1
|
Timmers M, Weterings J, van Geijn M, Bell R, Lenting PE, Rijcken CJ, Vermonden T, Hennink WE, Liskamp RM. A New Class of Tunable Acid-Sensitive Linkers for Native Drug Release Based on the Trityl Protecting Group. Bioconjug Chem 2022; 33:1707-1715. [PMID: 35979909 PMCID: PMC9501768 DOI: 10.1021/acs.bioconjchem.2c00310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/03/2022] [Indexed: 12/24/2022]
Abstract
Core-cross-linked polymeric micelles (CCPMs) are a promising nanoparticle platform due to favorable properties such as their long circulation and tumor disposition exploiting the enhanced permeability and retention (EPR) effect. Sustained release of covalently linked drugs from the hydrophobic core of the CCPM can be achieved by a biodegradable linker that connects the drug and the core. This study investigates the suitability of trityl-based linkers for the design of acid-triggered native active pharmaceutical ingredient (API) release from CCPMs. Trityl linker derivatives with different substituent patterns were synthesized and conjugated to model API compounds such as DMXAA-amine, doxorubicin, and gemcitabine, and their release kinetics were studied. Hereafter, API release from CCPMs based on mPEG-b-pHPMAmLac block copolymers was investigated. Variation of the trityl substitution pattern showed tunability of the API release rate from the trityl-based linker with t1/2 varying from <1.0 to 5.0 h at pH 5.0 and t1/2 from 6.5 to >24 h at pH 7.4, all at 37 °C. A clear difference in release kinetics was found between gemcitabine and doxorubicin, with gemcitabine showing no detectable release for 72 h at pH 5.0 and doxorubicin showing a t1/2 of less than 1 h. Based on these findings, we show that the reaction mechanism of trityl deprotection plays an important role in the API release kinetics. The first step in this mechanism, which is protonation of the trityl-bound amine, is pKa-dependent, which explains the difference in release rate. In conclusion, acid-sensitive and tunable trityl linkers are highly promising for the design of linker-API conjugates and for their use in CCPMs.
Collapse
Affiliation(s)
- Matt Timmers
- Cristal
Therapeutics, Maastricht 6229 EV, The Netherlands
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | | | | | - Roel Bell
- Symeres, Nijmegen 6546 BB, The
Netherlands
| | - Peter E. Lenting
- School
of Chemistry, University of Glasgow, Glasgow G12 8QQ, U.K.
| | | | - Tina Vermonden
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Rob M.J. Liskamp
- Cristal
Therapeutics, Maastricht 6229 EV, The Netherlands
- School
of Chemistry, University of Glasgow, Glasgow G12 8QQ, U.K.
- Department
of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht 6229 ER, The Netherlands
| |
Collapse
|
2
|
Phung CD, Tran TH, Pham LM, Nguyen HT, Jeong JH, Yong CS, Kim JO. Current developments in nanotechnology for improved cancer treatment, focusing on tumor hypoxia. J Control Release 2020; 324:413-429. [PMID: 32461115 DOI: 10.1016/j.jconrel.2020.05.029] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a common feature of the tumor microenvironment, which is characterized by tissue oxygen deficiency due to an aggressive proliferation of cancer cells. Hypoxia activates hypoxia-inducible factor-dependent signaling, which in turn regulates metabolic reprogramming, immune suppression, resistance to apoptosis, angiogenesis, metastasis, and invasion to secondary sites. In this review, we provide an overview of the use of nanotechnology to harmonize intra-tumoral oxygen or suppress hypoxia-related signaling for an improved efficacy of cancer treatment. The biological background was followed by conducting a literature review on the (1) nanoparticles responsible for enhancing oxygen levels within the tumor, (2) nanoparticles sensitizing hypoxia, (3) nanoparticles suppressing hypoxia-inducing factor, (4) nanoparticles that relieve tumor hypoxia for enhancement of chemotherapy, photodynamic therapy, and immunotherapy, either individually or in combination. Lastly, the heterogeneity of cancer and limitations of nanotechnology are discussed to facilitate translational therapeutic treatment.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam
| | - Le Minh Pham
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
3
|
Trebinska-Stryjewska A, Swiech O, Opuchlik LJ, Grzybowska EA, Bilewicz R. Impact of Medium pH on DOX Toxicity toward HeLa and A498 Cell Lines. ACS OMEGA 2020; 5:7979-7986. [PMID: 32309708 PMCID: PMC7161040 DOI: 10.1021/acsomega.9b04479] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/20/2020] [Indexed: 05/20/2023]
Abstract
The influence of the pH of the multicomponent cell medium on the performance of doxorubicin (DOX), an anticancer drug, was studied on the examples of cervical (HeLa) and kidney (A498) cancer cell lines. The change of pH of the cell medium to more acidic led to a decrease of DOX toxicity on both cell lines due to the change of drug permeability across the cell membrane as a result of drug protonation. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) studies and lactate dehydrogenase (LDH) release tests have shown low toxicity of the drug, especially in the case of A498 cells, which are characterized by an extremely high glycolytic metabolism. The behavior was ascribed primarily to the increased proton concentration in the peripheral blood follicle in the presence of products of the acidic glycolytic metabolism. It is not observed in the measurements performed in commercially available media since they usually have a neutral pH. In earlier reports on kidney cancer, several mechanisms were discussed, including the metabolism of DOX to its less toxic derivative, doxorubicinol, overexpression of ATP binding cassette subfamily B member 1 (ABCB1) transporters, that remove DOX from the inside of cells; however, there was no focus on the simple but very important contribution of drug protonation described in the present study. Drug pH-dependent equilibria in the cell medium should be considered since changes in the drug form may be an additional reason for multidrug resistance.
Collapse
Affiliation(s)
- Alicja Trebinska-Stryjewska
- Institute
of Optoelectronics, Biomedical Engineering Centre, Military University of Technology, 00-908 Warsaw, Poland
- Department
of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Olga Swiech
- Faculty
of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| | | | - Ewa A. Grzybowska
- Department
of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Renata Bilewicz
- Faculty
of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| |
Collapse
|
4
|
Wong AD, Russell LM, Katt ME, Searson PC. Chemotherapeutic Drug Delivery and Quantitative Analysis of Proliferation, Apoptosis, and Migration in a Tissue-Engineered Three-Dimensional Microvessel Model of the Tumor Microenvironment. ACS Biomater Sci Eng 2018; 5:633-643. [DOI: 10.1021/acsbiomaterials.8b00877] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Andrew D. Wong
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Luisa M. Russell
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Moriah E. Katt
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology (INBT), Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Oncology, Johns Hopkins University, 1650 Orleans Street, Baltimore, Maryland 21287, United States
| |
Collapse
|
5
|
Russell LM, Hultz M, Searson PC. Leakage kinetics of the liposomal chemotherapeutic agent Doxil: The role of dissolution, protonation, and passive transport, and implications for mechanism of action. J Control Release 2017; 269:171-176. [PMID: 29122661 DOI: 10.1016/j.jconrel.2017.11.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 10/25/2017] [Accepted: 11/04/2017] [Indexed: 01/24/2023]
Abstract
Doxil, a liposomal formulation of the chemotherapeutic drug doxorubicin, is FDA-approved for multiple indications. Doxil liposomes are designed to retain doxorubicin in circulation, minimize clearance by the mononuclear phagocyte system, and limit uptake in healthy tissue. Although pharmacokinetic data and survival statistics from clinical trials provide insight into distribution and efficacy, many details of the mechanism of action remain unresolved, despite the importance in translating liposome-based drug delivery systems to other molecules and cargo. Therefore, the objective of this study is to quantitatively assess the kinetics of doxorubicin leakage from Doxil liposomes. In contrast to previous studies, we consider three processes: dissolution of solid doxorubicin, protonation/deprotonation of soluble doxorubicin, and passive transport of neutral doxorubicin across the lipid bilayer of the liposomes. Experiments were performed for Doxil, Doxil-like liposomes, and Doxil-like liposomes with reduced cholesterol and pegylation. To mimic physiological conditions, we also performed experiments in serum and under slightly acidic conditions at pH5. We show that crystalline doxorubicin dissolution can be described by a first order rate constant of 1.0×10-9cms-1 at 37°C. Doxorubicin leakage can be described by first order rate constant for transport across the lipid bilayer with values in the range from 1 to 3×10-12cms-1 at 37°C. Based on these results we discuss implications for the mechanism of action, taking Doxil pharmacokinetics into account.
