1
|
Nemeikaitė-Čėnienė A, Haberkant P, Kučiauskas D, Stein F, Čėnas N. Redox Proteomic Profile of Tirapazamine-Resistant Murine Hepatoma Cells. Int J Mol Sci 2023; 24:ijms24076863. [PMID: 37047836 PMCID: PMC10094930 DOI: 10.3390/ijms24076863] [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: 02/09/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
3-Amino-1,2,4-benzotriazine-1,4-dioxide (tirapazamine, TPZ) and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities. Their action is attributed to the enzymatic single-electron reduction to free radicals that initiate the prooxidant processes. In order to clarify the mechanisms of aerobic mammalian cytotoxicity of ArN→O, we derived a TPZ-resistant subline of murine hepatoma MH22a cells (resistance index, 5.64). The quantitative proteomic of wild-type and TPZ-resistant cells revealed 5818 proteins, of which 237 were up- and 184 down-regulated. The expression of the antioxidant enzymes aldehyde- and alcohol dehydrogenases, carbonyl reductases, catalase, and glutathione reductase was increased 1.6-5.2 times, whereas the changes in the expression of glutathione peroxidase, superoxide dismutase, thioredoxin reductase, and peroxiredoxins were less pronounced. The expression of xenobiotics conjugating glutathione-S-transferases was increased by 1.6-2.6 times. On the other hand, the expression of NADPH:cytochrome P450 reductase was responsible for the single-electron reduction in TPZ and for the 2.1-fold decrease. These data support the fact that the main mechanism of action of TPZ under aerobic conditions is oxidative stress. The unchanged expression of intranuclear antioxidant proteins peroxiredoxin, glutaredoxin, and glutathione peroxidase, and a modest increase in the expression of DNA damage repair proteins, tend to support non-site-specific but not intranuclear oxidative stress as a main factor of TPZ aerobic cytotoxicity.
Collapse
Affiliation(s)
- Aušra Nemeikaitė-Čėnienė
- State Research Institute Center for Innovative Medicine, Santariškių St. 5, LT-08406 Vilnius, Lithuania
| | - Per Haberkant
- Proteomics Core Facility EMBL Heidelberg, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Dalius Kučiauskas
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Frank Stein
- Proteomics Core Facility EMBL Heidelberg, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Narimantas Čėnas
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| |
Collapse
|
2
|
Boyd NH, Tran AN, Bernstock JD, Etminan T, Jones AB, Gillespie GY, Friedman GK, Hjelmeland AB. Glioma stem cells and their roles within the hypoxic tumor microenvironment. Theranostics 2021; 11:665-683. [PMID: 33391498 PMCID: PMC7738846 DOI: 10.7150/thno.41692] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor microenvironments are the result of cellular alterations in cancer that support unrestricted growth and proliferation and result in further modifications in cell behavior, which are critical for tumor progression. Angiogenesis and therapeutic resistance are known to be modulated by hypoxia and other tumor microenvironments, such as acidic stress, both of which are core features of the glioblastoma microenvironment. Hypoxia has also been shown to promote a stem-like state in both non-neoplastic and tumor cells. In glial tumors, glioma stem cells (GSCs) are central in tumor growth, angiogenesis, and therapeutic resistance, and further investigation of the interplay between tumor microenvironments and GSCs is critical to the search for better treatment options for glioblastoma. Accordingly, we summarize the impact of hypoxia and acidic stress on GSC signaling and biologic phenotypes, and potential methods to inhibit these pathways.
