dependent on increased expression of a protein selectively found to be upregulated in cancers without impacting protein abundance illustrate that multi-omics methodologies are important to gain a deeper understanding of the mechanisms behind treatment induced cancer cell death.
Collapse
Affiliation(s)
- Avery M Runnebohm
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - H R Sagara Wijeratne
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Sarah A Peck Justice
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Department of Biology, Marian University, Indianapolis, IN
| | - Aruna B Wijeratne
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- IU Simon Comprehensive Cancer Center, Indianapolis, IN
| | - Gitanjali Roy
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | | | - Paul Hergenrother
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- IU Simon Comprehensive Cancer Center, Indianapolis, IN
| | - Edward A Motea
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- IU Simon Comprehensive Cancer Center, Indianapolis, IN
| | - Amber L Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- IU Simon Comprehensive Cancer Center, Indianapolis, IN
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
5
|
Song J, Zou G, Zhao Z, Zhu Y, Xue J, Ao L, Sun H, Hao H, Zhang B, Xu X. Discovery of proqodine A derivatives with antitumor activity targeting NAD(P)H: quinone oxidoreductase 1 and nicotinamide phosphoribosyltransferase. Chin J Nat Med 2024; 22:75-88. [PMID: 38278561 DOI: 10.1016/s1875-5364(24)60564-9] [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: 08/12/2023] [Indexed: 01/28/2024]
Abstract
NAD(P)H: quinone oxidoreductase 1 (NQO1) is a flavin protease highly expressed in various cancer cells. NQO1 catalyzes a futile redox cycle in substrates, leading to substantial reactive oxygen species (ROS) production. This ROS generation results in extensive DNA damage and elevated poly (ADP-ribose) polymerase 1 (PARP1)-mediated consumption of nicotinamide adenine dinucleotide (NAD+), ultimately causing cell death. Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD+ salvage synthesis pathway, emerges as a critical target in cancer therapy. The concurrent inhibition of NQO1 and NAMPT triggers hyperactivation of PARP1 and intensive NAD+ depletion. In this study, we designed, synthesized, and assessed a novel series of proqodine A derivatives targeting both NQO1 and NAMPT. Among these, compound T8 demonstrated potent antitumor properties. Specifically, T8 selectively inhibited the proliferation of MCF-7 cells and induced apoptosis through mechanisms dependent on both NQO1 and NAMPT. This discovery offers a promising new molecular entity for advancing anticancer research.
Collapse
Affiliation(s)
- Jiangzhou Song
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Guiqing Zou
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhou Zhao
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Ya Zhu
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Jiayu Xue
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Lanjia Ao
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Huiyong Sun
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| | - Bo Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaowei Xu
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
Wang J, Su X, Jiang L, Boudreau MW, Chatkewitz LE, Kilgore JA, Zahid KR, Williams NS, Chen Y, Liu S, Hergenrother PJ, Huang X. Augmented Concentration of Isopentyl-Deoxynyboquinone in Tumors Selectively Kills NAD(P)H Quinone Oxidoreductase 1-Positive Cancer Cells through Programmed Necrotic and Apoptotic Mechanisms. Cancers (Basel) 2023; 15:5844. [PMID: 38136388 PMCID: PMC10741405 DOI: 10.3390/cancers15245844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Lung and breast cancers rank as two of the most common and lethal tumors, accounting for a substantial number of cancer-related deaths worldwide. While the past two decades have witnessed promising progress in tumor therapy, developing targeted tumor therapies continues to pose a significant challenge. NAD(P)H quinone oxidoreductase 1 (NQO1), a two-electron reductase, has been reported as a promising therapeutic target across various solid tumors. β-Lapachone (β-Lap) and deoxynyboquinone (DNQ) are two NQO1 bioactivatable drugs that have demonstrated potent antitumor effects. However, their curative efficacy has been constrained by adverse effects and moderate lethality. To enhance the curative potential of NQO1 bioactivatable drugs, we developed a novel DNQ derivative termed isopentyl-deoxynyboquinone (IP-DNQ). Our study revealed that IP-DNQ treatment significantly increased reactive oxygen species generation, leading to double-strand break (DSB) formation, PARP1 hyperactivation, and catastrophic energy loss. Notably, we discovered that this novel drug induced both apoptosis and programmed necrosis events, which makes it entirely distinct from other NQO1 bioactivatable drugs. Furthermore, IP-DNQ monotherapy demonstrated significant antitumor efficacy and extended mice survival in A549 orthotopic xenograft models. Lastly, we identified that in mice IP-DNQ levels were significantly elevated in the plasma and tumor compared with IB-DNQ levels. This study provides novel preclinical evidence supporting IP-DNQ efficacy in NQO1+ NSCLC and breast cancer cells.
Collapse
Affiliation(s)
- Jiangwei Wang
- Department of Radiation Oncology, Indianapolis, IN 46202, USA
| | - Xiaolin Su
- Department of Biochemistry and Molecular Biology, Indianapolis, IN 46202, USA
| | - Lingxiang Jiang
- Department of Radiation Oncology, Indianapolis, IN 46202, USA
| | - Matthew W. Boudreau
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Lindsay E. Chatkewitz
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jessica A. Kilgore
- Department of Biochemistry, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA (N.S.W.)
| | | | - Noelle S. Williams
- Department of Biochemistry, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA (N.S.W.)
| | - Yaomin Chen
- Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shaohui Liu
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul J. Hergenrother
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiumei Huang
- Department of Radiation Oncology, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
7
|
Eisenbeis VB, Qiu D, Gorka O, Strotmann L, Liu G, Prucker I, Su XB, Wilson MSC, Ritter K, Loenarz C, Groß O, Saiardi A, Jessen HJ. β-lapachone regulates mammalian inositol pyrophosphate levels in an NQO1- and oxygen-dependent manner. Proc Natl Acad Sci U S A 2023; 120:e2306868120. [PMID: 37579180 PMCID: PMC10450438 DOI: 10.1073/pnas.2306868120] [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: 05/02/2023] [Accepted: 07/13/2023] [Indexed: 08/16/2023] Open
Abstract
Inositol pyrophosphates (PP-InsPs) are energetic signaling molecules with important functions in mammals. As their biosynthesis depends on ATP concentration, PP-InsPs are tightly connected to cellular energy homeostasis. Consequently, an increasing number of studies involve PP-InsPs in metabolic disorders, such as type 2 diabetes, aspects of tumorigenesis, and hyperphosphatemia. Research conducted in yeast suggests that the PP-InsP pathway is activated in response to reactive oxygen species (ROS). However, the precise modulation of PP-InsPs during cellular ROS signaling is unknown. Here, we report how mammalian PP-InsP levels are changing during exposure to exogenous (H2O2) and endogenous ROS. Using capillary electrophoresis electrospray ionization mass spectrometry (CE-ESI-MS), we found that PP-InsP levels decrease upon exposure to oxidative stressors in HCT116 cells. Application of quinone drugs, particularly β-lapachone (β-lap), under normoxic and hypoxic conditions enabled us to produce ROS in cellulo and to show that β-lap treatment caused PP-InsP changes that are oxygen-dependent. Experiments in MDA-MB-231 breast cancer cells deficient of NAD(P)H:quinone oxidoreductase-1 (NQO1) demonstrated that β-lap requires NQO1 bioactivation to regulate the cellular metabolism of PP-InsPs. Critically, significant reductions in cellular ATP concentrations were not directly mirrored in reduced PP-InsP levels as shown in NQO1-deficient MDA-MB-231 cells treated with β-lap. The data presented here unveil unique aspects of β-lap pharmacology and its impact on PP-InsP levels. The identification of different quinone drugs as modulators of PP-InsP synthesis will allow the overall impact on cellular function of such drugs to be better appreciated.
Collapse
Affiliation(s)
- Verena B. Eisenbeis
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Danye Qiu
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg79106, Germany
| | - Lisa Strotmann
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Guizhen Liu
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Isabel Prucker
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Xue Bessie Su
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, United Kingdom
| | - Miranda S. C. Wilson
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, United Kingdom
| | - Kevin Ritter
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Christoph Loenarz
- Faculty of Chemistry and Pharmacy, Institute for Pharmaceutical Sciences, Pharmaceutical and Medicinal Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Olaf Groß
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg79106, Germany
| | - Adolfo Saiardi
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, United Kingdom
| | - Henning J. Jessen
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| |
Collapse
|
8
|
Zeng Z, Zheng W, Hou P. The role of drug-metabolizing enzymes in synthetic lethality of cancer. Pharmacol Ther 2022; 240:108219. [PMID: 35636517 DOI: 10.1016/j.pharmthera.2022.108219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022]
Abstract
Drug-metabolizing enzymes (DMEs) have shown increasing importance in anticancer therapy. It is not only due to their effect on activation or deactivation of anticancer drugs, but also because of their extensive connections with pathological and biochemistry changes during tumorigenesis. Meanwhile, it has become more accessible to discovery anticancer drugs that selectively targeted cancer cells with the development of synthetic lethal screen technology. Synthetic lethal strategy makes use of unique genetic markers that different cancer cells from normal tissues to discovery anticancer agents. Dysregulation of DMEs has been found in various cancers, making them promising candidates for synthetic lethal strategy. In this review, we will systematically discuss about the role of DMEs in tumor progression, the application of synthetic lethality strategy in drug discovery, and a link between DMEs and synthetic lethal of cancer.
Collapse
Affiliation(s)
- Zekun Zeng
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Wenfang Zheng
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
9
|
Li C, Xue Y, Ba X, Wang R. The Role of 8-oxoG Repair Systems in Tumorigenesis and Cancer Therapy. Cells 2022; 11:cells11233798. [PMID: 36497058 PMCID: PMC9735852 DOI: 10.3390/cells11233798] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/09/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Tumorigenesis is highly correlated with the accumulation of mutations. The abundant and extensive DNA oxidation product, 8-Oxoguanine (8-oxoG), can cause mutations if it is not repaired by 8-oxoG repair systems. Therefore, the accumulation of 8-oxoG plays an essential role in tumorigenesis. To avoid the accumulation of 8-oxoG in the genome, base excision repair (BER), initiated by 8-oxoguanine DNA glycosylase1 (OGG1), is responsible for the removal of genomic 8-oxoG. It has been proven that 8-oxoG levels are significantly elevated in cancer cells compared with cells of normal tissues, and the induction of DNA damage by some antitumor drugs involves direct or indirect interference with BER, especially through inducing the production and accumulation of reactive oxygen species (ROS), which can lead to tumor cell death. In addition, the absence of the core components of BER can result in embryonic or early post-natal lethality in mice. Therefore, targeting 8-oxoG repair systems with inhibitors is a promising avenue for tumor therapy. In this study, we summarize the impact of 8-oxoG accumulation on tumorigenesis and the current status of cancer therapy approaches exploiting 8-oxoG repair enzyme targeting, as well as possible synergistic lethality strategies involving exogenous ROS-inducing agents.