Collapse
Affiliation(s)
- Luisa M Russell
- Department of Materials Science and Engineering, Johns Hopkins University, USA; Institute for Nanobiotechnology, Johns Hopkins University, USA
| | - Margot Hultz
- Department of Materials Science and Engineering, Johns Hopkins University, USA; Institute for Nanobiotechnology, Johns Hopkins University, USA
| | - Peter C Searson
- Department of Materials Science and Engineering, Johns Hopkins University, USA; Institute for Nanobiotechnology, Johns Hopkins University, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
6
|
Nikolaienko TY. Interaction of anticancer drug doxorubicin with sodium oleate bilayer: Insights from molecular dynamics simulations. J Mol Liq 2017. [DOI: 10.1016/j.molliq.2016.11.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
7
|
Zhang A, Yao L, An M. Reversing the undesirable pH-profile of doxorubicin via activation of a di-substituted maleamic acid prodrug at tumor acidity. Chem Commun (Camb) 2017; 53:12826-12829. [DOI: 10.1039/c7cc06843c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Selective drug release at pH 6.7 over pH 7.4 is achieved through a combination of ‘switch-on’ and ‘switch-off’ mechanisms.
Collapse
Affiliation(s)
- Anqi Zhang
- Department of Chemistry
- State University of New York (SUNY)
- Binghamton University
- Binghamton
- USA
| | - Lan Yao
- Department of Physics
- Applied Physics and Astronomy
- SUNY
- Binghamton University
- Binghamton
| | - Ming An
- Department of Chemistry
- State University of New York (SUNY)
- Binghamton University
- Binghamton
- USA
| |
Collapse
|
8
|
Bogorad MI, Searson PC. Real-time imaging and quantitative analysis of doxorubicin transport in a perfusable microvessel platform. Integr Biol (Camb) 2016; 8:976-84. [PMID: 27523481 PMCID: PMC5035133 DOI: 10.1039/c6ib00082g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Here we report on real-time imaging and quantitative analysis of solute transport in perfusable cylindrical microvessels formed from Madin-Darby canine kidney (MDCK) cells embedded in a collagen matrix. Fluorescence microscopy was used to image the kinetics of doxorubicin transport following injection. To assess the role of efflux pumps on transport, experiments were performed in microvessels formed from MDCK.2, MDCKII-w/t, and MDCKII-MDR1 cells. MDCKII-w/t and MDCKII-MDR1 showed significant doxorubicin accumulation in the cells, characteristic of the pharmacokinetics of doxorubicin. We present a model for doxorubicin transport that takes into account transport across the cell layer. These results demonstrate how real-time imaging of cell microvessels can be used to analyze the mechanisms of transport and distribution following systemic delivery.
Collapse
Affiliation(s)
- Max I Bogorad
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | | |
Collapse
|
9
|
Shahabi S, Döscher S, Bollhorst T, Treccani L, Maas M, Dringen R, Rezwan K. Enhancing Cellular Uptake and Doxorubicin Delivery of Mesoporous Silica Nanoparticles via Surface Functionalization: Effects of Serum. ACS APPLIED MATERIALS & INTERFACES 2015; 7:26880-91. [PMID: 26562468 DOI: 10.1021/acsami.5b09483] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In this study, we demonstrate how functional groups on the surface of mesoporous silica nanoparticles (MSNPs) can influence the encapsulation and release of the anticancer drug doxorubicin, as well as cancer cell response in the absence or presence of serum proteins. To this end, we synthesized four differently functionalized MSNPs with amine, sulfonate, polyethylene glycol, or polyethylene imine functional surface groups, as well as one type of antibody-conjugated MSNP for specific cellular targeting, and we characterized these MSNPs regarding their physicochemical properties, colloidal stability in physiological media, and uptake and release of doxorubicin in vitro. Then, the MSNPs were investigated for their cytotoxic potential on cancer cells. Cationic MSNPs could not be loaded with doxorubicin and did therefore not show any cytotoxic and antiproliferative potential on osteosarcoma cells, although they were efficiently taken up into the cells in the presence or absence of serum. In contrast, substantial amounts of doxorubicin were loaded into negatively charged and unfunctionalized MSNPs. Especially, sulfonate-functionalized doxorubicin-loaded MSNPs were efficiently taken up into the cells in the presence of serum and showed an accelerated toxic and antiproliferative potential compared to unfunctionalized MSNPs, antibody-conjugated MSNPs, and even free doxorubicin. These findings stress the high importance of the surface charge as well as of the protein corona for designing and applying nanoparticles for targeted drug delivery.
Collapse
Affiliation(s)
- Shakiba Shahabi
- Advanced Ceramics, University of Bremen , Am Biologischen Garten 2, 28359 Bremen, Germany
| | - Svea Döscher
- Advanced Ceramics, University of Bremen , Am Biologischen Garten 2, 28359 Bremen, Germany
| | - Tobias Bollhorst
- Advanced Ceramics, University of Bremen , Am Biologischen Garten 2, 28359 Bremen, Germany
| | - Laura Treccani
- Advanced Ceramics, University of Bremen , Am Biologischen Garten 2, 28359 Bremen, Germany
| | - Michael Maas
- Advanced Ceramics, University of Bremen , Am Biologischen Garten 2, 28359 Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen , 28359 Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen and Centre for Environmental Research and Sustainable Technology, Faculty 2 (Biology/Chemistry), University of Bremen , Leobener Strasse, NW2, 28359 Bremen, Germany
| | - Kurosch Rezwan
- Advanced Ceramics, University of Bremen , Am Biologischen Garten 2, 28359 Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen , 28359 Bremen, Germany
| |
Collapse
|
10
|
Gautier J, Munnier E, Soucé M, Chourpa I, Douziech Eyrolles L. Analysis of doxorubicin distribution in MCF-7 cells treated with drug-loaded nanoparticles by combination of two fluorescence-based techniques, confocal spectral imaging and capillary electrophoresis. Anal Bioanal Chem 2015; 407:3425-35. [PMID: 25749791 DOI: 10.1007/s00216-015-8566-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 01/23/2023]
Abstract
The intracellular distribution of the antiancer drug doxorubicin (DOX) was followed qualitatively by fluorescence confocal spectral imaging (FCSI) and quantitatively by capillary electrophoresis (CE). FCSI permits the localization of the major fluorescent species in cell compartments, with spectral shifts indicating the polarity of the respective environment. However, distinction between drug and metabolites by FCSI is difficult due to their similar fluorochromes, and direct quantification of their fluorescence is complicated by quantum yield variation between different subcellular environments. On the other hand, capillary electrophoresis with fluorescence detection (CE-LIF) is a quantitative method capable of separating doxorubicin and its metabolites. In this paper, we propose a method for determining drug and metabolite concentration in enriched nuclear and cytosolic fractions of cancer cells by CE-LIF, and we compare these data with those of FCSI. Significant differences in the subcellular distribution of DOX are observed between the drug administered as a molecular solution or as a suspension of drug-loaded iron oxide nanoparticles coated with polyethylene glycol. Comparative analysis of the CE-LIF vs FCSI data may lead to a tentative calibration of this latter method in terms of DOX fluorescence quantum yields in the nucleus and more or less polar regions of the cytosol.