Collapse
|
3
|
Nemeikaitė-Čėnienė A, Šarlauskas J, Misevičienė L, Marozienė A, Jonušienė V, Lesanavičius M, Čėnas N. Aerobic Cytotoxicity of Aromatic N-Oxides: The Role of NAD(P)H:Quinone Oxidoreductase (NQO1). Int J Mol Sci 2020; 21:ijms21228754. [PMID: 33228195 PMCID: PMC7699506 DOI: 10.3390/ijms21228754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities, which are typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the role of NAD(P)H:quinone oxidoreductase (NQO1) in ArN→O aerobic cytotoxicity. We synthesized 9 representatives of ArN→O with uncharacterized redox properties and examined their single-electron reduction by rat NADPH:cytochrome P-450 reductase (P-450R) and Plasmodium falciparum ferredoxin:NADP+ oxidoreductase (PfFNR), and by rat NQO1. NQO1 catalyzed both redox cycling and the formation of stable reduction products of ArN→O. The reactivity of ArN→O in NQO1-catalyzed reactions did not correlate with the geometric average of their activity towards P-450R- and PfFNR, which was taken for the parameter of their redox cycling efficacy. The cytotoxicity of compounds in murine hepatoma MH22a cells was decreased by antioxidants and the inhibitor of NQO1, dicoumarol. The multiparameter regression analysis of the data of this and a previous study (DOI: 10.3390/ijms20184602) shows that the cytotoxicity of ArN→O (n = 18) in MH22a and human colon carcinoma HCT-116 cells increases with the geometric average of their reactivity towards P-450R and PfFNR, and with their reactivity towards NQO1. These data demonstrate that NQO1 is a potentially important target of action of heteroaromatic N-oxides.
Collapse
Affiliation(s)
- Aušra Nemeikaitė-Čėnienė
- State Research Institute Center for Innovative Medicine, Santariškių St. 5, LT-08406 Vilnius, Lithuania;
| | - Jonas Šarlauskas
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Lina Misevičienė
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Audronė Marozienė
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Violeta Jonušienė
- Institute of Biosciences of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania;
| | - Mindaugas Lesanavičius
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
| | - Narimantas Čėnas
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania; (J.Š.); (L.M.); (A.M.); (M.L.)
- Correspondence: ; Tel.: +370-5-223-4392
| |
Collapse
|
4
|
Muhamad N, Na-Bangchang K. Metabolite Profiling in Anticancer Drug Development: A Systematic Review. Drug Des Devel Ther 2020; 14:1401-1444. [PMID: 32308372 PMCID: PMC7154001 DOI: 10.2147/dddt.s221518] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/20/2020] [Indexed: 12/24/2022] Open
Abstract
Drug metabolism is one of the most important pharmacokinetic processes and plays an important role during the stage of drug development. The metabolite profile investigation is important as the metabolites generated could be beneficial for therapy or leading to serious toxicity. This systematic review aims to summarize the research articles relating to the metabolite profile investigation of conventional drugs and herb-derived compounds for cancer chemotherapy, to examine factors influencing metabolite profiling of these drugs/compounds, and to determine the relationship between therapeutic efficacy and toxicity of their metabolites. The literature search was performed through PubMed and ScienceDirect databases up to January 2019. Out of 830 published articles, 78 articles were included in the analysis based on pre-defined inclusion and exclusion criteria. Both phase I and II enzymes metabolize the anticancer agents/herb-derived compounds . The major phase I reactions include oxidation/hydroxylation and hydrolysis, while the major phase II reactions are glucuronidation, methylation, and sulfation. Four main factors were found to influence metabolite formation, including species, gender, and route and dose of drug administration. Some metabolites were identified as active or toxic metabolites. This information is critical for cancer chemotherapy and anticancer drug development.