Collapse
Affiliation(s)
- Chunshuang Li
- Center for Cell Structure and Function, Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Sciences, Shandong Normal University, Jinan 250014, China
- The Key Laboratory of Molecular Epigenetics of Education, School of Life Science, Northeast Normal University, Changchun 130024, China
| | - Yaoyao Xue
- The Key Laboratory of Molecular Epigenetics of Education, School of Life Science, Northeast Normal University, Changchun 130024, China
| | - Xueqing Ba
- The Key Laboratory of Molecular Epigenetics of Education, School of Life Science, Northeast Normal University, Changchun 130024, China
- Correspondence: (X.B.); (R.W.)
| | - Ruoxi Wang
- Center for Cell Structure and Function, Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Sciences, Shandong Normal University, Jinan 250014, China
- Correspondence: (X.B.); (R.W.)
| |
Collapse
|
10
|
Dou L, Liu H, Wang K, Liu J, Liu L, Ye J, Wang R, Deng H, Qian F. Albumin binding revitalizes NQO1 bioactivatable drugs as novel therapeutics for pancreatic cancer. J Control Release 2022; 349:876-889. [PMID: 35907592 DOI: 10.1016/j.jconrel.2022.07.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/10/2022] [Accepted: 07/24/2022] [Indexed: 11/16/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) is an enzyme significantly overexpressed in pancreatic ductal adenocarcinoma (PDAC) tumors compared to the associated normal tissues. NQO1 bioactivatable drugs, such as β-lapachone (β-lap), can be catalyzed to generate reactive oxygen species (ROS) for direct tumor killing. However, the extremely narrow therapeutic window caused by methemoglobinemia and hemolytic anemia severely restricts its further clinical translation despite considerable efforts in the past 20 years. Previously, we demonstrated that albumin could be utilized to deliver cytotoxic drugs selectively into KRAS-mutant PDAC with a much expanded therapeutic window due to KRAS-enhanced macropinocytosis and reduced neonatal Fc receptor (FcRn) expression in PDAC. Herein, we analyzed the expression patterns of albumin and FcRn across major organs in LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) mice. The tumors were the predominant tissues with both elevated albumin and reduced FcRn expression, thus making them an ideal target for albumin-based drug delivery. Quantitative proteomics analysis of tissue samples from 5 human PDAC patients further confirmed the elevated albumin/FcRn ratio. Given such a compelling biological rationale, we designed a nanoparticle albumin-bound prodrug of β-lap, nab-(pro-β-lap), to achieve PDAC targeted delivery and expand the therapeutic window of β-lap. We found that nab-(pro-β-lap) uptake was profoundly enhanced by KRAS mutation. Compared to the solution formulation of the parent drug β-lap, nab-(pro-β-lap) showed enhanced safety due to much lower rates of methemoglobinemia and hemolytic anemia, which was confirmed both in vitro and in vivo. Furthermore, nab-(pro-β-lap) significantly inhibited tumor growth in subcutaneously implanted KPC xenografts and enhanced the pharmacodynamic endpoints (e.g., PARP1 hyperactivation, γ-H2AX). Thus, nab-(pro-β-lap), with improved safety and antitumor efficacy, offers a drug delivery strategy with translational viability for β-lap in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Lei Dou
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Huiqin Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Kaixin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Jing Liu
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, PR China
| | - Lei Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Junxiao Ye
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Rui Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, PR China
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
11
|
Chang MC, Mahar R, McLeod MA, Giacalone AG, Huang X, Boothman DA, Merritt ME. Synergistic Effect of β-Lapachone and Aminooxyacetic Acid on Central Metabolism in Breast Cancer. Nutrients 2022; 14:3020. [PMID: 35893874 PMCID: PMC9331106 DOI: 10.3390/nu14153020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Abstract
The compound β-lapachone, a naturally derived naphthoquinone, has been utilized as a potent medicinal nutrient to improve health. Over the last twelve years, numerous reports have demonstrated distinct associations of β-lapachone and NAD(P)H: quinone oxidoreductase 1 (NQO1) protein in the amelioration of various diseases. Comprehensive research of NQO1 bioactivity has clearly confirmed the tumoricidal effects of β-lapachone action through NAD+-keresis, in which severe DNA damage from reactive oxygen species (ROS) production triggers a poly-ADP-ribose polymerase-I (PARP1) hyperactivation cascade, culminating in NAD+/ATP depletion. Here, we report a novel combination strategy with aminooxyacetic acid (AOA), an aspartate aminotransferase inhibitor that blocks the malate-aspartate shuttle (MAS) and synergistically enhances the efficacy of β-lapachone metabolic perturbation in NQO1+ breast cancer. We evaluated metabolic turnover in MDA-MB-231 NQO1+, MDA-MB-231 NQO1-, MDA-MB-468, and T47D cancer cells by measuring the isotopic labeling of metabolites from a [U-13C]glucose tracer. We show that β-lapachone treatment significantly hampers lactate secretion by ~85% in NQO1+ cells. Our data demonstrate that combinatorial treatment decreases citrate, glutamate, and succinate enrichment by ~14%, ~50%, and ~65%, respectively. Differences in citrate, glutamate, and succinate fractional enrichments indicate synergistic effects on central metabolism based on the coefficient of drug interaction. Metabolic modeling suggests that increased glutamine anaplerosis is protective in the case of MAS inhibition.
Collapse
Affiliation(s)
- Mario C. Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (M.C.C.); (R.M.); (M.A.M.); (A.G.G.)
| | - Rohit Mahar
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (M.C.C.); (R.M.); (M.A.M.); (A.G.G.)
| | - Marc A. McLeod
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (M.C.C.); (R.M.); (M.A.M.); (A.G.G.)
| | - Anthony G. Giacalone
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (M.C.C.); (R.M.); (M.A.M.); (A.G.G.)
| | - Xiumei Huang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - David A. Boothman
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (M.C.C.); (R.M.); (M.A.M.); (A.G.G.)
| |
Collapse
|
12
|
Lee WS, Ham W, Kim J. Roles of NAD(P)H:quinone Oxidoreductase 1 in Diverse Diseases. Life (Basel) 2021; 11:life11121301. [PMID: 34947831 PMCID: PMC8703842 DOI: 10.3390/life11121301] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023] Open
Abstract
NAD(P)H:quinone oxidoreductase (NQO) is an antioxidant flavoprotein that catalyzes the reduction of highly reactive quinone metabolites by employing NAD(P)H as an electron donor. There are two NQO enzymes—NQO1 and NQO2—in mammalian systems. In particular, NQO1 exerts many biological activities, including antioxidant activities, anti-inflammatory effects, and interactions with tumor suppressors. Moreover, several recent studies have revealed the promising roles of NQO1 in protecting against cardiovascular damage and related diseases, such as dyslipidemia, atherosclerosis, insulin resistance, and metabolic syndrome. In this review, we discuss recent developments in the molecular regulation and biochemical properties of NQO1, and describe the potential beneficial roles of NQO1 in diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
- Correspondence: (W.-S.L.); (J.K.); Tel.: +82-2-6299-1419 (W.-S.L.); +82-2-6299-1397 (J.K.)
| | - Woojin Ham
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
- Correspondence: (W.-S.L.); (J.K.); Tel.: +82-2-6299-1419 (W.-S.L.); +82-2-6299-1397 (J.K.)
| |
Collapse
|
13
|
Zheng Y, Zhang H, Guo Y, Chen Y, Chen H, Liu Y. X-ray repair cross-complementing protein 1 (XRCC1) loss promotes β-lapachone -induced apoptosis in pancreatic cancer cells. BMC Cancer 2021; 21:1234. [PMID: 34789190 PMCID: PMC8600733 DOI: 10.1186/s12885-021-08979-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
Background β-lapachone (β-lap), the NQO1 bioactivatable drug, is thought to be a promising anticancer agent. However, the toxic side effects of β-lap limit the drug use, highlighting the need for a thorough understanding of β-lap’s mechanism of action. β-lap undergoes NQO1-dependent futile redox cycling, generating massive ROS and oxidative DNA lesions, leading to cell death. Thus, base excision repair (BER) pathway is an important resistance factor. XRCC1, a scaffolding component, plays a critical role in BER. Methods We knocked down XRCC1 expression by using pLVX-shXRCC1 in the MiaPaCa2 cells and BxPC3 cells and evaluated β-lap-induced DNA lesions by γH2AX foci formation and alkaline comet assay. The cell death induced by XRCC1 knockdown + β-lap treatment was analysed by relative survival, flow cytometry and Western blotting analysis. Results We found that knockdown of XRCC1 significantly increased β-lap-induced DNA double-strand breaks, comet tail lengths and cell death in PDA cells. Furthermore, we observed combining XRCC1 knockdown with β-lap treatment switched programmed necrosis with β-lap monotherapy to caspase-dependent apoptosis. Conclusions These results indicate that XRCC1 is involved in the repair of β-lap-induced DNA damage, and XRCC1 loss amplifies sensitivity to β-lap, suggesting targeting key components in BER pathways may have the potential to expand use and efficacy of β-lap for gene-based therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08979-y.
Collapse
Affiliation(s)
- Yansong Zheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Hengce Zhang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Science, Fujian Medical University, Fuzhou City, 350122, Fujian Province, China
| | - Yueting Guo
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Science, Fujian Medical University, Fuzhou City, 350122, Fujian Province, China
| | - Yuan Chen
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Science, Fujian Medical University, Fuzhou City, 350122, Fujian Province, China
| | - Hanglong Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou City, 350122, Fujian Province, China
| | - Yingchun Liu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Science, Fujian Medical University, Fuzhou City, 350122, Fujian Province, China.
| |
Collapse
|
14
|
Zhao W, Jiang L, Fang T, Fang F, Liu Y, Zhao Y, You Y, Zhou H, Su X, Wang J, Liu S, Chen Y, Wan J, Huang X. β-Lapachone Selectively Kills Hepatocellular Carcinoma Cells by Targeting NQO1 to Induce Extensive DNA Damage and PARP1 Hyperactivation. Front Oncol 2021; 11:747282. [PMID: 34676172 PMCID: PMC8523939 DOI: 10.3389/fonc.2021.747282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death globally. Currently there is a lack of tumor-selective and efficacious therapies for hepatocellular carcinoma. β-Lapachone (ARQ761 in clinical form) selectively kill NADPH: quinone oxidoreductase 1 (NQO1)-overexpressing cancer cells. However, the effect of β-Lapachone on HCC is virtually unknown. In this study, we found that relatively high NQO1 and low catalase levels were observed in both clinical specimens collected from HCC patients and HCC tumors from the TCGA database. β-Lapachone treatment induced NQO1-selective killing of HCC cells and caused ROS formation and PARP1 hyperactivation, resulting in a significant decrease in NAD+ and ATP levels and a dramatic increase in double-strand break (DSB) lesions over time in vitro. Administration of β-Lapachone significantly inhibited tumor growth and prolonged survival in a mouse xenograft model in vivo. Our data suggest that NQO1 is an ideal potential biomarker, and relatively high NQO1:CAT ratios in HCC tumors but low ratios in normal tissues offer an optimal therapeutic window to use β-Lapachone. This study provides novel preclinical evidence for β-Lapachone as a new promising chemotherapeutic agent for use in NQO1-positive HCC patients.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Lingxiang Jiang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ting Fang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Fei Fang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Yingchun Liu
- Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ye Zhao
- Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yuting You
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Hao Zhou
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaolin Su
- Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jiangwei Wang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yaomin Chen
- Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Computational Biology and Bioinformatics, Indiana University, School of Medicine, Indianapolis, IN, United States
| | - Xiumei Huang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
15
|
Mahar R, Chang MC, Merritt ME. Measuring NQO1 Bioactivation Using [ 2H 7]Glucose. Cancers (Basel) 2021; 13:4165. [PMID: 34439319 PMCID: PMC8392257 DOI: 10.3390/cancers13164165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 12/02/2022] Open
Abstract
Treatment of cancers with β-lapachone causes NAD(P)H: quinone oxidoreductase 1 (NQO1) to generate an unstable hydroquinone that regenerates itself in a futile cycle while producing reactive oxygen species (ROS) in the form of superoxide and subsequently hydrogen peroxide. Rapid accumulation of ROS damages DNA, hyperactivates poly-ADP-ribose polymerase-I, causes massive depletion of NAD+/ATP, and hampers glycolysis. Cells overexpressing NQO1 subsequently die rapidly through an NAD+-keresis mechanism. Assessing changes in glycolytic rates caused by NQO1 bioactivation would provide a means of assessing treatment efficacy, potentially lowering the chemotherapeutic dosage, and reducing off-target toxicities. NQO1-mediated changes in glycolytic flux were readily detected in A549 (lung), MiaPaCa2 (pancreatic), and HCT-116 (colon) cancer cell lines by 2H-NMR after administration of [2H7]glucose. The deuterated metabolic products 2H-lactate and HDO were quantified, and linear relationships with glucose consumption for both products were observed. The higher concentration of HDO compared to 2H-lactate allows for more sensitive measurement of the glycolytic flux in cancer. Gas chromatography-mass spectrometry analysis agreed with the NMR results and confirmed downregulated energy metabolism in NQO1+ cells after β-lapachone treatment. The demonstrated method is ideal for measuring glycolytic rates, the effects of chemotherapeutics that target glycolysis, and has the potential for in vivo translation.