Collapse
Affiliation(s)
- Juliette Gautier
- Universite Francois-Rabelais de Tours, EA6295 "Nanomedicaments et Nanosondes", 31 Avenue Monge, 37200, Tours, France
| | | | | | | | | |
Collapse
|
11
|
Zeng W, Liu P, Pan W, Singh SR, Wei Y. Hypoxia and hypoxia inducible factors in tumor metabolism. Cancer Lett 2014; 356:263-7. [PMID: 24508030 DOI: 10.1016/j.canlet.2014.01.032] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 01/01/2023]
Abstract
Because of the abnormal vasculature, most growing solid tumors contain regions that experience either acute or chronic hypoxia. However, tumor cells can maintain a high glycolytic rate even when there is enough oxygen supply. Hypoxia-inducible factors (HIFs) play crucial role in the response of tumor cells to this distinct microenvironment by shifting energy production from mitochondria towards glycolysis. In this review, we focus on the metabolism of tumor cell survival in hypoxic microenvironments. Furthermore, we also emphasize the mechanisms by which hypoxia and HIFs regulate tumor metabolism.
Collapse
Affiliation(s)
- Wen Zeng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China; Department of Joint and Bone Disease Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Peiyi Liu
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China; Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Weimin Pan
- Department of Human Movement Studies, Xi'an Physical Education University, Xi'an 710068, China
| | - Shree Ram Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Institutes of Health, National Cancer Institute, Frederick, MD 21702, USA.
| | - Yiyong Wei
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China; Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China.
| |
Collapse
|
12
|
Savatier J, Rharass T, Canal C, Gbankoto A, Vigo J, Salmon JM, Ribou AC. Adriamycin dose and time effects on cell cycle, cell death, and reactive oxygen species generation in leukaemia cells. Leuk Res 2012; 36:791-8. [PMID: 22417651 DOI: 10.1016/j.leukres.2012.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 01/10/2012] [Accepted: 02/20/2012] [Indexed: 01/03/2023]
Abstract
We investigate the relative importance of the different mechanisms of Adriamycin, an anthracycline, and their interrelations, in particular the link between cell cycle arrest, cell death, and generation of reactive oxygen species (ROS) that is suspected to be the origin of cardiotoxic side-effects. We introduced a lifetime fluorescence based technology and used videomicrofluorometry, two efficient analytical methods. We show that depending on the doses and time after incubation, ADR will not reach the same compartments (nucleus, mitochondria, cytosol) in the cells, having consequences on the production of ROS, growth arrest pathways and cell death pathways.
Collapse
Affiliation(s)
- Julien Savatier
- Institut Fresnel, Domaine Universitaire de St Jerôme, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
13
|
Enhancement of doxorubicin concentration in the M5076 ovarian sarcoma cells by cucurbitacin E co-treatment. Int J Pharm 2010; 383:186-91. [DOI: 10.1016/j.ijpharm.2009.08.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 08/27/2009] [Accepted: 08/28/2009] [Indexed: 11/19/2022]
|
14
|
Mhawi AA. Interaction of doxorubicin with the subcellular structures of the sensitive and Bcl-xL-overexpressing MCF-7 cell line: Confocal and low-energy-loss transmission electron microscopy. Micron 2009; 40:702-12. [DOI: 10.1016/j.micron.2009.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 05/09/2009] [Accepted: 05/09/2009] [Indexed: 10/20/2022]
|
15
|
Friche E, Skovsgaard T, Danø K. Multidrug resistance: drug extrusion and its counteraction by chemosensitizers. Eur J Haematol Suppl 2009; 48:59-67. [PMID: 3073961 DOI: 10.1111/j.1600-0609.1989.tb01239.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
16
|
Ota K, Ito K, Akahira JI, Sato N, Onogawa T, Moriya T, Unno M, Abe T, Niikura H, Takano T, Yaegashi N. Expression of Organic Cation Transporter SLC22A16 in Human Epithelial Ovarian Cancer. Int J Gynecol Pathol 2007; 26:334-40. [PMID: 17581421 DOI: 10.1097/01.pgp.0000236951.33914.1b] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The SLC22A16 is one of the newly isolated organic cation transporters, which is responsible for uptake and transport of adriamycin into cells. Adriamycin is considered to be an active agent for ovarian cancer. Recently, the benefit of adding adriamycin to the current standard regimen, paclitaxel and platinum, is evaluated to improve the outcome of patients with ovarian cancer. Therefore, we examined the expression of SLC22A16 in ovarian cancers. Twelve ovarian carcinoma cell lines were used for immunoblotting and reverse transcription-polymerase chain reaction to confirm the expression of SLC22A16 mRNA and protein. Five normal ovaries, 12 ovarian adenomas, and 94 ovarian cancer cases were obtained from patients after surgical therapy. The specimens were used for immunohistochemistry. The median value of relative SLC22A16 gene expression in cell lines derived from clear-cell adenocarcinoma was significantly higher than that in cell lines from other histologies (P < 0.001). Expression of SLC22A16 protein was also detected in cell lines derived from clear-cell adenocarcinoma. The SLC22A16 immunoreactivity was detected in 15 (16%) of 94 epithelial ovarian cancer, 1 (8.3%) of 12 benign adenomas, but 0 (0%) of 5 normal ovary cases. In ovarian cancer tissues, SLC22A16 immunoreactivity was detected in 2 (5%) of 38 serous adenocarcinoma, 1 (6.7%) of 15 endometrioid adenocarcinoma, 0 (0%) of 14 mucinous adenocarcinoma, and 12 (46.2%) of 26 clear-cell adenocarcinoma (P < 0.0001, clear-cell vs other histologies). In conclusion, SLC22A16 was abundantly expressed in clear-cell adenocarcinoma. Our results suggest that adriamycin-related chemicals that are taken up via SLC22A16 may have the potential to be effective against clear-cell adenocarcinoma.
Collapse
Affiliation(s)
- Kyoko Ota
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sato N, Ito K, Onogawa T, Akahira JI, Unno M, Abe T, Niikura H, Yaegashi N. Expression of Organic Cation Transporter SLC22A16 in Human Endometria. Int J Gynecol Pathol 2007; 26:53-60. [PMID: 17197897 DOI: 10.1097/01.pgp.0000225845.67245.b3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
SLC22A16 is one of newly isolated organic cation transporters, which is responsible for uptake and transport of adriamycin into cells. Adriamycin is one of the key drugs for treatment of endometrial cancer. Therefore, we examined expression of SLC22A16 in human endometrium and its disorders. Protein and mRNA expression levels of SLC22A16 were examined in 124 endometrial cancer specimens, 25 normal endometrial tissue samples (15 in proliferative phase, 10 in secretory phase), and 7 endometrial cancer cell lines using immunohistochemical analysis and reverse transcription-polymerase chain reaction. Changes in SLC22A16 mRNA expression level after progesterone exposure were also examined. Immunohistochemical analysis showed that SLC22A16 protein was highly expressed in endometrium during the normal secretory phase, but its level was significantly reduced in the proliferative phase. SLC22A16 protein was detected in 59 of 124 (48%) endometrial cancer specimens and 3 of 7 (43%) endometrial cancer cell lines. The mRNA levels measured by quantitative reverse transcription-polymerase chain reaction were comparable with levels of protein expression. Furthermore, SLC22A16 mRNA levels were increased in endometrial cancer cell lines in the presence of progesterone. In conclusion, SLC22A16 is expressed in various endometrial tissues. Its expression level is high during the secretory phase and may be regulated by progesterone. Our findings also suggest that it may be possible to use progestins to increase the response of endometrioid endometrial carcinoma with SLC22A16 expression to adriamycin-based chemotherapeutic regimens.