Collapse
Affiliation(s)
- Nadda Muhamad
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Kesara Na-Bangchang
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand.,Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand.,Drug Discovery and Development Center, Office of Advanced Sciences and Technology, Thammasat University, Pathum Thani 12120, Thailand
| |
Collapse
|
5
|
Gerard Y, Voissière A, Peyrode C, Galmier MJ, Maubert E, Ghedira D, Tarrit S, Gaumet V, Canitrot D, Miot-Noirault E, Chezal JM, Weber V. Design, synthesis and evaluation of targeted hypoxia-activated prodrugs applied to chondrosarcoma chemotherapy. Bioorg Chem 2020; 98:103747. [PMID: 32208207 DOI: 10.1016/j.bioorg.2020.103747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/13/2020] [Accepted: 03/09/2020] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment in chondrosarcoma (CHS), a chemo- and radio-resistant cancer provides unique hallmarks for developing a chondrosarcoma targeted drug-delivery system. Tumor targeting could be achieved using a quaternary ammonium function (QA) as a ligand for aggrecan, the main high negative charged proteoglycan of the extracellular matrix of CHS, and a 2-nitroimidazole as trigger that enables hypoxia-responsive drug release. In a previous work, ICF05016 was identified as efficient proteoglycan-targeting hypoxia-activated prodrug in a human extraskeletal myxoid chondrosarcoma model in mice and a first study of the structure-activity relationship of the QA function and the alkyl linker length was conducted. Here, we report the second part of the study, namely the modification of the nitro-aromatic trigger and the position of the proteoglycan-targeting ligand at the aromatic ring as well as the nature of the alkylating mustard. Synthetic approaches have been established to functionalize the 2-nitroimidazole ring at the N-1 and C-4 positions with a terminal tertiary alkyl amine, and to perform the phosphorylation step namely through the use of an amine borane complex, leading to phosphoramide and isophosphoramide mustards and also to a phosphoramide mustard bearing four 2-chloroethyl chains. In a preliminary study using a reductive chemical activation, QA-conjugates, except the 4-nitrobenzyl one, were showed to undergo efficient cleavage with release of the corresponding mustard. However N,N,N-trimethylpropylaminium tethered to the N-1 or C-4 positions of the imidazole seemed to hamper the enzymatic reduction of the prodrugs and all tested compounds featured moderate selectivity toward hypoxic cells, likely not sufficient for application as hypoxia-activated prodrugs.
Collapse
Affiliation(s)
- Yvain Gerard
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Aurélien Voissière
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Caroline Peyrode
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Marie-Josephe Galmier
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Elise Maubert
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Donia Ghedira
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Sebastien Tarrit
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Vincent Gaumet
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Damien Canitrot
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Elisabeth Miot-Noirault
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Jean-Michel Chezal
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Valérie Weber
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
6
|
Wong WW, Jackson RK, Liew LP, Dickson BD, Cheng GJ, Lipert B, Gu Y, Hunter FW, Wilson WR, Hay MP. Hypoxia-selective radiosensitisation by SN38023, a bioreductive prodrug of DNA-dependent protein kinase inhibitor IC87361. Biochem Pharmacol 2019; 169:113641. [PMID: 31541630 DOI: 10.1016/j.bcp.2019.113641] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023]
Abstract
DNA-dependent protein kinase (DNA-PK) plays a key role in repair of radiation-induced DNA double strand breaks (DSB) by non-homologous end-joining. DNA-PK inhibitors (DNA-PKi) are therefore efficient radiosensitisers, but normal tissue radiosensitisation represents a risk for their use in radiation oncology. Here we describe a novel prodrug, SN38023, that is metabolised to a potent DNA-PKi (IC87361) selectively in radioresistant hypoxic cells. DNA-PK inhibitory potency of SN38023 was 24-fold lower than IC87361 in cell-free assays, consistent with molecular modelling studies suggesting that SN38023 is unable to occupy one of the predicted DNA-PK binding modes of IC87361. One-electron reduction of the prodrug by radiolysis of anoxic formate solutions, and by metabolic reduction in anoxic HCT116/POR cells that overexpress cytochrome P450 oxidoreductase (POR), generated IC87361 efficiently as assessed by LC-MS. SN38023 inhibited radiation-induced Ser2056 autophosphorylation of DNA-PK catalytic subunit and radiosensitised HCT116/POR and UT-SCC-54C cells selectively under anoxia. SN38023 was an effective radiosensitiser in anoxic HCT116 spheroids, demonstrating potential for penetration into hypoxic tumour tissue, but in spheroid co-cultures of high-POR and POR-null cells it showed no evidence of bystander effects resulting from local diffusion of IC87361. Pharmacokinetics of IC87361 and SN38023 at maximum achievable doses in NIH-III mice demonstrated sub-optimal exposure of UT-SCC-54C tumour xenografts and did not provide significant tumour radiosensitisation. In conclusion, SN38023 has potential for exploiting hypoxia for selective delivery of a potent DNA-PKi to the most radioresistant subpopulation of cells in tumours. However, prodrugs providing improved systemic pharmacokinetics and that release DNA-PKi that elicit bystander effects are needed to maximise therapeutic utility.