Collapse
Affiliation(s)
| | | | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.M.); (M.C.C.)
| |
Collapse
|
16
|
Ross D, Siegel D. The diverse functionality of NQO1 and its roles in redox control. Redox Biol 2021; 41:101950. [PMID: 33774477 PMCID: PMC8027776 DOI: 10.1016/j.redox.2021.101950] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
In this review, we summarize the multiple functions of NQO1, its established roles in redox processes and potential roles in redox control that are currently emerging. NQO1 has attracted interest due to its roles in cell defense and marked inducibility during cellular stress. Exogenous substrates for NQO1 include many xenobiotic quinones. Since NQO1 is highly expressed in many solid tumors, including via upregulation of Nrf2, the design of compounds activated by NQO1 and NQO1-targeted drug delivery have been active areas of research. Endogenous substrates have also been proposed and of relevance to redox stress are ubiquinone and vitamin E quinone, components of the plasma membrane redox system. Established roles for NQO1 include a superoxide reductase activity, NAD+ generation, interaction with proteins and their stabilization against proteasomal degradation, binding and regulation of mRNA translation and binding to microtubules including the mitotic spindles. We also summarize potential roles for NQO1 in regulation of glucose and insulin metabolism with relevance to diabetes and the metabolic syndrome, in Alzheimer's disease and in aging. The conformation and molecular interactions of NQO1 can be modulated by changes in the pyridine nucleotide redox balance suggesting that NQO1 may function as a redox-dependent molecular switch.
Collapse
Affiliation(s)
- David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
17
|
Viera T, Patidar PL. DNA damage induced by KP372-1 hyperactivates PARP1 and enhances lethality of pancreatic cancer cells with PARP inhibition. Sci Rep 2020; 10:20210. [PMID: 33214574 PMCID: PMC7677541 DOI: 10.1038/s41598-020-76850-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/28/2020] [Indexed: 12/23/2022] Open
Abstract
The overall prognosis for pancreatic cancer remains dismal and potent chemotherapeutic agents that selectively target this cancer are critically needed. Elevated expression of NAD(P)H:quinone oxidoreductase 1 (NQO1) is frequent in pancreatic cancer, and it offers promising tumor-selective targeting. Recently, KP372-1 was identified as a novel NQO1 redox cycling agent that induces cytotoxicity in cancer cells by creating redox imbalance; however, the mechanistic basis of KP372-1-induced cytotoxicity remains elusive. Here, we show that KP372-1 sensitizes NQO1-expressing pancreatic cancer cells and spares immortalized normal pancreatic duct cells, hTERT-HPNE. Notably, we found that KP372-1 is ~ 10- to 20-fold more potent than β-lapachone, another NQO1 substrate, against pancreatic cancer cells. Mechanistically, our data strongly suggest that reactive oxygen species produced by NQO1-dependent redox cycling of KP372-1 cause robust DNA damage, including DNA breaks. Furthermore, we found that KP372-1-induced DNA damage hyperactivates the central DNA damage sensor protein poly(ADP-ribose) polymerase 1 (PARP1) and activates caspase-3 to initiate cell death. Our data also show that the combination of KP372-1 with PARP inhibition creates enhanced cytotoxicity in pancreatic cancer cells. Collectively, our study provides mechanistic insights into the cytotoxicity instigated by KP372-1 and lays an essential foundation to establish it as a promising chemotherapeutic agent against cancer.
Collapse
Affiliation(s)
- Talysa Viera
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Pl, Socorro, NM, 87801, USA
| | - Praveen L Patidar
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Pl, Socorro, NM, 87801, USA.
| |
Collapse
|
18
|
Inokuchi S, Itoh S, Yoshizumi T, Yugawa K, Yoshiya S, Toshima T, Takeishi K, Iguchi T, Sanefuji K, Harada N, Sugimachi K, Ikegami T, Kohashi K, Taguchi K, Yonemasu H, Fukuzawa K, Oda Y, Mori M. Mitochondrial expression of the DNA repair enzyme OGG1 improves the prognosis of pancreatic ductal adenocarcinoma. Pancreatology 2020; 20:1175-1182. [PMID: 32741713 DOI: 10.1016/j.pan.2020.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 07/05/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES 8-Hydroxydeoxyguanosine (8-OHdG) is an indicator of oxidative stress and causes transversion mutations and carcinogenesis. 8-OHdG is excision repaired by 8-OHdG DNA glycosylase 1 (OGG1), which is classified as nuclear and mitochondrial subtypes. We aimed to clarify the role of OGG1 in pancreatic ductal adenocarcinoma (PDAC). METHODS Ninety-two patients with PDAC who had undergone surgical resection at multiple institutions were immunohistochemically analyzed. The OGG1 and 8-OHdG expression levels were scored using the Germann Immunoreactive Score. The cutoff values of OGG1, as well as that of 8-OHdG, were determined. RESULTS The low nuclear OGG1 expression group (n = 41) showed significantly higher carbohydrate antigen (CA)19-9 (p = 0.026), and higher s-pancreas antigen (SPAN)-1 (p = 0.017) than the high expression group (n = 51). Nuclear OGG1 expression has no effect on the prognosis. The low mitochondrial OGG1 expression group (n = 40) showed higher CA19-9 (p = 0.041), higher SPAN-1 (p = 0.032), and more histological perineural invasion (p = 0.037) than the high expression group (n = 52). The low mitochondrial OGG1 expression group had a significantly shorter recurrence-free survival (p = 0.0080) and overall survival (p = 0.0073) rates. The Cox proportional hazards model revealed that low mitochondrial OGG1 expression is an independent risk factor of the PDAC prognosis. OGG1 expression was negatively correlated with 8-OHdG expression (p = 0.0004), and high 8-OHdG expression shortened the recurrence-free survival of patients with PDAC. CONCLUSIONS Low mitochondrial OGG1 expression might aggravate the PDAC prognosis.
Collapse
Affiliation(s)
- Shoichi Inokuchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan.
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Kyohei Yugawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan; Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 812-8582, Japan
| | - Shohei Yoshiya
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Takeo Toshima
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Kazuki Takeishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Tomohiro Iguchi
- Department of Hepatobiliary-Pancreatic Surgery, National Hospital Organization Kyushu Cancer Center, Fukuoka, 811-1395, Japan
| | - Kensaku Sanefuji
- Department of Surgery, Oita Red Cross Hospital, 870-0033, Oita, Japan
| | - Noboru Harada
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Keishi Sugimachi
- Department of Hepatobiliary-Pancreatic Surgery, National Hospital Organization Kyushu Cancer Center, Fukuoka, 811-1395, Japan
| | - Toru Ikegami
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 812-8582, Japan
| | - Kenichi Taguchi
- Department of Pathology, National Hospital Organization Kyushu Cancer Center, Fukuoka, 811-1395, Japan
| | - Hirotoshi Yonemasu
- Department of Anatomic Pathology, Oita Red Cross Hospital, 870-0033, Oita, Japan
| | - Kengo Fukuzawa
- Department of Surgery, Oita Red Cross Hospital, 870-0033, Oita, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 812-8582, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan
| |
Collapse
|
19
|
Starcher CL, Pay SL, Singh N, Yeh IJ, Bhandare SB, Su X, Huang X, Bey EA, Motea EA, Boothman DA. Targeting Base Excision Repair in Cancer: NQO1-Bioactivatable Drugs Improve Tumor Selectivity and Reduce Treatment Toxicity Through Radiosensitization of Human Cancer. Front Oncol 2020; 10:1575. [PMID: 32974194 PMCID: PMC7468503 DOI: 10.3389/fonc.2020.01575] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/21/2020] [Indexed: 01/23/2023] Open
Abstract
Ionizing radiation (IR) creates lethal DNA damage that can effectively kill tumor cells. However, the high dose required for a therapeutic outcome also damages healthy tissue. Thus, a therapeutic strategy with predictive biomarkers to enhance the beneficial effects of IR allowing a dose reduction without losing efficacy is highly desirable. NAD(P)H:quinone oxidoreductase 1 (NQO1) is overexpressed in the majority of recalcitrant solid tumors in comparison with normal tissue. Studies have shown that NQO1 can bioactivate certain quinone molecules (e.g., ortho-naphthoquinone and β-lapachone) to induce a futile redox cycle leading to the formation of oxidative DNA damage, hyperactivation of poly(ADP-ribose) polymerase 1 (PARP1), and catastrophic depletion of NAD+ and ATP, which culminates in cellular lethality via NAD+-Keresis. However, NQO1-bioactivatable drugs induce methemoglobinemia and hemolytic anemia at high doses. To circumvent this, NQO1-bioactivatable agents have been shown to synergize with PARP1 inhibitors, pyrimidine radiosensitizers, and IR. This therapeutic strategy allows for a reduction in the dose of the combined agents to decrease unwanted side effects by increasing tumor selectivity. In this review, we discuss the mechanisms of radiosensitization between NQO1-bioactivatable drugs and IR with a focus on the involvement of base excision repair (BER). This combination therapeutic strategy presents a unique tumor-selective and minimally toxic approach for targeting solid tumors that overexpress NQO1.
Collapse
Affiliation(s)
- Colton L Starcher
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - S Louise Pay
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Naveen Singh
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - I-Ju Yeh
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Snehal B Bhandare
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaolin Su
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiumei Huang
- Department of Radiation Oncology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Erik A Bey
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Edward A Motea
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
20
|
Guo G, Gao Z, Tong M, Zhan D, Wang G, Wang Y, Qin J. NQO1 is a determinant for cellular sensitivity to anti-tumor agent Napabucasin. Am J Cancer Res 2020; 10:1442-1454. [PMID: 32509390 PMCID: PMC7269777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023] Open
Abstract
Napabucasin (NAPA) is thought to be a potent cancer stemness inhibitor in different types of cancer cell lines. While it has shown promising activity in early phase clinical trials, two recent phase III NAPA clinical trials failed to meet the primary endpoint of overall survival. The reason for the failure is not clear, but a possible way to revive the clinical trial is to stratify patients with biomarkers that could predict NAPA response. Here, we report the identification of NAD(P)H dehydrogenase 1 (NQO1) as a major determinant of NAPA efficacy. A proteomic profiling of cancer cell lines revealed that NQO1 abundance is negatively correlated with IC50; in vitro assays showed that NAPA is a substrate for NQO1, which mediates the generation of ROS that leads to cell death. Furthermore, activation of an NQO1 transcription factor NRF2 by chemicals, including an FDA approved drug, can increase the NAPA cytotoxicity. Our findings suggest a potential use of NQO1 expression as a companion diagnostic test to identify patients in future NAPA trials and a combination strategy to expand the application of NAPA-based regimens for cancer therapy.
Collapse
Affiliation(s)
- Gaigai Guo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijing 102206, China
| | - Zhouyong Gao
- Joint Center for Translational Medical Medicine, Baodi HospitalTianjin 301800, China
| | - Mengsha Tong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijing 102206, China
- School of Life Sciences, Tsinghua UniversityBeijing 100084, China
| | - Dongdong Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijing 102206, China
- Center for Bioinformatics, East China Normal UniversityShanghai 200241, China
| | - Guangshun Wang
- Joint Center for Translational Medical Medicine, Baodi HospitalTianjin 301800, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijing 102206, China
- Joint Center for Translational Medical Medicine, Baodi HospitalTianjin 301800, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of LifeomicsBeijing 102206, China
- Joint Center for Translational Medical Medicine, Baodi HospitalTianjin 301800, China
| |
Collapse
|
21
|
Mechetin GV, Endutkin AV, Diatlova EA, Zharkov DO. Inhibitors of DNA Glycosylases as Prospective Drugs. Int J Mol Sci 2020; 21:ijms21093118. [PMID: 32354123 PMCID: PMC7247160 DOI: 10.3390/ijms21093118] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/22/2022] Open
Abstract
DNA glycosylases are enzymes that initiate the base excision repair pathway, a major biochemical process that protects the genomes of all living organisms from intrinsically and environmentally inflicted damage. Recently, base excision repair inhibition proved to be a viable strategy for the therapy of tumors that have lost alternative repair pathways, such as BRCA-deficient cancers sensitive to poly(ADP-ribose)polymerase inhibition. However, drugs targeting DNA glycosylases are still in development and so far have not advanced to clinical trials. In this review, we cover the attempts to validate DNA glycosylases as suitable targets for inhibition in the pharmacological treatment of cancer, neurodegenerative diseases, chronic inflammation, bacterial and viral infections. We discuss the glycosylase inhibitors described so far and survey the advances in the assays for DNA glycosylase reactions that may be used to screen pharmacological libraries for new active compounds.