Collapse
Affiliation(s)
- Naoko Sato
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther 2006; 5:1275-9. [PMID: 16731760 DOI: 10.1158/1535-7163.mct-06-0024] [Citation(s) in RCA: 259] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The extracellular pH of tumor tissue is significantly lower than the extracellular pH of normal tissue, whereas the intracellular pH of both tissues is similar. In principle, extracellular acidity may be expected to enhance the intracellular uptake and cytotoxicity of weak acid chemotherapeutics that are membrane permeable in their uncharged state and inhibit the efficacy of weak bases. However, procedures for assessing the role of the gradient as a determinant of drug efficacy in vivo by altering the pH gradient may also alter drug availability and thus mask or exaggerate the effect of the gradient change. In the present study, we have altered the extracellular pH of tumors and compared the effect of the resultant pH gradient change on the efficacy of a weak acid versus a weak base. This experimental design gives rise to a change in the ratio of chlorambucil- to doxorubicin-induced tumor growth delay, independent of possible changes in drug availability. The extracellular pH of the 54A human tumor in NCr/Sed/nu/nu mice was altered by administration of 5 mg/g i.v. glucose. The resultant 0.2 pH unit increase in the tumor cell pH gradient gives rise to a predicted 2.3-fold increase in the ratio of chlorambucil to doxorubicin growth delay. The experimentally measured change in the growth delay ratio was 2.1. The results provide compelling evidence that the pH gradient in a determinant of the efficacy of weak electrolytes in the complex in vivo environment and may be exploited for the treatment of cancer.
Collapse
Affiliation(s)
- Leo E Gerweck
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | |
Collapse
|
19
|
Okabe M, Unno M, Harigae H, Kaku M, Okitsu Y, Sasaki T, Mizoi T, Shiiba K, Takanaga H, Terasaki T, Matsuno S, Sasaki I, Ito S, Abe T. Characterization of the organic cation transporter SLC22A16: A doxorubicin importer. Biochem Biophys Res Commun 2005; 333:754-62. [PMID: 15963465 DOI: 10.1016/j.bbrc.2005.05.174] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Accepted: 05/25/2005] [Indexed: 11/28/2022]
Abstract
Specific efflux transporters, such as P-glycoprotein, have been shown to confer drug resistance by decreasing the intracellular accumulation of anticancer drugs. Understanding influx transporters, as well as efflux transporters, is essential to overcome this resistance. We report the expression profile and pharmacological characterization of an organic cation transporter, SLC22A16. The results of our experiments indicate that SLC22A16 is a mediator of doxorubicin uptake in cancer cells. Quantitative real-time RT-PCR analyses show that SLC22A16 is expressed in primary samples taken from patients with acute leukemia. Xenopus oocytes injected with SLC22A16 cRNA import doxorubicin, a widely used anticancer drug for hematological malignancies, in a saturable and dose-dependent manner. The apparent Km value for doxorubicin import was 5.2+/-0.4 microM. In cytotoxic assays, stable transfectants of leukemic Jurkat cells overexpressing SLC22A16 cells became significantly more sensitive to doxorubicin (2 microM) treatment. Characterization of SLC22A16 will help in designing novel therapies targeting hematological malignancies.
Collapse
Affiliation(s)
- Mitsunori Okabe
- Division of General and Alimentary Tract Surgery, Department of Surgery, Tohoku University School of Medicine, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ikeda R, Che XF, Yamaguchi T, Ushiyama M, Zheng CL, Okumura H, Takeda Y, Shibayama Y, Nakamura K, Jeung HC, Furukawa T, Sumizawa T, Haraguchi M, Akiyama SI, Yamada K. Cepharanthine potently enhances the sensitivity of anticancer agents in K562 cells. Cancer Sci 2005; 96:372-6. [PMID: 15958061 PMCID: PMC11159796 DOI: 10.1111/j.1349-7006.2005.00057.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A major impediment to cancer treatment is the development of resistance by the tumor. P-glycoprotein (P-gp) and multidrug resistance protein 1 (MRP1) are involved in multidrug resistance. In addition to the extrusion of chemotherapeutic agents through these transporters, it has been reported that there are differences in the intracellular distribution of chemotherapeutic agents between drug resistant cells and sensitive cells. Cepharanthine is a plant alkaloid that effectively reverses resistance to anticancer agents. It has been previously shown that cepharanthine is an effective agent for the reversal of resistance in P-gp-overexpressing cells. Cepharanthine has also been reported to have numerous pharmacological effects besides the inhibition of P-gp. It has also been found that cepharanthine enhanced sensitivity to doxorubicin (ADM) and vincristine (VCR), and enhanced apoptosis induced by ADM and VCR of P-gp negative K562 cells. Cepharanthine changed the distribution of ADM from cytoplasmic vesicles to nucleoplasm in K562 cells by inhibiting the acidification of cytoplasmic organelles. Cepharanthine in combination with ADM should be useful for treating patients with tumors.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology
- Alkaloids/pharmacology
- Antibiotics, Antineoplastic/pharmacology
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis
- Benzylisoquinolines
- Doxorubicin/pharmacology
- Drug Interactions
- Drug Resistance, Multiple
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Pancreatic Neoplasms/pathology
- Tumor Cells, Cultured
- Vincristine/pharmacology
Collapse
Affiliation(s)
- Ryuji Ikeda
- Department of Clinical Pharmacy and Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hofland KF, Thougaard AV, Sehested M, Jensen PB. Dexrazoxane Protects against Myelosuppression from the DNA Cleavage–Enhancing Drugs Etoposide and Daunorubicin but not Doxorubicin. Clin Cancer Res 2005; 11:3915-24. [PMID: 15897593 DOI: 10.1158/1078-0432.ccr-04-2343] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The anthracyclines daunorubicin and doxorubicin and the epipodophyllotoxin etoposide are potent DNA cleavage-enhancing drugs that are widely used in clinical oncology; however, myelosuppression and cardiac toxicity limit their use. Dexrazoxane (ICRF-187) is recommended for protection against anthracycline-induced cardiotoxicity. EXPERIMENTAL DESIGN Because of their widespread use, the hematologic toxicity following coadministration of dexrazoxane and these three structurally different DNA cleavage enhancers was investigated: Sensitivity of human and murine blood progenitor cells to etoposide, daunorubicin, and doxorubicin +/- dexrazoxane was determined in granulocyte-macrophage colony forming assays. Likewise, in vivo, B6D2F1 mice were treated with etoposide, daunorubicin, and doxorubicin, with or without dexrazoxane over a wide range of doses: posttreatment, a full hematologic evaluation was done. RESULTS Nontoxic doses of dexrazoxane reduced myelosuppression and weight loss from daunorubicin and etoposide in mice and antagonized their antiproliferative effects in the colony assay; however, dexrazoxane neither reduced myelosuppression, weight loss, nor the in vitro cytotoxicity from doxorubicin. CONCLUSION Although our findings support the observation that dexrazoxane reduces neither hematologic activity nor antitumor activity from doxorubicin clinically, the potent antagonism of daunorubicin activity raises concern; a possible interference with anticancer efficacy certainly would call for renewed attention. Our data also suggest that significant etoposide dose escalation is perhaps possible by the use of dexrazoxane. Clinical trials in patients with brain metastases combining dexrazoxane and high doses of etoposide is ongoing with the aim of improving efficacy without aggravating hematologic toxicity. If successful, this represents an exciting mechanism for pharmacologic regulation of side effects from cytotoxic chemotherapy.