Collapse
Affiliation(s)
- Way Wua Wong
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| | - Rosanna K Jackson
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| | - Lydia P Liew
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Benjamin D Dickson
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| | - Gary J Cheng
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| | - Barbara Lipert
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| | - Yongchuan Gu
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| | - Michael P Hay
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
7
|
Nemeikaitė-Čėnienė A, Šarlauskas J, Jonušienė V, Marozienė A, Misevičienė L, Yantsevich AV, Čėnas N. Kinetics of Flavoenzyme-Catalyzed Reduction of Tirapazamine Derivatives: Implications for Their Prooxidant Cytotoxicity. Int J Mol Sci 2019; 20:ijms20184602. [PMID: 31533349 PMCID: PMC6769651 DOI: 10.3390/ijms20184602] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 12/23/2022] Open
Abstract
Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit promising antibacterial, antiprotozoal, and tumoricidal activities. Their action is typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the mechanism(s) of aerobic mammalian cell cytotoxicity of ArN→O performing the parallel studies of their reactions with NADPH:cytochrome P-450 reductase (P-450R), adrenodoxin reductase/adrenodoxin (ADR/ADX), and NAD(P)H:quinone oxidoreductase (NQO1); we found that in P-450R and ADR/ADX-catalyzed single-electron reduction, the reactivity of ArN→O (n = 9) increased with their single-electron reduction midpoint potential (E17), and correlated with the reactivity of quinones. NQO1 reduced ArN→O at low rates with concomitant superoxide production. The cytotoxicity of ArN→O in murine hepatoma MH22a and human colon adenocarcinoma HCT-116 cells increased with their E17, being systematically higher than that of quinones. The cytotoxicity of both groups of compounds was prooxidant. Inhibitor of NQO1, dicoumarol, and inhibitors of cytochromes P-450 α-naphthoflavone, isoniazid and miconazole statistically significantly (p < 0.02) decreased the toxicity of ArN→O, and potentiated the cytotoxicity of quinones. One may conclude that in spite of similar enzymatic redox cycling rates, the cytotoxicity of ArN→O is higher than that of quinones. This is partly attributed to ArN→O activation by NQO1 and cytochromes P-450. A possible additional factor in the aerobic cytotoxicity of ArN→O is their reductive activation in oxygen-poor cell compartments, leading to the formation of DNA-damaging species similar to those forming under hypoxia.
Collapse
Affiliation(s)
- Aušra Nemeikaitė-Čėnienė
- State Research Institute Center for Innovative Medicine, Santariškių St. 5, LT-08406 Vilnius, Lithuania.
| | - Jonas Šarlauskas
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Violeta Jonušienė
- Department of Biochemistry and Molecular Biology, Institute of Biosciences of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Audronė Marozienė
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Lina Misevičienė
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| | - Aliaksei V Yantsevich
- Institute of Bioorganic Chemistry, NAS of Belarus, Kuprevicha 5/2, BY-220072 Minsk, Belarus.
| | - Narimantas Čėnas
- Department of Xenobiotics Biochemistry, Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| |
Collapse
|
8
|
Hay MP, Shin HN, Wong WW, Sahimi WW, Vaz ATD, Yadav P, Anderson RF, Hicks KO, Wilson WR. Benzotriazine Di-Oxide Prodrugs for Exploiting Hypoxia and Low Extracellular pH in Tumors. Molecules 2019; 24:E2524. [PMID: 31295864 PMCID: PMC6680510 DOI: 10.3390/molecules24142524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 12/31/2022] Open
Abstract
Extracellular acidification is an important feature of tumor microenvironments but has yet to be successfully exploited in cancer therapy. The reversal of the pH gradient across the plasma membrane in cells that regulate intracellular pH (pHi) has potential to drive the selective uptake of weak acids at low extracellular pH (pHe). Here, we investigate the dual targeting of low pHe and hypoxia, another key feature of tumor microenvironments. We prepared eight bioreductive prodrugs based on the benzotriazine di-oxide (BTO) nucleus by appending alkanoic or aminoalkanoic acid sidechains. The BTO acids showed modest selectivity for both low pHe (pH 6.5 versus 7.4, ratios 2 to 5-fold) and anoxia (ratios 2 to 8-fold) in SiHa and FaDu cell cultures. Related neutral BTOs were not selective for acidosis, but had greater cytotoxic potency and hypoxic selectivity than the BTO acids. Investigation of the uptake and metabolism of representative BTO acids confirmed enhanced uptake at low pHe, but lower intracellular concentrations than expected for passive diffusion. Further, the modulation of intracellular reductase activity and competition by the cell-excluded electron acceptor WST-1 suggests that the majority of metabolic reductions of BTO acids occur at the cell surface, compromising the engagement of the resulting free radicals with intracellular targets. Thus, the present study provides support for designing bioreductive prodrugs that exploit pH-dependent partitioning, suggesting, however, that that the approach should be applied to prodrugs with obligate intracellular activation.