Collapse
Affiliation(s)
- Grigory V. Mechetin
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
| | - Anton V. Endutkin
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
| | - Evgeniia A. Diatlova
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
| | - Dmitry O. Zharkov
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
- Novosibirsk State University, 2 Pirogova St., 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-383-363-5187
| |
Collapse
|
22
|
Lipner MB, Peng XL, Jin C, Xu Y, Gao Y, East MP, Rashid NU, Moffitt RA, Herrera Loeza SG, Morrison AB, Golitz BT, Vaziri C, Graves LM, Johnson GL, Yeh JJ. Irreversible JNK1-JUN inhibition by JNK-IN-8 sensitizes pancreatic cancer to 5-FU/FOLFOX chemotherapy. JCI Insight 2020; 5:129905. [PMID: 32213714 DOI: 10.1172/jci.insight.129905] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Over 55,000 people in the United States are diagnosed with pancreatic ductal adenocarcinoma (PDAC) yearly, and fewer than 20% of these patients survive a year beyond diagnosis. Chemotherapies are considered or used in nearly every PDAC case, but there is limited understanding of the complex signaling responses underlying resistance to these common treatments. Here, we take an unbiased approach to study protein kinase network changes following chemotherapies in patient-derived xenograft (PDX) models of PDAC to facilitate design of rational drug combinations. Proteomics profiling following chemotherapy regimens reveals that activation of JNK-JUN signaling occurs after 5-fluorouracil plus leucovorin (5-FU + LEU) and FOLFOX (5-FU + LEU plus oxaliplatin [OX]), but not after OX alone or gemcitabine. Cell and tumor growth assays with the irreversible inhibitor JNK-IN-8 and genetic manipulations demonstrate that JNK and JUN each contribute to chemoresistance and cancer cell survival after FOLFOX. Active JNK1 and JUN are specifically implicated in these effects, and synergy with JNK-IN-8 is linked to FOLFOX-mediated JUN activation, cell cycle dysregulation, and DNA damage response. This study highlights the potential for JNK-IN-8 as a biological tool and potential combination therapy with FOLFOX in PDAC and reinforces the need to tailor treatment to functional characteristics of individual tumors.
Collapse
Affiliation(s)
- Matthew B Lipner
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | | | - Chong Jin
- Lineberger Comprehensive Cancer Center.,Department of Biostatistics
| | - Yi Xu
- Lineberger Comprehensive Cancer Center
| | - Yanzhe Gao
- Lineberger Comprehensive Cancer Center.,Department of Pathology, and
| | - Michael P East
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | - Naim U Rashid
- Lineberger Comprehensive Cancer Center.,Department of Biostatistics
| | | | | | | | | | - Cyrus Vaziri
- Lineberger Comprehensive Cancer Center.,Department of Pathology, and
| | - Lee M Graves
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | - Gary L Johnson
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | - Jen Jen Yeh
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center.,Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Torrente L, Prieto-Farigua N, Falzone A, Elkins CM, Boothman DA, Haura EB, DeNicola GM. Inhibition of TXNRD or SOD1 overcomes NRF2-mediated resistance to β-lapachone. Redox Biol 2020; 30:101440. [PMID: 32007910 PMCID: PMC6997906 DOI: 10.1016/j.redox.2020.101440] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Alterations in the NRF2/KEAP1 pathway result in the constitutive activation of NRF2, leading to the aberrant induction of antioxidant and detoxification enzymes, including NQO1. The NQO1 bioactivatable agent β-lapachone can target cells with high NQO1 expression but relies in the generation of reactive oxygen species (ROS), which are actively scavenged in cells with NRF2/KEAP1 mutations. However, whether NRF2/KEAP1 mutations influence the response to β-lapachone treatment remains unknown. To address this question, we assessed the cytotoxicity of β-lapachone in a panel of NSCLC cell lines bearing either wild-type or mutant KEAP1. We found that, despite overexpression of NQO1, KEAP1 mutant cells were resistant to β-lapachone due to enhanced detoxification of ROS, which prevented DNA damage and cell death. To evaluate whether specific inhibition of the NRF2-regulated antioxidant enzymes could abrogate resistance to β-lapachone, we systematically inhibited the four major antioxidant cellular systems using genetic and/or pharmacologic approaches. We demonstrated that inhibition of the thioredoxin-dependent system or copper-zinc superoxide dismutase (SOD1) could abrogate NRF2-mediated resistance to β-lapachone, while depletion of catalase or glutathione was ineffective. Interestingly, inhibition of SOD1 selectively sensitized KEAP1 mutant cells to β-lapachone exposure. Our results suggest that NRF2/KEAP1 mutational status might serve as a predictive biomarker for response to NQO1-bioactivatable quinones in patients. Further, our results suggest SOD1 inhibition may have potential utility in combination with other ROS inducers in patients with KEAP1/NRF2 mutations.
Collapse
Affiliation(s)
- Laura Torrente
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Nicolas Prieto-Farigua
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Aimee Falzone
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Cody M Elkins
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Simon Cancer Center Indiana, University School of Medicine, Indianapolis, IN, 46202, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
24
|
Chen J, Zhang S, Zhang S, Gao S, Wang J, Lei D, Du P, Xu Z, Zhu C, Sun H. Mesoporous Silica Nanoparticle-Based Combination of NQO1 Inhibitor and 5-Fluorouracil for Potent Antitumor Effect Against Head and Neck Squamous Cell Carcinoma (HNSCC). NANOSCALE RESEARCH LETTERS 2019; 14:387. [PMID: 31858276 PMCID: PMC6923313 DOI: 10.1186/s11671-019-3224-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/27/2019] [Indexed: 06/10/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCC) are one of the deadliest forms of cancer, and 90% of its origin is from squamous cells. NAD(P)H:quinone oxidoreductase 1 (NQO1), an enzyme overexpressed in squamous cell carcinoma, plays an important role in proliferation and chemoresistance. The main aims were to study the inhibitory effect of ß-lapachone (ARQ761 in clinical form) in HNSCC and to study the combinational effect of 5-FU and ß-lap in improving the therapeutic efficacy in HNSCC. Lipid bilayer-assembled mesoporous silica nanoparticles loaded with 5-FU/ß-lap were prepared and studied for its physicochemical and biological properties. ß-lap showed a concentration-dependent inhibition of NQO1 enzyme activity in Cal33 cells. Notably, significant inhibitory effect was observed at a dose of 20-50 μg/ml of ß-lap. Combination of 5-FU+ß-lap resulted in lower cell viability; most notably, 5-FU/ß-lap-loaded mesoporous silica nanoparticles (FNQ-MSN) exhibited significantly lower cell viability compared with that of any of the individual drug or physical combinations. ß-lap resulted in a decrease in the protein band of NQO1 compared with control; however, most notable decrease in the NQO1 level was observed in the FNQ-MSN-treated cell group. FNQ-MSN resulted in more than 60% of cell apoptosis (early and late apoptosis) and predominant nuclear fragmentation of cancer cells indicating the superior anticancer effect of a carrier-based combination regimen. Notable decrease in tumor volume was observed with the physical mixture of 5-FU+ß-lap; however, combined treatment of carrier-based 5-FU and ß-lap (FNQ-MSN) significantly delayed the tumor growth and prolonged the survival of tumor-bearing xenograft mice. These findings suggest the potential of NQO1 inhibitor in enhancing the chemotherapeutic potential of 5-FU in the treatment of HNSCC.
Collapse
Affiliation(s)
- Jing Chen
- Department of Dermatology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan China
| | - Shuzhen Zhang
- Department of Dermatology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan China
| | - Shuai Zhang
- Department of Dermatology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan China
| | - Shanjun Gao
- Microbiome Laboratory, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan China
| | - Jianbo Wang
- Department of Dermatology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan China
| | - Dongchun Lei
- Department of Dermatology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan China
| | - Pengqiang Du
- Department of Pharmacy, Henan Provincial People’s Hospital, Zhengzhou, Henan China
- Department of Pharmacy of Central China Fuwai Hospital, Zhengzhou University, Zhengzhou, Henan China
| | - Zhiwei Xu
- Clinical Research Service Center, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan China
| | - Cailiang Zhu
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou, Henan China
| | - Hongbin Sun
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou, Henan China
| |
Collapse
|
25
|
Froeling FEM, Swamynathan MM, Deschênes A, Chio IIC, Brosnan E, Yao MA, Alagesan P, Lucito M, Li J, Chang AY, Trotman LC, Belleau P, Park Y, Rogoff HA, Watson JD, Tuveson DA. Bioactivation of Napabucasin Triggers Reactive Oxygen Species-Mediated Cancer Cell Death. Clin Cancer Res 2019; 25:7162-7174. [PMID: 31527169 PMCID: PMC6891204 DOI: 10.1158/1078-0432.ccr-19-0302] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/12/2019] [Accepted: 09/11/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE Napabucasin (2-acetylfuro-1,4-naphthoquinone or BBI-608) is a small molecule currently being clinically evaluated in various cancer types. It has mostly been recognized for its ability to inhibit STAT3 signaling. However, based on its chemical structure, we hypothesized that napabucasin is a substrate for intracellular oxidoreductases and therefore may exert its anticancer effect through redox cycling, resulting in reactive oxygen species (ROS) production and cell death. EXPERIMENTAL DESIGN Binding of napabucasin to NAD(P)H:quinone oxidoreductase-1 (NQO1), and other oxidoreductases, was measured. Pancreatic cancer cell lines were treated with napabucasin, and cell survival, ROS generation, DNA damage, transcriptomic changes, and alterations in STAT3 activation were assayed in vitro and in vivo. Genetic knockout or pharmacologic inhibition with dicoumarol was used to evaluate the dependency on NQO1. RESULTS Napabucasin was found to bind with high affinity to NQO1 and to a lesser degree to cytochrome P450 oxidoreductase (POR). Treatment resulted in marked induction of ROS and DNA damage with an NQO1- and ROS-dependent decrease in STAT3 phosphorylation. Differential cytotoxic effects were observed, where NQO1-expressing cells generating cytotoxic levels of ROS at low napabucasin concentrations were more sensitive. Cells with low or no baseline NQO1 expression also produced ROS in response to napabucasin, albeit to a lesser extent, through the one-electron reductase POR. CONCLUSIONS Napabucasin is bioactivated by NQO1, and to a lesser degree by POR, resulting in futile redox cycling and ROS generation. The increased ROS levels result in DNA damage and multiple intracellular changes, one of which is a reduction in STAT3 phosphorylation.
Collapse
Affiliation(s)
- Fieke E M Froeling
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
- Northwell Cancer Institute, Lake Success, New York
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Manojit Mosur Swamynathan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York
| | - Astrid Deschênes
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Iok In Christine Chio
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
- Institute for Cancer Genetics, Columbia University, New York, New York
| | - Erin Brosnan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Melissa A Yao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
- Icahn School of Medicine at Mount Sinai, New York, New York
| | - Priya Alagesan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Matthew Lucito
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Juying Li
- Boston Biomedical Inc., Cambridge, Massachusetts
| | - An-Yun Chang
- Boston Biomedical Inc., Cambridge, Massachusetts
| | | | - Pascal Belleau
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | | | - James D Watson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| |
Collapse
|
26
|
Wu X, Chantemargue B, Di Meo F, Bourgaux C, Chapron D, Trouillas P, Rosilio V. Deciphering the Peculiar Behavior of β-Lapachone in Lipid Monolayers and Bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:14603-14615. [PMID: 31619039 DOI: 10.1021/acs.langmuir.9b02886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
β-Lapachone (β-Lap) is a promising anticancer drug whose applications have been limited so far because of its poor solubility and stability. Its encapsulation in liposomes has been proposed to overcome these issues. However, surface pressure measurements show that β-Lap exhibits atypical interfacial behavior when mixed with lipids. Although the drug does not seem to be retained in lipid monolayers as deduced from the π-A isotherms, small changes in compressibility moduli suggest that β-Lap actually interacts with lipids, either disorganizing or rigidifying their monolayers. Thermal and structural analyses of lipid bilayers confirm the existence of β-Lap/lipid interactions and show that the drug inserts between hydrophobic chains, close to the polar headgroup in DPPC bilayers and deeper in the acyl chains in POPC bilayers. Molecular dynamics simulations allow a comprehensive description of the drug position and orientation in DOPC and POPC bilayers in the presence or absence of cholesterol.