Collapse
|
22
|
Mikuni T, He G, Petryakov S, Fallouh MM, Deng Y, Ishihara R, Kuppusamy P, Tatsuta M, Zweier JL. In vivo detection of gastric cancer in rats by electron paramagnetic resonance imaging. Cancer Res 2004; 64:6495-502. [PMID: 15374960 DOI: 10.1158/0008-5472.can-04-0319] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Electron paramagnetic resonance imaging (EPRI) enables noninvasive spatial mapping of free radical metabolism and has recently been shown to provide in vivo physiologic information regarding alterations in the redox state of tumors and neoplastic tissues. With the use of nitroxide spin probes, it has been shown that certain tumors possess a highly reduced state. To determine whether EPRI can be used for early detection and visualization of gastric carcinoma based on its altered redox metabolism, studies were performed in a rat gastric cancer model induced by 1-methyl-3-nitro-1-nitrosoguanidine. Using a specialized 750 MHz resonator and EPRI instrument, a technique was developed for imaging nitroxide radicals in the whole stomach. In vivo three-dimensional EPRI of the stomach of rats with continuous intravenous administration of nitroxide 3-carboxamido-2,2,5,5-tetramethylpyrrolidine-N-oxyl (3-carbamoyl-proxyl) [3-CP] was performed. Whereas electron paramagnetic resonance images from untreated controls provide a uniform visualization of the stomach mucosa and wall, in the treated rats with gastric cancer, holes were present in the image at the locations of tumors. With localized spectroscopy, it was confirmed that the tumor regions were devoid of signal, and this was largely due to the presence of a more reduced state with rapid reduction of nitroxide. Pharmacokinetic studies indicated that 3-CP in tumors was rapidly reduced to an undetectable level, whereas the 3-CP levels in normal stomach tissue persisted. Near-infrared reflectance measurements of indocyanine green dye uptake indicated that there were no significant differences in tumor versus normal mucosal perfusion. From these results, we concluded that gastric cancer tumors could be distinguished from normal tissue based primarily on the marked difference in their rate of radical metabolism. Because alterations in cellular redox state and radical metabolism are of critical importance in tumor biology and treatment, this methodology should provide an important new tool for the study and visualization of gastric carcinoma and may also be of use in other cancer models.
Collapse
Affiliation(s)
- Tomiko Mikuni
- Center for Biomedical Electron Paramagnetic Resonance Spectroscopy and Imaging, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sadzuka Y, Sugiyama T, Suzuki H, Sawanishi H, Miyamoto KI. Increased effects of MPDAX, a novel xanthine derivative, on antitumor activity of doxorubicin. Toxicol Lett 2004; 150:341-9. [PMID: 15110086 DOI: 10.1016/j.toxlet.2004.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2003] [Revised: 02/10/2004] [Accepted: 02/10/2004] [Indexed: 10/26/2022]
Abstract
To clarify the effect of 1-methyl-3-propyl-7-N,N-dimethylpropylamide-xanthine (MPDAX) on doxorubicin (DOX) transport, we examined the efficacy of MPDAX as an amplifier of the antitumor activity of DOX in mice bearing tumors with different properties as to DOX transport across cell membranes. MPDAX significantly enhanced the DOX-induced antitumor activity on DOX-sensitive tumors. It is expected that the increase in antitumor activity caused by MPDAX contributes to the increased DOX concentration in tumors due to the MPDAX-induced change in DOX transport via the transporter expressed in sensitive tumor cells. In contrast, in M5076, a lower sensitive to DOX, MPDAX decreased the tumor weight by half at an otherwise ineffective dose of DOX. Furthermore, in P388/DOX, DOX has no effect, but MPDAX caused an elevation of the DOX-induced antitumor activity with an increase in the DOX concentration in the tumors. The results suggested that MPDAX is a novel amplifier for antitumor agents as it significantly increased the antitumor activity toward tumors with different properties. The DOX concentrations in the MPDAX + DOX group for all tumor lines were about two-fold those in the DOX alone group. Furthermore, MPDAX and DOX exhibited significant inhibitory effects on uridine and thymidine uptake. It is known that nucleoside transporters increase the membrane permeability of DOX. We speculated that MPDAX inhibits the cell membrane transport of uridine and thymidine via nucleoside transporters. MPDAX, acting via nucleoside transporters, increases the DOX-induced antitumor activity toward many tumor types and is an useful biochemical modulator.
Collapse
Affiliation(s)
- Yasuyuki Sadzuka
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Shizuoka 422-8526, Japan.
| | | | | | | | | |
Collapse
|
24
|
Mahoney BP, Raghunand N, Baggett B, Gillies RJ. Tumor acidity, ion trapping and chemotherapeutics. I. Acid pH affects the distribution of chemotherapeutic agents in vitro. Biochem Pharmacol 2003; 66:1207-18. [PMID: 14505800 DOI: 10.1016/s0006-2952(03)00467-2] [Citation(s) in RCA: 244] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Resistance to anti-cancer chemotherapies often leads to regional failure, and can be caused by biochemical and/or physiological mechanisms. Biochemical mechanisms include the overexpression of resistance-conferring proteins. In contrast, physiological resistance involves the tumor microenvironment, and can be caused by poor perfusion, hypoxia and/or acidity. This communication investigates the role of tumor acidity in resistance to a panel of chemotherapeutic agents commonly used against breast cancer, such as anthracyclines, taxanes, anti-metabolites and alkylating agents. The effects of pH on the cytotoxicity of these agents were determined, and ion trapping was confirmed by monitoring the effect of pH on the cellular uptake of radiolabeled anthracyclines. Furthermore, pH-dependent cytotoxicity and uptake were compared between parental drug sensitive MCF-7 cells and variants overexpressing p-glycoprotein (MDR-1) and Breast Cancer Resistance Protein. These data indicate that the magnitude of physiological resistance from pH-dependent ion trapping is comparable to biochemical resistance caused by overexpression of drug efflux pumps. Hence, microenvironment-based ion trapping is a significant barrier to anthracycline-based chemotherapy and can itself be a therapeutic target to enhance the efficacy of existing chemotherapies.
Collapse
Affiliation(s)
- Brent P Mahoney
- Department of Biochemistry and Cancer Biology Program, Arizona Cancer Center, University of Arizona Health Sciences Center, Tucson, AZ 85724-5024, USA
| | | | | | | |
Collapse
|
25
|
Shannon AM, Bouchier-Hayes DJ, Condron CM, Toomey D. Tumour hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat Rev 2003; 29:297-307. [PMID: 12927570 DOI: 10.1016/s0305-7372(03)00003-3] [Citation(s) in RCA: 398] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tissue hypoxia occurs where there is an imbalance between oxygen supply and consumption. Hypoxia occurs in solid tumours as a result of an inadequate supply of oxygen, due to exponential cellular proliferation and an inefficient vascular supply. It is an adverse prognostic indicator in cancer as it is associated with tumour progression and resistance to therapy. The expression of several genes controlling tumour cell survival are regulated by hypoxia, e.g., growth factors governing the formation of new blood vessels, and hypoxia-responsive transcription factors modulating the expression of genes, which promote tumour cell survival. This review outlines some of the pathways by which tumour hypoxia leads to chemotherapeutic resistance, directly due to lack of oxygen availability, and indirectly due to alterations in the proteome/genome, angiogenesis and pH changes. Some innovative therapies are also detailed which may potentially minimise or eliminate these problems associated with targeting solid tumours.