Collapse
Affiliation(s)
- Michael P Hay
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand
| | - Hong Nam Shin
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Way Wua Wong
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Wan Wan Sahimi
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Aaron T D Vaz
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Pooja Yadav
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Robert F Anderson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Symonds St, Auckland 1142, New Zealand.
| |
Collapse
|
9
|
Mao X, McManaway S, Jaiswal JK, Hong CR, Wilson WR, Hicks KO. Schedule-dependent potentiation of chemotherapy drugs by the hypoxia-activated prodrug SN30000. Cancer Biol Ther 2019; 20:1258-1269. [PMID: 31131698 DOI: 10.1080/15384047.2019.1617570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypoxia-activated prodrugs (HAPs) are hypothesized to improve the therapeutic index of chemotherapy drugs that are ineffective against tumor cells in hypoxic microenvironments. SN30000 (CEN-209) is a benzotriazine di-N-oxide HAP that potentiates radiotherapy in preclinical models, but its combination with chemotherapy has not been explored. Here we apply multiple models (monolayers, multicellular spheroids and tumor xenografts) to identify promising SN30000/chemotherapy combinations (with chemotherapy drugs before, during or after SN30000 exposure). SN30000, unlike doxorubicin, cisplatin, gemcitabine or paclitaxel, was more active against cells in spheroids than monolayers by clonogenic assay. Combinations of SN30000 and chemotherapy drugs in HCT116/GFP and SiHa spheroids demonstrated hypoxia-and schedule-dependent potentiation of gemcitabine or doxorubicin in growth inhibition and clonogenic assays. Co-administration with SN30000 suppressed clearance of gemcitabine in NIH-III mice, likely due to SN30000-induced hypothermia which also modulated extravascular transport of gemcitabine in tumor tissue as assessed from its diffusion through HCT116 multicellular layer cultures. Despite these systemic effects, the same schedules that gave therapeutic synergy in spheroids (SN30000 3 h before or during gemcitabine, but not gemcitabine 3 h before SN30000) enhanced growth delay of HCT116 xenografts without increasing host toxicity. Identification of hypoxic and S-phase cells by immunohistochemistry and flow cytometry established that hypoxic cells initially spared by gemcitabine subsequently reoxygenate and re-enter the cell cycle, and that this repopulation is prevented by SN30000 only when administered with or before gemcitabine. This illustrates the value of spheroids in modeling tumor microenvironment-dependent drug interactions, and the potential of HAPs for overcoming hypoxia-mediated drug resistance.
Collapse
Affiliation(s)
- Xinjian Mao
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Sarah McManaway
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Jagdish K Jaiswal
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Cho R Hong
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland , Auckland , New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland , Auckland , New Zealand
| |
Collapse
|
10
|
Marx S, Van Gysel M, Breuer A, Dal Maso T, Michiels C, Wouters J, Le Calvé B. Potentialization of anticancer agents by identification of new chemosensitizers active under hypoxia. Biochem Pharmacol 2019; 162:224-236. [DOI: 10.1016/j.bcp.2019.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022]
|
11
|
Shen X, Gates KS. Enzyme-Activated Generation of Reactive Oxygen Species from Heterocyclic N-Oxides under Aerobic and Anaerobic Conditions and Its Relevance to Hypoxia-Selective Prodrugs. Chem Res Toxicol 2019; 32:348-361. [PMID: 30817135 DOI: 10.1021/acs.chemrestox.9b00036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Enzymatic one-electron reduction of heterocyclic N-oxides can lead to the intracellular generation of reactive oxygen species via several different chemical pathways. These reactions may be relevant to hypoxia-selective anticancer drugs, antimicrobial agents, and unwanted toxicity of heterocylic nitrogen compounds.