Collapse
Affiliation(s)
- Xiao Wu
- Institut Galien Paris Sud , Univ Paris-Sud, CNRS, Université Paris-Saclay , 92296 Châtenay-Malabry , France
| | - Benjamin Chantemargue
- INSERM U1248, Faculty of Pharmacy , Université de Limoges , 2 rue du Docteur Marcland , 87025 Limoges, Cedex France
- Centre of Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science , Palacký University , tr. 17 listopadu 12 , 771 46 Olomouc , Czech Republic
| | - Florent Di Meo
- INSERM U1248, Faculty of Pharmacy , Université de Limoges , 2 rue du Docteur Marcland , 87025 Limoges, Cedex France
| | - Claudie Bourgaux
- Institut Galien Paris Sud , Univ Paris-Sud, CNRS, Université Paris-Saclay , 92296 Châtenay-Malabry , France
| | - David Chapron
- Institut Galien Paris Sud , Univ Paris-Sud, CNRS, Université Paris-Saclay , 92296 Châtenay-Malabry , France
| | - Patrick Trouillas
- INSERM U1248, Faculty of Pharmacy , Université de Limoges , 2 rue du Docteur Marcland , 87025 Limoges, Cedex France
- Centre of Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science , Palacký University , tr. 17 listopadu 12 , 771 46 Olomouc , Czech Republic
| | - Véronique Rosilio
- Institut Galien Paris Sud , Univ Paris-Sud, CNRS, Université Paris-Saclay , 92296 Châtenay-Malabry , France
| |
Collapse
|
27
|
Purohit V, Simeone DM, Lyssiotis CA. Metabolic Regulation of Redox Balance in Cancer. Cancers (Basel) 2019; 11:cancers11070955. [PMID: 31288436 PMCID: PMC6678865 DOI: 10.3390/cancers11070955] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are chemically active free radicals produced by partial reduction of oxygen that can activate discrete signaling pathways or disrupt redox homeostasis depending on their concentration. ROS interacts with biomolecules, including DNA, and can cause mutations that can transform normal cells into cancer cells. Furthermore, certain cancer-causing mutations trigger alterations in cellular metabolism that can increase ROS production, resulting in genomic instability, additional DNA mutations, and tumor evolution. To prevent excess ROS-mediated toxicity, cancer-causing mutations concurrently activate pathways that manage this oxidative burden. Hence, an understanding of the metabolic pathways that regulate ROS levels is imperative for devising therapies that target tumor cells. In this review, we summarize the dual role of metabolism as a generator and inhibitor of ROS in cancer and discuss current strategies to target the ROS axis.
Collapse
Affiliation(s)
- Vinee Purohit
- Perlmutter Cancer Center, New York University, New York, NY 10016, USA
| | - Diane M Simeone
- Perlmutter Cancer Center, New York University, New York, NY 10016, USA
- Department of Surgery, New York University, New York, NY 10016, USA
- Department of Pathology, New York University, New York, NY 10016, USA
| | - Costas A Lyssiotis
- Departments of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
- Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Yang Y, Zhu G, Dong B, Piao J, Chen L, Lin Z. The NQO1/PKLR axis promotes lymph node metastasis and breast cancer progression by modulating glycolytic reprogramming. Cancer Lett 2019; 453:170-183. [PMID: 30954648 DOI: 10.1016/j.canlet.2019.03.054] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 12/23/2022]
Abstract
Overexpression of NQO1 is associated with poor prognosis in human cancers including lung, stomach, colon, cervical, and pancreatic cancers. However, the molecular mechanisms underlying the protumorigenic capacities of NQO1 have not been fully elucidated. Here, we investigated this question and determined the molecular mechanisms underlying the roles of NQO1 in glycolysis reprogramming, proliferation, and metastasis breast cancer (BC) cells. The results indicated that NQO1 overexpression in BC cells raises glucose metabolism and metastasis related behaviors. Mechanistically, NQO1 bound to PKLR, activated the AMPK and AKT/mTOR signaling pathway and consequently induced glycolytic reprogramming. In addition, 2-deoxy-d-glucose (2-DG) or 3-bromopyruvate (3-BrPA) influenced proliferation and regulated the expression of genes involved in the epithelial-to-mesenchymal transition (EMT) by restraining glycolytic reprogramming. Finally, overexpression of NQO1 and PKLR in human BC tissues was remarkably related to lymph node (LN) metastasis and poor prognosis. Together, these results demonstrate that the NQO1/PKLR axis can promote the progression of BC by modulating glycolytic reprogramming and suggest that targeting NQO1 and its downstream effectors are promising therapeutic targets for preventing the BC progression.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Guang Zhu
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Bing Dong
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Liyan Chen
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China; Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, 133002, China.
| | - Zhenhua Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| |
Collapse
|
29
|
Rai P, Sobol RW. Mechanisms of MTH1 inhibition-induced DNA strand breaks: The slippery slope from the oxidized nucleotide pool to genotoxic damage. DNA Repair (Amst) 2019; 77:18-26. [PMID: 30852368 DOI: 10.1016/j.dnarep.2019.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Unlike normal tissues, tumor cells possess a propensity for genomic instability, resulting from elevated oxidant levels produced by oncogenic signaling and aberrant cellular metabolism. Thus, targeting mechanisms that protect cancer cells from the tumor-inhibitory consequences of their redox imbalance and spontaneous DNA-damaging events is expected to have broad-spectrum efficacy and a high therapeutic index. One critical mechanism for tumor cell protection from oxidant stress is the hydrolysis of oxidized nucleotides. Human MutT homolog 1 (MTH1), the mammalian nudix (nucleoside diphosphate X) pyrophosphatase (NUDT1), protects tumor cells from oxidative stress-induced genomic DNA damage by cleansing the nucleotide pool of oxidized purine nucleotides. Depletion or pharmacologic inhibition of MTH1 results in genomic DNA strand breaks in many cancer cells. However, the mechanisms underlying how oxidized nucleotides, thought mainly to be mutagenic rather than genotoxic, induce DNA strand breaks are largely unknown. Given the recent therapeutic interest in targeting MTH1, a better understanding of such mechanisms is crucial to its successful translation into the clinic and in identifying the molecular contexts under which its inhibition is likely to be beneficial. Here we provide a comprehensive perspective on MTH1 function and its importance in protecting genome integrity, in the context of tumor-associated oxidative stress and the mechanisms that likely lead to irreparable DNA strand breaks as a result of MTH1 inhibition.
Collapse
Affiliation(s)
- Priyamvada Rai
- Department of Medicine/Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, United States; Sylvester Comprehensive Cancer Center, Miami, FL, 33136, United States.
| | - Robert W Sobol
- Mitchell Cancer Institute, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, United States.
| |
Collapse
|
30
|
SANIE-JAHROMI F, MAHMOUDINASAB H, SAADAT M. Effects of β-Lapachone at Non-Toxic and Toxic Concentrations on the mRNA Levels of XRCC1, GADD45A and LIG4 Genes. IRANIAN JOURNAL OF PUBLIC HEALTH 2019; 48:559-560. [PMID: 31223588 PMCID: PMC6570797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 02/24/2018] [Indexed: 11/18/2022]
Affiliation(s)
| | | | - Mostafa SAADAT
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| |
Collapse
|
31
|
Motea EA, Huang X, Singh N, Kilgore JA, Williams NS, Xie XJ, Gerber DE, Beg MS, Bey EA, Boothman DA. NQO1-dependent, Tumor-selective Radiosensitization of Non-small Cell Lung Cancers. Clin Cancer Res 2019; 25:2601-2609. [PMID: 30617135 DOI: 10.1158/1078-0432.ccr-18-2560] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/30/2018] [Accepted: 01/04/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Development of tumor-specific therapies for the treatment of recalcitrant non-small cell lung cancers (NSCLC) is urgently needed. Here, we investigated the ability of β-lapachone (β-lap, ARQ761 in clinical form) to selectively potentiate the effects of ionizing radiation (IR, 1-3 Gy) in NSCLCs that overexpress NAD(P)H:Quinone Oxidoreductase 1 (NQO1). EXPERIMENTAL DESIGN The mechanism of lethality of low-dose IR in combination with sublethal doses of β-lap was evaluated in NSCLC lines in vitro and validated in subcutaneous and orthotopic xenograft models in vivo. Pharmacokinetics and pharmacodynamics (PK/PD) studies comparing single versus cotreatments were performed to validate therapeutic efficacy and mechanism of action. RESULTS β-Lap administration after IR treatment hyperactivated PARP, greatly lowered NAD+/ATP levels, and increased double-strand break (DSB) lesions over time in vitro. Radiosensitization of orthotopic, as well as subcutaneous, NSCLCs occurred with high apparent cures (>70%), even though 1/8 β-lap doses reach subcutaneous versus orthotopic tumors. No methemoglobinemia or long-term toxicities were noted in any normal tissues, including mouse liver that expresses the highest level of NQO1 (∼12 units) of any normal tissue. PK/PD responses confirm that IR + β-lap treatments hyperactivate PARP activity, greatly lower NAD+/ATP levels, and dramatically inhibit DSB repair in exposed NQO1+ cancer tissue, whereas low NQO1 levels and high levels of catalase in associated normal tissue were protective. CONCLUSIONS Our data suggest that combination of sublethal doses of β-lap and IR is a viable approach to selectively treat NQO1-overexpressing NSCLC and warrant a clinical trial using low-dose IR + β-lap against patients with NQO1+ NSCLCs.
Collapse
Affiliation(s)
- Edward A Motea
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiumei Huang
- Department of Radiation Oncology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Naveen Singh
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica A Kilgore
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Noelle S Williams
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xian-Jin Xie
- Department of Biostatistics, UT Southwestern Medical Center, Dallas, Texas
| | - David E Gerber
- Department of Internal Medicine, Division of Hematology-Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Muhammad S Beg
- Department of Internal Medicine, Division of Hematology-Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Erik A Bey
- Department of Pharmaceutical Sciences, West Virginia University Cancer Institute, Morgantown, West Virginia.
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
32
|
Chen X, Mims J, Huang X, Singh N, Motea E, Planchon SM, Beg M, Tsang AW, Porosnicu M, Kemp ML, Boothman DA, Furdui CM. Modulators of Redox Metabolism in Head and Neck Cancer. Antioxid Redox Signal 2018; 29:1660-1690. [PMID: 29113454 PMCID: PMC6207163 DOI: 10.1089/ars.2017.7423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Head and neck squamous cell cancer (HNSCC) is a complex disease characterized by high genetic and metabolic heterogeneity. Radiation therapy (RT) alone or combined with systemic chemotherapy is widely used for treatment of HNSCC as definitive treatment or as adjuvant treatment after surgery. Antibodies against epidermal growth factor receptor are used in definitive or palliative treatment. Recent Advances: Emerging targeted therapies against other proteins of interest as well as programmed cell death protein 1 and programmed death-ligand 1 immunotherapies are being explored in clinical trials. CRITICAL ISSUES The disease heterogeneity, invasiveness, and resistance to standard of care RT or chemoradiation therapy continue to constitute significant roadblocks for treatment and patients' quality of life (QOL) despite improvements in treatment modality and the emergence of new therapies over the past two decades. FUTURE DIRECTIONS As reviewed here, alterations in redox metabolism occur at all stages of HNSCC management, providing opportunities for improved prevention, early detection, response to therapies, and QOL. Bioinformatics and computational systems biology approaches are key to integrate redox effects with multiomics data from cells and clinical specimens and to identify redox modifiers or modifiable target proteins to achieve improved clinical outcomes. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Xiaofei Chen
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jade Mims
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Xiumei Huang
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Naveen Singh
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Edward Motea
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | | | - Muhammad Beg
- Department of Internal Medicine, Division of Hematology-Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Allen W. Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mercedes Porosnicu
- Department of Internal Medicine, Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Melissa L. Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - David A. Boothman
- Departments of Pharmacology, Radiation Oncology, and Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
33
|
Gerber DE, Beg MS, Fattah F, Frankel AE, Fatunde O, Arriaga Y, Dowell JE, Bisen A, Leff RD, Meek CC, Putnam WC, Kallem RR, Subramaniyan I, Dong Y, Bolluyt J, Sarode V, Luo X, Xie Y, Schwartz B, Boothman DA. Phase 1 study of ARQ 761, a β-lapachone analogue that promotes NQO1-mediated programmed cancer cell necrosis. Br J Cancer 2018; 119:928-936. [PMID: 30318513 PMCID: PMC6203852 DOI: 10.1038/s41416-018-0278-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/23/2018] [Accepted: 09/04/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND NAD(P)H:quinone oxidoreductase 1 (NQO1) is a two-electron oxidoreductase expressed in multiple tumour types. ARQ 761 is a β-lapachone (β-lap) analogue that exploits the unique elevation of NQO1 found in solid tumours to cause tumour-specific cell death. METHODS We performed a 3+3 dose escalation study of 3 schedules (weekly, every other week, 2/3 weeks) of ARQ 761 in patients with refractory advanced solid tumours. Tumour tissue was analysed for NQO1 expression. After 20 patients were analysed, enrolment was restricted to patients with NQO1-high tumours (H-score ≥ 200). RESULTS A total of 42 patients were treated. Median number of prior lines of therapy was 4. Maximum tolerated dose was 390 mg/m2 as a 2-h infusion every other week. Dose-limiting toxicity was anaemia. The most common treatment-related adverse events were anaemia (79%), fatigue (45%), hypoxia (33%), nausea (17%), and vomiting (17%). Transient grade 3 hypoxia, reflecting possible methemoglobinaemia, occurred in 26% of patients. Among 32 evaluable patients, best response was stable disease (n = 12); 6 patients had tumour shrinkage. There was a trend towards improved efficacy in NQO1-high tumours (P = 0.06). CONCLUSIONS ARQ 761 has modest single-agent activity, which appears associated with tumour NQO1 expression. Principal toxicities include anaemia and possible methemoglobinaemia.