Collapse
Affiliation(s)
- Aoife M Shannon
- Department of Surgery, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, 9, Dublin, Ireland.
| | | | | | | |
Collapse
|
26
|
Nagai K, Nagasawa K, Ishimoto A, Fujimoto S. Pirarubicin is taken up by a uridine-transportable sodium-dependent concentrative nucleoside transporter in Ehrlich ascites carcinoma cells. Cancer Chemother Pharmacol 2003; 51:512-8. [PMID: 12679883 DOI: 10.1007/s00280-003-0569-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2002] [Accepted: 12/13/2002] [Indexed: 11/28/2022]
Abstract
PURPOSE We evaluated the contribution of a nucleoside transporter (NT) consisting of an equilibrative NT (ENT) and a concentrative Na(+)/nucleoside cotransporter (CNT) to the uptake of THP and DOX by mouse Ehrlich ascites carcinoma cells. METHODS. Transport experiments were performed using a silicone layer method. The expression of CNT isoforms was confirmed by RT-PCR analysis. RESULTS The effects of inhibition of the ENT inhibitors, nitrobenzylthioinosine (NBMPR) and nitrobenzylthioguanosine, on THP and DOX uptake by Ehrlich cells was negligible. THP uptake, but not DOX uptake, partially depended on an inwardly directed Na(+) gradient, and the uptake was inhibited by all the inhibitors of CNT examined. Furthermore, efflux of [(3)H]uridine from Ehrlich cells was stimulated by the addition of THP to the extracellular compartment, which was definitive evidence of CNT-mediated uptake of THP. The mRNA for CNT2, but not that for CNT3, was detected in Ehrlich cells, which is consistent with the characteristics of [(3)H]uridine uptake. In the cells, formycin B, a representative CNT2 ligand, had cis-inhibitory and trans-stimulatory effects on THP uptake. CONCLUSION These results demonstrate that THP, but not DOX, is taken up into Ehrlich cells partially via a uridine-transportable CNT.
Collapse
Affiliation(s)
- Katsuhito Nagai
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, 607-8414, Kyoto, Japan
| | | | | | | |
Collapse
|
27
|
Gerweck LE, Kozin SV, Stocks SJ. The pH partition theory predicts the accumulation and toxicity of doxorubicin in normal and low-pH-adapted cells. Br J Cancer 1999; 79:838-42. [PMID: 10070878 PMCID: PMC2362684 DOI: 10.1038/sj.bjc.6690134] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The accumulation and toxicity of the weak base doxorubicin has been investigated as a function of extracellular pH, intracellular pH and the cellular pH gradient in cells previously cultured under normal (pH 7.4) and low-pH (6.8) conditions. Low-pH-adapted cells exhibit transmembrane pH gradients which substantially differ from normal cells at the same extracellular pH. No relationship was obtained between intracellular pH and the uptake or toxicity of doxorubicin in the two cell types. In contrast, doxorubicin accumulation and toxicity increased with increasing extracellular pH in both normal and low-pH-adapted cells. However, at the same extracellular pH, drug cytotoxicity was more pronounced in normal than in low-pH-adapted cells. The difference in doxorubicin accumulation and cytotoxicity at the same extracellular pH was found to be dependent on the difference in the transmembrane pH gradient of the two cell types. As the cellular pH gradient differs between tumour and normal tissue, this observation suggests a basis for enhancing cellular drug uptake in either tissue type.
Collapse
Affiliation(s)
- L E Gerweck
- Department of Radiation Oncology, Massachusetts General Hospital, Boston 02114, USA
| | | | | |
Collapse
|
28
|
Demant EJ, Friche E. Kinetics of anthracycline accumulation in multidrug-resistant tumor cells: relationship to drug lipophilicity and serum albumin binding. Biochem Pharmacol 1998; 56:1209-17. [PMID: 9802333 DOI: 10.1016/s0006-2952(98)00255-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A multidrug-resistant Ehrlich ascites tumor cell line (EHR2/DNR+) was used to examine the membrane transport kinetics of lipophilic anthracycline derivatives in the presence of serum albumin. We present a model for theoretical data analysis with consideration of drug-albumin complex formation. For a set of five derivatives (doxorubicin, daunorubicin, 4-demethoxydaunorubicin, 4'-deoxy-4'-iododoxorubicin, and 13-dihydro-4'-deoxy-4'-iododoxorubicin), data were given on the rates of diffusional drug uptake, and membrane permeability coefficients of the noncharged molecules were estimated. Both the initial rates and the steady-state levels of drug uptake were found to decrease by addition of BSA at concentrations ranging from 5 to 75 mg/mL. For each drug, this effect of serum albumin could be accounted for by the altered distribution between free and protein-bound drug molecules in the bulk aqueous medium. A good fit of theoretical accumulation curves to the experimental data was obtained. It was concluded that a mathematical simulation method makes it possible to predict the uptake characteristics of lipophilic anthracycline compounds into tumor cells under serum conditions.
Collapse
Affiliation(s)
- E J Demant
- Department of Medical Biochemistry and Genetics, The Panum Institute, University of Copenhagen, Denmark
| | | |
Collapse
|
29
|
Bray PG, Ward SA. A comparison of the phenomenology and genetics of multidrug resistance in cancer cells and quinoline resistance in Plasmodium falciparum. Pharmacol Ther 1998; 77:1-28. [PMID: 9500157 DOI: 10.1016/s0163-7258(97)00083-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Plasmodium falciparum is the causative agent of the most deadly form of human malaria. Chemotherapy traditionally has been the main line of defense against this parasite, and chloroquine, the drug of choice, has been one of the most successful drugs ever developed. Unfortunately, the evolution and spread of resistance to chloroquine and other quinoline-containing drugs means that these compounds are now virtually useless in many endemic areas. Future prospects for the use of quinoline compounds improved considerably when it was demonstrated that chloroquine resistance could be circumvented in vitro by a number of structurally and functionally unrelated compounds such as verapamil and desipramine. The phenomenon of resistance reversal by compounds such as verapamil is also a key feature of drug resistance in mammalian cells, and this has raised the possibility that the underlying mechanisms of drug resistance of the two cell types could be similar. This hypothesis has prompted a large number of studies into the genetics and biochemistry of resistance to quinoline-containing drugs in P. falciparum. Both the genetic and the biochemical studies have raised issues of controversy and stimulated much debate. These issues are discussed in this review, in the context of a comparison with the genetics and biochemistry of multidrug resistance in mammalian cells.
Collapse
Affiliation(s)
- P G Bray
- Department of Pharmacology and Therapeutics, University of Liverpool, UK
| | | |
Collapse
|
30
|
C-SNARF-1 as a pHi fluoroprobe: discrepancies between conventional and intracellular data do not result from protein interactions. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 1997. [DOI: 10.1016/s1011-1344(96)07339-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
31
|
Goda K, Balkay L, Márián T, Trón L, Aszalós A, Szabó G. Intracellular pH does not affect drug extrusion by P-glycoprotein. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 1996; 34:177-82. [PMID: 8810535 DOI: 10.1016/1011-1344(95)07282-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The intracellular pH (pH(i)) of cells exhibiting multidrug resistance (MDR) related to the expression of the P-glycoprotein (Pgp) is often more alkaline than that of the parental cells, as also observed for the KB-V1/KB-3-1 system in this paper. The possible role of an elevated pH(i) in Pgp-related MDR has been investigated by shifting back the pH(i) of the MDR+ cells to a more acidic value using the mobile proton ionophore carbonylcyanide m-chlorophenylhydrazone (CCCP). The influence of CCCP-evoked delta pH(i) on relative daunorubicin (DNR) accumulation was similar in the case of several Pgp positive and negative cell lines, in view of flow cytometric and radioactive drug accumulation studies and measuring DNR levels in the medium in a flow-through system. Our data argue against a significant effect of pH(i) on Pgp pumping efficiency. However, an indirect connection between pH(i) regulation and the MDR phenotype is suggested by the fact that acidification of the external medium in the presence of verpamil could be observed exclusively in MDR+ cells.