Collapse
|
12
|
Sharma A, Arambula JF, Koo S, Kumar R, Singh H, Sessler JL, Kim JS. Hypoxia-targeted drug delivery. Chem Soc Rev 2019; 48:771-813. [PMID: 30575832 PMCID: PMC6361706 DOI: 10.1039/c8cs00304a] [Citation(s) in RCA: 324] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hypoxia is a state of low oxygen tension found in numerous solid tumours. It is typically associated with abnormal vasculature, which results in a reduced supply of oxygen and nutrients, as well as impaired delivery of drugs. The hypoxic nature of tumours often leads to the development of localized heterogeneous environments characterized by variable oxygen concentrations, relatively low pH, and increased levels of reactive oxygen species (ROS). The hypoxic heterogeneity promotes tumour invasiveness, metastasis, angiogenesis, and an increase in multidrug-resistant proteins. These factors decrease the therapeutic efficacy of anticancer drugs and can provide a barrier to advancing drug leads beyond the early stages of preclinical development. This review highlights various hypoxia-targeted and activated design strategies for the formulation of drugs or prodrugs and their mechanism of action for tumour diagnosis and treatment.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Chemistry, Korea University, Seoul, 02841, Korea.
| | | | | | | | | | | | | |
Collapse
|
13
|
DiGiacomo JW, Gilkes DM. Therapeutic Strategies to Block the Hypoxic Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:141-157. [DOI: 10.1007/978-3-030-12734-3_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Mao X, McManaway S, Jaiswal JK, Patel PB, Wilson WR, Hicks KO, Bogle G. An agent-based model for drug-radiation interactions in the tumour microenvironment: Hypoxia-activated prodrug SN30000 in multicellular tumour spheroids. PLoS Comput Biol 2018; 14:e1006469. [PMID: 30356233 PMCID: PMC6218095 DOI: 10.1371/journal.pcbi.1006469] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 11/05/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023] Open
Abstract
Multicellular tumour spheroids capture many characteristics of human tumour microenvironments, including hypoxia, and represent an experimentally tractable in vitro model for studying interactions between radiotherapy and anticancer drugs. However, interpreting spheroid data is challenging because of limited ability to observe cell fate within spheroids dynamically. To overcome this limitation, we have developed a hybrid continuum/agent-based model (ABM) for HCT116 tumour spheroids, parameterised using experimental models (monolayers and multilayers) in which reaction and diffusion can be measured directly. In the ABM, cell fate is simulated as a function of local oxygen, glucose and drug concentrations, determined by solving diffusion equations and intracellular reactions. The model is lattice-based, with cells occupying discrete locations on a 3D grid embedded within a coarser grid that encompasses the culture medium; separate solvers are employed for each grid. The generated concentration fields account for depletion in the medium and specify concentration-time profiles within the spheroid. Cell growth and survival are determined by intracellular oxygen and glucose concentrations, the latter based on direct measurement of glucose diffusion/reaction (in multilayers) for the first time. The ABM reproduces known features of spheroids including overall growth rate, its oxygen and glucose dependence, peripheral cell proliferation, central hypoxia and necrosis. We extended the ABM to describe in detail the hypoxia-dependent interaction between ionising radiation and a hypoxia-activated prodrug (SN30000), again using experimentally determined parameters; the model accurately simulated clonogenic cell killing in spheroids, while inclusion of reversible cell cycle delay was required to account for the marked spheroid growth delay after combined radiation and SN30000. This ABM of spheroid growth and response exemplifies the utility of integrating computational and experimental tools for investigating radiation/drug interactions, and highlights the critical importance of understanding oxygen, glucose and drug concentration gradients in interpreting activity of therapeutic agents in spheroid models. Studies in 3D cultures, notably multicellular tumour spheroids that mimic many features of solid tumours, have great potential for speeding up anticancer drug development. However the increased complexity of 3D cultures makes interpretation of experiments more difficult. We have developed a hybrid continuum/agent-based mathematical model, validated by experiments, to aid interpretation of spheroid experiments in developing drugs designed to eliminate radiation-resistant hypoxic cells. This model includes key features of the tumour microenvironment including oxygen and glucose transport and regions of hypoxia where the cells are resistant to radiation, but sensitive to hypoxia-activated prodrugs such as SN30000. This enables us to predict the growth and cell response in untreated spheroids and compare the results to spheroids treated with radiation and SN30000. We demonstrate good prediction of cellular responses in spheroids treated with radiation and SN30000 and good agreement with spheroid regrowth after treatment when additional effects of cellular growth delay are added. This demonstrates that the modelling approach has potential to improve interpretation of experimental investigations of drug and radiation combinations.