Collapse
Affiliation(s)
- David E Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - M Shaalan Beg
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Farjana Fattah
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Arthur E Frankel
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Oluwatomilade Fatunde
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yull Arriaga
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jonathan E Dowell
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ajit Bisen
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Richard D Leff
- Texas Tech University Health Sciences Center School of Pharmacy, Dallas, TX, 75390, USA
| | - Claudia C Meek
- Texas Tech University Health Sciences Center School of Pharmacy, Dallas, TX, 75390, USA
| | - William C Putnam
- Texas Tech University Health Sciences Center School of Pharmacy, Dallas, TX, 75390, USA
| | - Raja Reddy Kallem
- Texas Tech University Health Sciences Center School of Pharmacy, Dallas, TX, 75390, USA
| | | | - Ying Dong
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Joyce Bolluyt
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Venetia Sarode
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xin Luo
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yang Xie
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | | | - David A Boothman
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
34
|
Lewis JE, Costantini F, Mims J, Chen X, Furdui CM, Boothman DA, Kemp ML. Genome-Scale Modeling of NADPH-Driven β-Lapachone Sensitization in Head and Neck Squamous Cell Carcinoma. Antioxid Redox Signal 2018; 29:937-952. [PMID: 28762750 PMCID: PMC6104251 DOI: 10.1089/ars.2017.7048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS The purpose of this study was to investigate differential nicotinamide adenine dinucleotide phosphate, reduced (NADPH) production between radiation-sensitive and -resistant head and neck squamous cell carcinoma (HNSCC) cell lines and whether these differences are predictive of sensitivity to the chemotherapeutic β-lapachone. RESULTS We have developed a novel human genome-scale metabolic modeling platform that combines transcriptomic, kinetic, thermodynamic, and metabolite concentration data. Upon incorporation of this information into cell line-specific models, we observed that the radiation-resistant HNSCC model redistributed flux through several major NADPH-producing reactions. Upon RNA interference of canonical NADPH-producing genes, the metabolic network can further reroute flux through alternate NADPH biosynthesis pathways in a cell line-specific manner. Model predictions of perturbations in cellular NADPH production after gene knockdown match well with experimentally verified effects of β-lapachone treatment on NADPH/NADP+ ratio and cell viability. This computational approach accurately predicts HNSCC-specific oxidoreductase genes that differentially affect cell viability between radiation-responsive and radiation-resistant cancer cells upon β-lapachone treatment. INNOVATION Quantitative genome-scale metabolic models that incorporate multiple levels of biological data are applied to provide accurate predictions of responses to a NADPH-dependent redox cycling chemotherapeutic drug under a variety of perturbations. CONCLUSION Our modeling approach suggests differences in metabolism and β-lapachone redox cycling that underlie phenotypic differences in radiation-sensitive and -resistant cancer cells. This approach can be extended to investigate the synergistic action of NAD(P)H: quinone oxidoreductase 1 bioactivatable drugs and radiation therapy. Antioxid. Redox Signal. 29, 937-952.
Collapse
Affiliation(s)
- Joshua E Lewis
- 1 The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| | - Francesco Costantini
- 2 School of Chemical and Biomolecular Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Jade Mims
- 3 Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Xiaofei Chen
- 3 Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - Cristina M Furdui
- 3 Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | - David A Boothman
- 4 Department of Pharmacology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Melissa L Kemp
- 1 The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| |
Collapse
|
35
|
Van Sciver RE, Lee MP, Lee CD, Lafever AC, Svyatova E, Kanda K, Colliver AL, Siewertsz van Reesema LL, Tang-Tan AM, Zheleva V, Bwayi MN, Bian M, Schmidt RL, Matrisian LM, Petersen GM, Tang AH. A New Strategy to Control and Eradicate "Undruggable" Oncogenic K-RAS-Driven Pancreatic Cancer: Molecular Insights and Core Principles Learned from Developmental and Evolutionary Biology. Cancers (Basel) 2018; 10:142. [PMID: 29757973 PMCID: PMC5977115 DOI: 10.3390/cancers10050142] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022] Open
Abstract
Oncogenic K-RAS mutations are found in virtually all pancreatic cancers, making K-RAS one of the most targeted oncoproteins for drug development in cancer therapies. Despite intense research efforts over the past three decades, oncogenic K-RAS has remained largely "undruggable". Rather than targeting an upstream component of the RAS signaling pathway (i.e., EGFR/HER2) and/or the midstream effector kinases (i.e., RAF/MEK/ERK/PI3K/mTOR), we propose an alternative strategy to control oncogenic K-RAS signal by targeting its most downstream signaling module, Seven-In-Absentia Homolog (SIAH). SIAH E3 ligase controls the signal output of oncogenic K-RAS hyperactivation that drives unchecked cell proliferation, uncontrolled tumor growth, and rapid cancer cell dissemination in human pancreatic cancer. Therefore, SIAH is an ideal therapeutic target as it is an extraordinarily conserved downstream signaling gatekeeper indispensable for proper RAS signaling. Guided by molecular insights and core principles obtained from developmental and evolutionary biology, we propose an anti-SIAH-centered anti-K-RAS strategy as a logical and alternative anticancer strategy to dampen uncontrolled K-RAS hyperactivation and halt tumor growth and metastasis in pancreatic cancer. The clinical utility of developing SIAH as both a tumor-specific and therapy-responsive biomarker, as well as a viable anti-K-RAS drug target, is logically simple and conceptually innovative. SIAH clearly constitutes a major tumor vulnerability and K-RAS signaling bottleneck in pancreatic ductal adenocarcinoma (PDAC). Given the high degree of evolutionary conservation in the K-RAS/SIAH signaling pathway, an anti-SIAH-based anti-PDAC therapy will synergize with covalent K-RAS inhibitors and direct K-RAS targeted initiatives to control and eradicate pancreatic cancer in the future.
Collapse
Affiliation(s)
- Robert E Van Sciver
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Michael P Lee
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Caroline Dasom Lee
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Alex C Lafever
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Elizaveta Svyatova
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Kevin Kanda
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Amber L Colliver
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | | | - Angela M Tang-Tan
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA.
| | - Vasilena Zheleva
- Department of Surgery, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Monicah N Bwayi
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Minglei Bian
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| | - Rebecca L Schmidt
- Department of Biology, Upper Iowa University, Fayette, IA 52142, USA.
| | - Lynn M Matrisian
- Pancreatic Cancer Action Network, 1050 Connecticut Ave NW, Suite 500, Washington, DC 20036, USA.
- Pancreatic Cancer Action Network, 1500 Rosecrans Ave, Suite 200, Manhattan Beach, CA 90266, USA.
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic Cancer Center, Mayo Clinic Pancreatic Cancer SPORE, BioBusiness 5-85, 200 First Street SW, Rochester, MN 55905, USA.
| | - Amy H Tang
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| |
Collapse
|
36
|
Candido ACL, da Silva MPG, da Silva EG, de Abreu FC. Electrochemical and spectroscopic characterization of the interaction between β-lapachone and PAMAM derivatives immobilized on surface electrodes. J Solid State Electrochem 2018. [DOI: 10.1007/s10008-018-3880-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
37
|
Zhang K, Chen D, Ma K, Wu X, Hao H, Jiang S. NAD(P)H:Quinone Oxidoreductase 1 (NQO1) as a Therapeutic and Diagnostic Target in Cancer. J Med Chem 2018; 61:6983-7003. [DOI: 10.1021/acs.jmedchem.8b00124] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kuojun Zhang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Dong Chen
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kun Ma
- Center for Drug Evaluation, China Food and Drug Administration, Beijing 100038, China
| | - Xiaoxing Wu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sheng Jiang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
38
|
Targeting of stress response pathways in the prevention and treatment of cancer. Biotechnol Adv 2018; 36:583-602. [PMID: 29339119 DOI: 10.1016/j.biotechadv.2018.01.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
The hallmarks of tumor tissue are not only genetic aberrations but also the presence of metabolic and oxidative stress as a result of hypoxia and lactic acidosis. The stress activates several prosurvival pathways including metabolic remodeling, autophagy, antioxidant response, mitohormesis, and glutaminolysis, whose upregulation in tumors is associated with a poor survival of patients, while their activation in healthy tissue with statins, metformin, physical activity, and natural compounds prevents carcinogenesis. This review emphasizes the dual role of stress response pathways in cancer and suggests the integrative understanding as a basis for the development of rational therapy targeting the stress response.
Collapse
|
39
|
Arakawa N, Okubo A, Yasuhira S, Takahashi K, Amano H, Akasaka T, Masuda T, Shibazaki M, Maesawa C. Carnosic acid, an inducer of NAD(P)H quinone oxidoreductase 1, enhances the cytotoxicity of β-lapachone in melanoma cell lines. Oncol Lett 2017; 15:2393-2400. [PMID: 29434949 DOI: 10.3892/ol.2017.7618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 11/02/2017] [Indexed: 01/06/2023] Open
Abstract
NAD(P)H quinone oxidoreductase 1 (NQO1)-dependent antitumor drugs such as β-lapachone (β-lap) are attractive candidates for cancer chemotherapy because several tumors exhibit higher expression of NQO1 than adjacent tissues. Although the association between NQO1 and β-lap has been elucidated, the effects of a NQO1-inducer and β-lap used in combination remain to be clarified. It has previously been reported that melanoma cell lines have detectable levels of NQO1 expression and are sensitive to NQO1-dependent drugs such as 17-allylamino-17-demethoxygeldanamycin. The present study was conducted to investigate the involvement of NQO1 in β-lap-mediated toxicity and the utility of combination treatment with a NQO1-inducer and β-lap in malignant melanoma cell lines. Decreased expression or inhibition of NQO1 caused these cell lines to become less sensitive to β-lap, indicating a requirement of NQO1 activity for β-lap-mediated toxicity. Of note was that carnosic acid (CA), a compound extracted from rosemary, was able to induce further expression of NQO1 through NF-E2 related factor 2 (NRF2) stabilization, thus significantly enhancing the cytotoxicity of β-lap in all of the melanoma cell lines tested. Taken together, the data presented in the current study indicated that the NRF2-NQO1 axis may have potential value as a therapeutic target in malignant melanoma to improve the rate of clinical response to NQO1-dependent antitumor drugs.