Collapse
Affiliation(s)
- K Goda
- Department of Biophysics, University Medical School of Debrecen, Hungary
| | | | | | | | | | | |
Collapse
|
32
|
Ghauharali RI, Westerhoff HV, Dekker H, Lankelma J. Saturable P-glycoprotein kinetics assayed by fluorescence studies of drug efflux from suspended human KB8-5 cells. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1278:213-22. [PMID: 8593279 DOI: 10.1016/0005-2736(95)00224-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article describes a new and rapid method to determine the pumping rate of P-glycoprotein (P-gp) in intact cells. Multidrug resistant (MDR) human epidermoid carcinoma KB8-5 cells (containing P-gp) were loaded with daunorubicin (DNR) in the absence or in the presence of verapamil, sufficient to inhibit DNR pumping by P-gp. In either case, the cells were resuspended in medium devoid of DNR and the subsequent increase of the DNR fluorescence intensity was measured as a function of time. For cells loaded with the same amount of drug, the free cytosolic drug concentration (Ci(t)) was a unique function of the DNR medium concentration (Co(t)). The cellular drug content in the presence of verapamil decreased nonlinearly with decreasing extracellular drug concentration, indicating that the intracellular drug apparent distribution volume increased with decreasing cellular drug content. At each fluorescence intensity, we calculated the P-gp mediated (verapamil-inhibitable) DNR transport rate from the rate of increase of the DNR fluorescence intensity in the absence of verapamil minus the rate of increase of the DNR fluorescence intensity in the presence of verapamil. When plotted against the intracellular free drug concentration (as calculated from the total cellular drug content and a separately determined relation between the total cellular drug content and the intracellular free drug concentration: the apparent distribution volume), this P-gp mediated DNR transport rate showed saturation of P-gp at higher DNR concentrations. The results imply that P-gp mediated DNR transport is saturable (the value of Km is in the order of 1 microM).
Collapse
Affiliation(s)
- R I Ghauharali
- Department of Medical Oncology, Free University Hospital, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
33
|
Roepe PD. The role of the MDR protein in altered drug translocation across tumor cell membranes. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1241:385-405. [PMID: 8547302 DOI: 10.1016/0304-4157(95)00013-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- P D Roepe
- Program in Molecular Pharmacology and Therapeutics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021-6007, USA
| |
Collapse
|
34
|
Nielsen D, Maare C, Skovsgaard T. Influx of daunorubicin in multidrug resistant Ehrlich ascites tumour cells: correlation to expression of P-glycoprotein and efflux. Influence of verapamil. Biochem Pharmacol 1995; 50:443-50. [PMID: 7646548 DOI: 10.1016/0006-2952(95)00172-v] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Classic multidrug resistance is characterized by a decrease in the intracellular concentration of drugs in resistant cells as compared to sensitive cells. This is correlated with the presence of P-glycoprotein in the membrane. P-glycoprotein is responsible for an active efflux of drug. In this study we investigated the correlation between P-glycoprotein and influx of daunorubicin. Four Ehrlich ascites tumour cell lines selected in vivo for resistance to daunorubicin were investigated. The sublines EHR2/0.1, EHR2/0.2, passage no. 12 of EHR2/0.8, EHR2/0.4, and passage no. 72 of EHR2/0.8 were 6-, 6-, 5-, 33-, and 35-fold resistant to daunorubicin, respectively. All sublines overexpressed P-glycoprotein as determined with Western blot. Influx was measured over 40 sec. In glucose-enriched medium influx was significantly decreased in all but one of the resistant sublines. A correlation between P-glycoprotein, degrees of resistance, and influx was demonstrated in four sublines. Comparing influx experiments with efflux experiments (Nielsen et al., Biochem Pharmacol 1994, 47, 2125-2135) we found a linear relationship between influx and efflux in the resistant sublines (r = 0.97). Verapamil (5.5 microM, 11.0 microM) increased influx significantly in all resistant sublines, whereas the drug had no effect on sensitive cells. Verapamil (3.3 microM) increased influx in the EHR2/0.8 (passage no. 72) subline to the level of sensitive cells. Comparing this result with efflux experiments, verapamil was found to increase influx preferentially. Depletion of energy (medium without glucose including Na(+)-azide) increased influx in all resistant sublines. In EHR2/0.4 and EHR2/0.8 (passage no. 72) the influx, however, was still significantly decreased after depletion of energy. In these cells further addition of verapamil increased influx to the level of EHR2. These data were consistent with the hypothesis that P-glycoprotein effluxes drug directly from the plasma membrane.
Collapse
Affiliation(s)
- D Nielsen
- Department of Oncology R, Herlev Hospital, University of Copenhagen, Denmark
| | | | | |
Collapse
|
35
|
Nielsen D, Maare C, Skovsgaard T. Kinetics of daunorubicin transport in Ehrlich ascites tumor cells with different expression of P-glycoprotein. Biochem Pharmacol 1994; 47:2125-35. [PMID: 7913318 DOI: 10.1016/0006-2952(94)90247-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The classical multidrug resistance (MDR) phenotype is characterized by a decrease in the intracellular drug concentration in resistant cells as compared to sensitive cells. P-glycoprotein (P-gp) is thought to be responsible for an active efflux of lipophilic drugs. Four Ehrlich ascites tumor cell lines selected in vivo for resistance to daunorubicin (DNR) and their sensitive counterpart were investigated. The resistant sublines EHR2/0.1, EHR2/0.2, EHR2/0.4, and EHR2/0.8 were developed by treatment of tumor bearing mice with DNR 0.1, 0.2, 0.4, and 0.8 mg/kg x 4 weekly, respectively. One passage from EHR2/0.1, EHR2/0.2, and EHR2/0.4 and two passages from EHR2/0.8 were investigated. Western blot analysis showed significantly different amounts of P-gp (a 6-fold variation). Efflux of DNR in a drug free medium was investigated with and without presence of verapamil (VER). Efflux from sensitive and resistant cells was described by mono- and bi-exponential kinetics, respectively. In all cases but one, a correlation between resistance, expression of P-gp, P-gp mediated efflux capacity and effect of VER was established. In passage No. 12 of EHR2/0.8, however, a high expression of P-gp was found in spite of a low degree of resistance and a low efflux capacity. In this subline the effect of VER did not correlate to the expression of P-gp. Active efflux seemed to be saturable and was suggested to constitute the major route of efflux in MDR cells. A dose-response relationship was established for the effect of VER on efflux. In conclusion, the results support that P-gp acts as a drug efflux pump. No simple correlation, however, could be established between P-gp and drug transport in all the investigated cell lines. Other factors which might influence transmembranous transportation of DNR are suggested. The active efflux capacity of the cell lines seemed to determine the degree of resistance and the sensitivity to circumvention by VER.
Collapse
Affiliation(s)
- D Nielsen
- Department of Oncology R, Herlev Hospital, University of Copenhagen, Denmark
| | | | | |
Collapse
|
36
|
Simon SM, Schindler M. Cell biological mechanisms of multidrug resistance in tumors. Proc Natl Acad Sci U S A 1994; 91:3497-504. [PMID: 7909602 PMCID: PMC43607 DOI: 10.1073/pnas.91.9.3497] [Citation(s) in RCA: 256] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Multidrug resistance (MDR) is a generic term for the variety of strategies tumor cells use to evade the cytotoxic effects of anticancer drugs. MDR is characterized by a decreased sensitivity of tumor cells not only to the drug employed for chemotherapy but also to a broad spectrum of drugs with neither obvious structural homology nor common targets. This pleiotropic resistance is one of the major obstacles to the successful treatment of tumors. MDR may result from structural or functional changes at the plasma membrane or within the cytoplasm, cellular compartments, or nucleus. Molecular mechanisms of MDR are discussed in terms of modifications in detoxification and DNA repair pathways, changes in cellular sites of drug sequestration, decreases in drug-target affinity, synthesis of specific drug inhibitors within cells, altered or inappropriate targeting of proteins, and accelerated removal or secretion of drugs.