Collapse
Affiliation(s)
- Xinjian Mao
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Sarah McManaway
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Jagdish K. Jaiswal
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Priyanka B. Patel
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - William R. Wilson
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Kevin O. Hicks
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- * E-mail:
| | - Gib Bogle
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
15
|
Hong CR, Bogle G, Wang J, Patel K, Pruijn FB, Wilson WR, Hicks KO. Bystander Effects of Hypoxia-Activated Prodrugs: Agent-Based Modeling Using Three Dimensional Cell Cultures. Front Pharmacol 2018; 9:1013. [PMID: 30279659 PMCID: PMC6153434 DOI: 10.3389/fphar.2018.01013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022] Open
Abstract
Intra-tumor heterogeneity represents a major barrier to anti-cancer therapies. One strategy to minimize this limitation relies on bystander effects via diffusion of cytotoxins from targeted cells. Hypoxia-activated prodrugs (HAPs) have the potential to exploit hypoxia in this way, but robust methods for measuring bystander effects are lacking. The objective of this study is to develop experimental models (monolayer, multilayer, and multicellular spheroid co-cultures) comprising 'activator' cells with high expression of prodrug-activating reductases and reductase-deficient 'target' cells, and to couple these with agent-based models (ABMs) that describe diffusion and reaction of prodrugs and their active metabolites, and killing probability for each cell. HCT116 cells were engineered as activators by overexpressing P450 oxidoreductase (POR) and as targets by knockout of POR, with fluorescent protein and antibiotic resistance markers to enable their quantitation in co-cultures. We investigated two HAPs with very different pharmacology: SN30000 is metabolized to DNA-breaking free radicals under hypoxia, while the dinitrobenzamide PR104A generates DNA-crosslinking nitrogen mustard metabolites. In anoxic spheroid co-cultures, increasing the proportion of activator cells decreased killing of both activators and targets by SN30000. An ABM parameterized by measuring SN30000 cytotoxicity in monolayers and diffusion-reaction in multilayers accurately predicted SN30000 activity in spheroids, demonstrating the lack of bystander effects and that rapid metabolic consumption of SN30000 inhibited prodrug penetration. In contrast, killing of targets by PR104A in anoxic spheroids was markedly increased by activators, demonstrating that a bystander effect more than compensates any penetration limitation. However, the ABM based on the well-studied hydroxylamine and amine metabolites of PR104A did not fit the cell survival data, indicating a need to reassess its cellular pharmacology. Characterization of extracellular metabolites of PR104A in anoxic cultures identified more stable, lipophilic, activated dichloro mustards with greater tissue diffusion distances. Including these metabolites explicitly in the ABM provided a good description of activator and target cell killing by PR104A in spheroids. This study represents the most direct demonstration of a hypoxic bystander effect for PR104A to date, and demonstrates the power of combining mathematical modeling of pharmacokinetics/pharmacodynamics with multicellular culture models to dissect bystander effects of targeted drug carriers.
Collapse
Affiliation(s)
- Cho R. Hong
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Gib Bogle
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Jingli Wang
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Kashyap Patel
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Frederik B. Pruijn
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - William R. Wilson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Kevin O. Hicks
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|