Collapse
Affiliation(s)
- Nobuyuki Arakawa
- Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, Iwate 028-8505, Japan
| | - Ayaka Okubo
- Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, Iwate 028-8505, Japan
| | - Shinji Yasuhira
- Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, Iwate 028-8505, Japan
| | - Kazuhiro Takahashi
- Department of Dermatology, Iwate Medical University, Iwate 028-3694, Japan
| | - Hiroo Amano
- Department of Dermatology, Iwate Medical University, Iwate 028-3694, Japan
| | - Toshihide Akasaka
- Division of Dermatology, Kitakami Saiseikai Hospital, Iwate 024-8506, Japan
| | - Tomoyuki Masuda
- Department of Pathology, School of Medicine, Iwate Medical University, Iwate 028-3694, Japan
| | - Masahiko Shibazaki
- Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, Iwate 028-8505, Japan
| | - Chihaya Maesawa
- Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, Iwate 028-8505, Japan
| |
Collapse
|
40
|
Silvers MA, Deja S, Singh N, Egnatchik RA, Sudderth J, Luo X, Beg MS, Burgess SC, DeBerardinis RJ, Boothman DA, Merritt ME. The NQO1 bioactivatable drug, β-lapachone, alters the redox state of NQO1+ pancreatic cancer cells, causing perturbation in central carbon metabolism. J Biol Chem 2017; 292:18203-18216. [PMID: 28916726 DOI: 10.1074/jbc.m117.813923] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/13/2017] [Indexed: 12/21/2022] Open
Abstract
Many cancer treatments, such as those for managing recalcitrant tumors like pancreatic ductal adenocarcinoma, cause off-target toxicities in normal, healthy tissue, highlighting the need for more tumor-selective chemotherapies. β-Lapachone is bioactivated by NAD(P)H:quinone oxidoreductase 1 (NQO1). This enzyme exhibits elevated expression in most solid cancers and therefore is a potential cancer-specific target. β-Lapachone's therapeutic efficacy partially stems from the drug's induction of a futile NQO1-mediated redox cycle that causes high levels of superoxide and then peroxide formation, which damages DNA and causes hyperactivation of poly(ADP-ribose) polymerase, resulting in extensive NAD+/ATP depletion. However, the effects of this drug on energy metabolism due to NAD+ depletion were never described. The futile redox cycle rapidly consumes O2, rendering standard assays of Krebs cycle turnover unusable. In this study, a multimodal analysis, including metabolic imaging using hyperpolarized pyruvate, points to reduced oxidative flux due to NAD+ depletion after β-lapachone treatment of NQO1+ human pancreatic cancer cells. NAD+-sensitive pathways, such as glycolysis, flux through lactate dehydrogenase, and the citric acid cycle (as inferred by flux through pyruvate dehydrogenase), were down-regulated by β-lapachone treatment. Changes in flux through these pathways should generate biomarkers useful for in vivo dose responses of β-lapachone treatment in humans, avoiding toxic side effects. Targeting the enzymes in these pathways for therapeutic treatment may have the potential to synergize with β-lapachone treatment, creating unique NQO1-selective combinatorial therapies for specific cancers. These findings warrant future studies of intermediary metabolism in patients treated with β-lapachone.
Collapse
Affiliation(s)
- Molly A Silvers
- From the Departments of Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center
| | - Stanislaw Deja
- Advanced Imaging Research Center (AIRC), Division of Metabolic Mechanisms of Disease
| | - Naveen Singh
- From the Departments of Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center
| | - Robert A Egnatchik
- the Children's Medical Center Research Institute, Simmons Comprehensive Cancer Center
| | - Jessica Sudderth
- the Children's Medical Center Research Institute, Simmons Comprehensive Cancer Center
| | - Xiuquan Luo
- From the Departments of Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center
| | | | - Shawn C Burgess
- Advanced Imaging Research Center (AIRC), Division of Metabolic Mechanisms of Disease
| | - Ralph J DeBerardinis
- the Children's Medical Center Research Institute, Simmons Comprehensive Cancer Center
| | - David A Boothman
- From the Departments of Pharmacology and Radiation Oncology, Simmons Comprehensive Cancer Center,
| | - Matthew E Merritt
- the AIRC, University of Texas Southwestern Medical Center, Dallas, Texas 75390 and .,the Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
41
|
Espindola-Netto JM, Chini CCS, Tarragó M, Wang E, Dutta S, Pal K, Mukhopadhyay D, Sola-Penna M, Chini EN. Preclinical efficacy of the novel competitive NAMPT inhibitor STF-118804 in pancreatic cancer. Oncotarget 2017; 8:85054-85067. [PMID: 29156703 PMCID: PMC5689593 DOI: 10.18632/oncotarget.18841] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 06/05/2017] [Indexed: 11/25/2022] Open
Abstract
NAD salvage is one of the pathways used to generate NAD in mammals. Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in this pathway, uses nicotinamide (NAM) to generate nicotinamide mononucleotide (NMN). NMN is one of the main precursors of NAD synthesis in cells. Our previous study showed the importance of NAMPT in maintaining NAD levels in pancreatic ductal adenocarcinoma cells (PDAC), and that the NAMPT inhibitor FK866 decreased pancreatic cancer growth. We now tested the effect of STF-118804, a new highly specific NAMPT inhibitor, in models of pancreatic ductal adenocarcinoma. STF-118804 reduced viability and growth of different PDAC lines, as well as the formation of colonies in soft agar. In addition, STF-118804 decreased glucose uptake, lactate excretion, and ATP levels, resulting in metabolic collapse. STF-118804 treatment activated AMPK and inhibited of mTOR pathways in these cells. This effect was significantly potentiated by pharmacological AMPK activation and mTOR inhibition. Exogenous NMN blocked both the activation of the AMPK pathway and the decrease in cell viability. Panc-1 cells expressing GFP-luciferase were orthotopically implanted on mice pancreas to test the in vivo effectiveness of STF-118804. Both STF-118804 and FK866 reduced tumor size after 21 days of treatment. Combinations of STF-118804 with chemotherapeutic agents such as paclitaxel, gemcitabine, and etoposide showed an additive effect in decreasing cell viability and growth. In conclusion, our preclinical study shows that the NAMPT inhibitor STF-118804 reduced the growth of PDAC in vitro and in vivo and had an additive effect in combination with main current chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jair Machado Espindola-Netto
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A.,Laboratório de Enzimologia e Controle do Metabolismo (LabECoM), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmacia, Centro de Ciencias da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia C S Chini
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A
| | - Mariana Tarragó
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmacia, Centro de Ciencias da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo N Chini
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A
| |
Collapse
|
42
|
Yang Y, Zhou X, Xu M, Piao J, Zhang Y, Lin Z, Chen L. β-lapachone suppresses tumour progression by inhibiting epithelial-to-mesenchymal transition in NQO1-positive breast cancers. Sci Rep 2017; 7:2681. [PMID: 28578385 PMCID: PMC5457413 DOI: 10.1038/s41598-017-02937-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/20/2017] [Indexed: 01/28/2023] Open
Abstract
NQO1 is a FAD-binding protein that can form homodimers and reduce quinones to hydroquinones, and a growing body of evidence currently suggests that NQO1 is dramatically elevated in solid cancers. Here, we demonstrated that NQO1 was elevated in breast cancer and that its expression level was positively correlated with invasion and reduced disease free survival (DFS) and overall survival (OS) rates. Next, we found that β-lapachone exerted significant anti-proliferation and anti-metastasis effects in breast cancer cell lines due to its effects on NQO1 expression. Moreover, we revealed that the anti-cancer effects of β-lapachone were mediated by the inactivation of the Akt/mTOR pathway. In conclusion, these results demonstrated that NQO1 could be a useful prognostic biomarker for patients with breast cancer, and its bioactivatable drug, β-lapachone represented a promising new development and an effective strategy for indicating the progression of NQO1-positive breast cancers.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Xianchun Zhou
- Department of Internal Medicine, Yanbian University Hospital, Yanji, 133000, China
| | - Ming Xu
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.,Department of Internal Medicine, Yanbian University Hospital, Yanji, 133000, China
| | - Yuan Zhang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China
| | - Zhenhua Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| | - Liyan Chen
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, 133002, China.
| |
Collapse
|
43
|
Furuse J, Nagashima F. Emerging protein kinase inhibitors for treating pancreatic cancer. Expert Opin Emerg Drugs 2017; 22:77-86. [PMID: 28253828 DOI: 10.1080/14728214.2017.1293648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Pancreatic cancer, the incidence and mortality of which are increasing around the world, has the most dismal prognosis among the commonly encountered cancers. Systemic chemotherapy plays an important role in the treatment of patients with pancreatic cancer, and development of more effective chemotherapies is being sought. Areas covered: This review article provides a summary about protein kinase inhibitors that have been investigated for the treatment of pancreatic cancer, not only existing agents targeting RAS, EGFR, VEGFR, MEK, etc., but also various compounds targeting, including the MAPK, PI3 K/Akt/mTOR, and JAK/STAT signaling pathways, trials of which are currently ongoing. To date, none has shown sufficient efficacy as to merit becoming established as a standard treatment agent for pancreatic cancer. Expert opinion: As the toxicities of protein kinase inhibitors usually differ from those of cytotoxic agents, it could be of value to use these agents in combination with gemcitabine plus nab-paclitaxel. It may be reasonable to identify a suitable disease and/or predictive markers for new compounds in proof of concept trials. It is an urgent need to conduct phase III trials, on the basis of the results obtained, in subpopulations with biomarkers to predict the efficacy of these drugs.
Collapse
Affiliation(s)
- Junji Furuse
- a Department of Medical Oncology , Kyorin University, Faculty of Medicine , Tokyo , Japan
| | - Fumio Nagashima
- a Department of Medical Oncology , Kyorin University, Faculty of Medicine , Tokyo , Japan
| |
Collapse
|
44
|
Kolossov VL, Ponnuraj N, Beaudoin JN, Leslie MT, Kenis PJ, Gaskins HR. Distinct responses of compartmentalized glutathione redox potentials to pharmacologic quinones targeting NQO1. Biochem Biophys Res Commun 2017; 483:680-686. [PMID: 27986568 PMCID: PMC5253246 DOI: 10.1016/j.bbrc.2016.12.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 12/29/2022]
Abstract
Deoxynyboquinone (DNQ), a potent novel quinone-based antineoplastic agent, selectively kills solid cancers with overexpressed cytosolic NAD(P)H:quinone oxidoreductase-1 (NQO1) via excessive ROS production. A genetically encoded redox-sensitive probe was used to monitor intraorganellar glutathione redox potentials (EGSH) as a direct indicator of cellular oxidative stress following chemotherapeutic administration. Beta-lapachone (β-lap) and DNQ-induced spatiotemporal redox responses were monitored in human lung A549 and pancreatic MIA-PaCa-2 adenocarcinoma cells incubated with or without dicumarol and ES936, potent NQO1 inhibitors. Immediate oxidation of EGSH in both the cytosol and mitochondrial matrix was observed in response to DNQ and β-lap. The DNQ-induced cytosolic oxidation was fully prevented with NQO1 inhibition, whereas mitochondrial oxidation in A549 was NQO1-independent in contrast to MIA-PaCa-2 cells. However, at pharmacologic concentrations of β-lap both quinone-based substrates directly oxidized the redox probe, a possible sign of off-target reactivity with cellular thiols. Together, these data provide new evidence that DNQ's direct and discerning NQO1 substrate specificity underlies its pharmacologic potency, while β-lap elicits off-target responses at its effective doses.
Collapse
Affiliation(s)
- Vladimir L Kolossov
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States.
| | - Nagendraprabhu Ponnuraj
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Jessica N Beaudoin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Matthew T Leslie
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Paul J Kenis
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - H Rex Gaskins
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| |
Collapse
|
45
|
Nukuzuma S, Nakamichi K, Kameoka M, Sugiura S, Nukuzuma C, Tasaki T, Takegami T. Suppressive effect of topoisomerase inhibitors on JC polyomavirus propagation in human neuroblastoma cells. Microbiol Immunol 2017; 60:253-60. [PMID: 26935240 DOI: 10.1111/1348-0421.12372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 11/28/2022]
Abstract
JC polyomavirus (JCPyV) causes progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease of the central nervous system, in immunocompromised patients. Because no drugs have been approved for treating PML, many antiviral agents are currently being investigated for this purpose. The inhibitory effects of the topoisomerase I inhibitors topotecan and β-lapachone were assessed by investigating viral replication, propagation and viral protein 1 (VP1) production in cultured cells. JCPyV replication was assayed using the human neuroblastoma cell line IMR-32 transfected with the JCPyV plasmid and RT- PCR combined with Dpn I treatment. Dpn I digests the input plasmid DNA containing methylated adenosine, but not newly replicated JCPyV DNA, in IMR-32 cells. It was found that JCPyV replicates less in IMR-32 cells treated with topotecan or β-lapachone than in untreated cells. Moreover, drug treatment of JCI cells, which are IMR-32 cells persistently infected with JCPyV, led to a reduction in the amount of JCPyV DNA and population of VP1-positive cells. These results demonstrate that topotecan and β-lapachone affects JCPyV propagation in human neuroblastoma cell lines, suggesting that topotecan and β-lapachone could potentially be used to treat PML.
Collapse
Affiliation(s)
- Souichi Nukuzuma
- Department of Infectious Diseases, Kobe Institute of Health, 4-6-5, Minatojima-Nakamachi, Chuo-ku, Kobe 650-0046
| | - Kazuo Nakamichi
- Department of Virology 1, National Institute of Infectious Diseases, Toyama, Shinjuku, Tokyo 162-8640
| | - Masanori Kameoka
- Department of International Health, Kobe University Graduate School of Health Sciences, Suma-ku, Kobe 615-0124
| | - Shigeki Sugiura
- Medical Genetics Research Center, Nara Medical University, Kashihara, Nara 634-8521
| | | | - Takafumi Tasaki
- Divison of Protein Regulation Research, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293
| | - Tsutomu Takegami
- Division of Molecular Oncology and Virology, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| |
Collapse
|
46
|
Zhou Y, Dong Y, Huang G, Wang Y, Huang X, Zhang F, Boothman DA, Gao J, Liang W. Lysosome-oriented, dual-stage pH-responsive polymeric micelles for β-Lapachone delivery. J Mater Chem B 2016; 4:7429-7440. [PMID: 28580145 PMCID: PMC5452003 DOI: 10.1039/c6tb02049f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
β-Lapachone (β-lap), a novel anticancer agent, is bioactivated by NADP(H):quinone oxidoreductase 1 (NQO1), an enzyme over-expressed in numerous tumors, including lung, pancreas, breast, and prostate cancers. Fast renal clearance and methemaglobinemia / hemolytic side-effects from the clinical formulation (β-lap-hydroxyl propyl-β-cyclodextrin complex) hindered its clinical translation. Here, we investigated a dual model pH responsive polymers for β-lap delivery. Three pH-sensitive linkages, including acylhydrazone, ketal and imine bonds for β-lap prodrug syntheses result in an aryl imine linkage the most optimal linkage. The conversion to β-lap was 2.8%, 4.5% and 100% at pH 7.4, 6.5 and 5.0 in 8 h, respectively. β-lap aryl imine prodrug conjugated ultra pH-sensitive (UPS) polymer reached high β-lap loading density (8.3%) and exhibited dual-stages responsiveness to pH variation. In pHs under pHt, at stage I, micelle immediately dissociation and subsequently entering stage II, micelles start quickly release β-lap. In vitro release study showed that the micelles constantly release β-lap (14.9 ± 0.1%) at pHs above pHt in 72 h, whereas boosted release of β-lap (79.4 ± 1.2%) at pH 5.0. Micelle intracellular distribution predominantly in the lysosome organelle guaranteed their pH responsive dissociation and subsequently β-lap controlled release. The M-P micelles retained NQO1-dependent cytotoxicity in A549 lung cancer cells, similar to free drug in both efficacy and mechanism of cell death. The lysosome-oriented dual-stage ultra pH responsive β-lap prodrug micelles potentially offer an alternative nanotherapeutic strategy for lung, as well as other NQO1+ cancer therapies.
Collapse
Affiliation(s)
- Yinjian Zhou
- Protein and Peptide Pharmaceutical Laboratory, Institute of
Biophysics, Chinese Academy of Sciences, Beijing, China
- Department of Pharmacology, Harold C. Simmons Comprehensive
Cancer Center, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd,
Dallas, Texas 75390
| | - Ying Dong
- Laboratory of Molecular Stresses, Departments of
Pharmacology and Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center,
UT Southwestern Medical Center at Dallas, 6001 Forest Park Drive, ND2.210K Texas
75390-8807
| | - Gang Huang
- Department of Pharmacology, Harold C. Simmons Comprehensive
Cancer Center, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd,
Dallas, Texas 75390
| | - Yiguang Wang
- Department of Pharmacology, Harold C. Simmons Comprehensive
Cancer Center, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd,
Dallas, Texas 75390
| | - Xiaonan Huang
- Department of Pharmacology, Harold C. Simmons Comprehensive
Cancer Center, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd,
Dallas, Texas 75390
| | - Fayun Zhang
- Protein and Peptide Pharmaceutical Laboratory, Institute of
Biophysics, Chinese Academy of Sciences, Beijing, China
| | - David A. Boothman
- Laboratory of Molecular Stresses, Departments of
Pharmacology and Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center,
UT Southwestern Medical Center at Dallas, 6001 Forest Park Drive, ND2.210K Texas
75390-8807
| | - Jinming Gao
- Department of Pharmacology, Harold C. Simmons Comprehensive
Cancer Center, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd,
Dallas, Texas 75390
| | - Wei Liang
- Protein and Peptide Pharmaceutical Laboratory, Institute of
Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
47
|
Durand N, Storz P. Targeting reactive oxygen species in development and progression of pancreatic cancer. Expert Rev Anticancer Ther 2016; 17:19-31. [PMID: 27841037 DOI: 10.1080/14737140.2017.1261017] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDA) is characterized by expression of oncogenic KRas which drives all aspects of tumorigenesis. Oncogenic KRas induces the formation of reactive oxygen species (ROS) which have been implicated in initiation and progression of PDA. To facilitate tumor promoting levels and to avoid oncogene-induced senescence or cytotoxicity, ROS homeostasis in PDA cells is balanced by additional up-regulation of antioxidant systems. Areas covered: We examine the sources of ROS in PDA, the mechanisms by which ROS homeostasis is maintained, and the biological consequences of ROS in PDA. Additionally, we discuss the potential mechanisms for targeting ROS homoeostasis as a point of therapeutic intervention. An extensive review of the relevant literature as it relates to the topic was conducted using PubMed. Expert commentary: Even though oncogenic mutations in the KRAS gene have been detected in over 95% of human pancreatic adenocarcinoma, targeting its gene product, KRas, has been difficult. The dependency of PDA cells on balancing ROS homeostasis could be an angle for new prevention or treatment strategies. These include use of antioxidants to prevent formation or progression of precancerous lesions, or methods to increase ROS in tumor cells to toxic levels.
Collapse
Affiliation(s)
- Nisha Durand
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Peter Storz
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| |
Collapse
|
48
|
Bermejo M, Mangas-Sanjuan V, Gonzalez-Alvarez I, Gonzalez-Alvarez M. Enhancing Oral Absorption of β-Lapachone: Progress Till Date. Eur J Drug Metab Pharmacokinet 2016; 42:1-10. [DOI: 10.1007/s13318-016-0369-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Park JS, Lee YY, Kim J, Seo H, Kim HS. β-Lapachone increases phase II antioxidant enzyme expression via NQO1-AMPK/PI3K-Nrf2/ARE signaling in rat primary astrocytes. Free Radic Biol Med 2016; 97:168-178. [PMID: 27242267 DOI: 10.1016/j.freeradbiomed.2016.05.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/11/2016] [Accepted: 05/26/2016] [Indexed: 12/12/2022]
Abstract
β-Lapachone (β-LAP) is a naturally occurring quinine that exerts a number of pharmacological actions including antibacterial, antifungal, antimalarial, and antitumor activities. In the present study, we investigated whether β-LAP has an antioxidant effect in rat primary astrocytes. β-LAP suppressed intracellular reactive oxygen species (ROS) production induced by hydrogen peroxide and inhibited astroglial cell death. It also increased astrocytic expression of phase II antioxidant enzymes such as heme oxygenase-1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1), manganese superoxide dismutase (MnSOD), and catalase. Further mechanistic studies revealed that β-LAP activated AMPK and Akt, and pretreatment of cells with an AMPK inhibitor (compound C) or PI3K/Akt inhibitor (LY294002) suppressed β-LAP-induced antioxidant enzyme expression by inhibiting Nrf2/antioxidant response element (ARE) signaling. Compound C also decreased Akt phosphorylation, suggesting that AMPK is upstream of PI3K/Akt. Furthermore, the AMPK activator 5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside mimicked the effect of β-LAP by increasing Akt phosphorylation and ARE-mediated transcription, suggesting that AMPK plays a pivotal role in β-LAP-mediated antioxidant enzyme expression. Because β-LAP effects are usually associated with NQO1 activity, we examined the effect of NQO1 knockdown on antioxidant enzyme expression. Small interfering RNA (siRNA) specific for NQO1 inhibited β-LAP-induced AMPK/Akt phosphorylation and downstream antioxidant enzyme expression. Collectively, the results suggest that β-LAP increases antioxidant enzyme gene expression in astrocytes by modulating NQO1-AMPK/PI3K-Nrf2/ARE signaling.
Collapse
Affiliation(s)
- Jin-Sun Park
- Department of Molecular Medicine, Tissue Injury Defense Research Center, Ewha Womans University Medical School, Seoul, Republic of Korea
| | - Yu-Young Lee
- Department of Molecular Medicine, Tissue Injury Defense Research Center, Ewha Womans University Medical School, Seoul, Republic of Korea
| | - Jisun Kim
- Department of Molecular & Life Sciences, Hanyang University, Republic of Korea
| | - Hyemyung Seo
- Department of Molecular & Life Sciences, Hanyang University, Republic of Korea.
| | - Hee-Sun Kim
- Department of Molecular Medicine, Tissue Injury Defense Research Center, Ewha Womans University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Kahanda D, Chakrabarti G, Mcwilliams MA, Boothman DA, Slinker JD. Using DNA devices to track anticancer drug activity. Biosens Bioelectron 2016; 80:647-653. [PMID: 26901461 DOI: 10.1016/j.bios.2016.02.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 01/13/2023]
Abstract
It is beneficial to develop systems that reproduce complex reactions of biological systems while maintaining control over specific factors involved in such processes. We demonstrated a DNA device for following the repair of DNA damage produced by a redox-cycling anticancer drug, beta-lapachone (β-lap). These chips supported ß-lap-induced biological redox cycle and tracked subsequent DNA damage repair activity with redox-modified DNA monolayers on gold. We observed drug-specific changes in square wave voltammetry from these chips at therapeutic ß-lap concentrations of high statistical significance over drug-free control. We also demonstrated a high correlation of this change with the specific ß-lap-induced redox cycle using rational controls. The concentration dependence of ß-lap revealed significant signal changes at levels of high clinical significance as well as sensitivity to sub-lethal levels of ß-lap. Catalase, an enzyme decomposing peroxide, was found to suppress DNA damage at a NQO1/catalase ratio found in healthy cells, but was clearly overcome at a higher NQO1/catalase ratio consistent with cancer cells. We found that it was necessary to reproduce key features of the cellular environment to observe this activity. Thus, this chip-based platform enabled tracking of ß-lap-induced DNA damage repair when biological criteria were met, providing a unique synthetic platform for uncovering activity normally confined to inside cells.
Collapse
Affiliation(s)
- Dimithree Kahanda
- Department of Physics, The University of Texas at Dallas, 800 W. Campbell Rd., PHY 36, Richardson, TX 75080, United States
| | - Gaurab Chakrabarti
- Departments of Pharmacology, Oncology and Radiation Oncology, Laboratory of Molecular Stress Responses and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, ND2.210K 601 Forest Park Drive, Dallas, TX 75390-8807, United States
| | - Marc A Mcwilliams
- Department of Physics, The University of Texas at Dallas, 800 W. Campbell Rd., PHY 36, Richardson, TX 75080, United States
| | - David A Boothman
- Departments of Pharmacology, Oncology and Radiation Oncology, Laboratory of Molecular Stress Responses and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, ND2.210K 601 Forest Park Drive, Dallas, TX 75390-8807, United States
| | - Jason D Slinker
- Department of Physics, The University of Texas at Dallas, 800 W. Campbell Rd., PHY 36, Richardson, TX 75080, United States.
| |
Collapse
|