Collapse
Affiliation(s)
- S M Simon
- Laboratory of Cellular Biophysics, Rockefeller University, New York, NY 10021
| | | |
Collapse
|
37
|
Simon S, Roy D, Schindler M. Intracellular pH and the control of multidrug resistance. Proc Natl Acad Sci U S A 1994; 91:1128-32. [PMID: 8302842 PMCID: PMC521467 DOI: 10.1073/pnas.91.3.1128] [Citation(s) in RCA: 189] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Many anticancer drugs are classified as either weak bases or molecules whose binding to cellular structures is pH dependent. Accumulation of these drugs within tumor cells should be affected by transmembrane pH gradients. Indeed, development of multidrug resistance (MDR) in tumor cells has been correlated with an alkaline shift of cytosolic pH. To examine the role of pH in drug partitioning, the distribution of two drugs, doxorubicin and daunomycin, was monitored in fibroblasts and myeloma cells. In both cell types the drugs rapidly accumulated within the cells. The highest concentrations were measured in the most acidic compartments--e.g., lysosomes. Modifying the cellular pH in drug-sensitive cells to mimic reported shifts in MDR caused an immediate change in the cellular drug concentration. Drug accumulation was enhanced by acidic shifts and reversed by alkaline shifts. All of these effects were rapid and reversible. These results demonstrate that the alkaline shift observed in MDR is sufficient to prevent the accumulation of chemotherapeutic drugs independent of active drug efflux.
Collapse
Affiliation(s)
- S Simon
- Laboratory of Cellular Biophysics, Rockefeller University, New York, NY 10021
| | | | | |
Collapse
|
38
|
Mülder HS, van Grondelle R, Westerhoff HV, Lankelma J. A plasma membrane 'vacuum cleaner' for daunorubicin in non-P-glycoprotein multidrug-resistant SW-1573 human non-small cell lung carcinoma cells. A study using fluorescence resonance energy transfer. EUROPEAN JOURNAL OF BIOCHEMISTRY 1993; 218:871-82. [PMID: 8281939 DOI: 10.1111/j.1432-1033.1993.tb18443.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A multidrug resistant (MDR) human non-small cell lung carcinoma cell line, SW-1573/2R120 (2R120), not containing the drug-efflux pump P-glycoprotein (PgP), has been studied for the transport of daunorubicin (DN) across the cellular plasma membrane. Earlier, reduced initial DN-uptake rates and lower cellular DN steady-state concentrations were found for this cell line, when it was compared to the SW-1573 wild-type cell line. This finding was an indication for the presence of another cellular drug-efflux pump. However, we found similar DN-efflux rates in drug-free medium for the two cell lines, while for Pgp-containing MDR SW-1573/2R160 (2R160) cells the efflux rate was increased compared to wild-type cells. In order to elucidate differences in DN transport across the cellular plasma membrane, the association of DN with plasma membranes of intact cells was investigated, using fluorescence-resonance-energy transfer. For this purpose, the plasma-membrane probe 1-(4-trimethyl-ammoniumphenyl)-6-phenyl-1,3,5-hexatriene (TMA-DPH) was chosen since, because of the overlap between the emission spectrum of TMA-DPH and the excitation spectrum of DN, transfer of energy can be achieved from TMA-DPH to DN. Cells were loaded with TMA-DPH and, after addition of 10 microM DN, the TMA-DPH fluorescence was quenched. Rapid initial quenching proved to be similar in the MDR 2R160 (Pgp-containing) cells and in the SW-1573 wild-type cells (21 +/- 1% and 20 +/- 2%, respectively), but was less in the MDR 2R120 cells not containing Pgp (14 +/- 1%). This finding correlated with a lowered amount of DN dissolved in the plasma membrane of 2R120 cells. We interpret these data to be the result of a 'vacuum-cleaner' pumping system other than Pgp which removes DN from a plasma membrane compartment and equilibrates relatively slowly with the interior of the cell.
Collapse
Affiliation(s)
- H S Mülder
- Department of Oncology, Free University Hospital, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
39
|
Coley HM, Twentyman PR, Workman P. The efflux of anthracyclines in multidrug-resistant cell lines. Biochem Pharmacol 1993; 46:1317-26. [PMID: 7902089 DOI: 10.1016/0006-2952(93)90094-d] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In order to address the association of enhanced drug efflux with the multidrug-resistant (MDR) phenotype, we have studied the cellular pharmacokinetics of anthracyclines in the P-glycoprotein (Pgp)-positive MDR cell lines H69/LX4 (human small cell lung cancer) and EMT6/AR1.0 (mouse mammary tumour). Both doxorubicin (DOX) and daunorubicin (DNR) were accumulated to a lesser extent and effluxed at a higher rate by MDR cells than by their drug-sensitive counterparts. In contrast, the 9-alkyl substituted compound, aclacinomycin A (ACL), was accumulated and effluxed from parent and MDR cells at an identical rate. In experiments designed to examine energy-dependent efflux, DOX and DNR were shown to be efficiently effluxed against the concentration gradient in the presence of glucose. However, in the same experiments the analogues ACL and Ro 31-3294 (9-alkyl and morpholinyl substituted), which have previously been shown to retain activity against MDR cell lines, were accumulated and effluxed at identical rates in parent and MDR EMT6 cells. Hence, 9-alkyl and morpholinyl substituted compounds appear to behave less favourably as substrates for energy-driven drug efflux by Pgp-positive MDR cells than do DOX or DNR. Resistance modifiers verapamil and cyclosporin A appeared to abolish energy-dependent efflux for DOX and DNR in both the EMT6 and H69 MDR lines whereas they had no effect on the cellular efflux of ACL. The altered cellular pharmacology in MDR cell lines may provide a rational basis for the use of modified anthracycline analogues (e.g. 9-alkyl and morpholinyl (substituted) and resistance of modifying agent in the treatment of tumours expressing a Pgp-mediated phenotype.
Collapse
Affiliation(s)
- H M Coley
- MRC Clinical Oncology and Radiotherapeutics Unit, Cambridge, U.K
| | | | | |
Collapse
|
40
|
Jähde E, Roszinski S, Volk T, Glüsenkamp KH, Wiedemann G, Rajewsky MF. Metabolic response of AH13r rat tumours to cyclophosphamide as monitored by pO2 and pH semi-microelectrodes. Eur J Cancer 1993; 29A:116-22. [PMID: 1445727 DOI: 10.1016/0959-8049(93)90587-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The composition of the microenvironment has an important influence on the cellular response to cytotoxic agents. Using pH and pO2 semi-microelectrodes, we have monitored metabolic changes in AH13r rat tumours as a function of time after subcurative chemotherapy. Prior to therapy, tumours contained large areas considered hypoxic (mean pO2 approximately 4 mmHg) and are characterised by a marked accumulation of acidic metabolites (mean pH 6.65). Administration of cyclophosphamide (40 mg/kg body weight) resulted in tumour regression to 15% of pretreatment volumes and a growth delay of 12 days. Concomitant with volume reduction, tumours became reoxygenated (mean pO2 approximately 7 mmHg), with maximum values being reached within 2-4 days, paralleled by a shift of pH to more alkaline values (0.17 U on average). These changes coincided with the development of subtotal necrosis. During early tumour regrowth, the pH and pO2 histograms returned to control values. These data corroborate and extend the results of previous studies in which noninvasive techniques had been applied for the monitoring of treatment-induced metabolic changes in malignant tumours in vivo. In addition, these results support the notion that the effectiveness of anticancer therapy might be improved by selecting and scheduling therapeutic agents in consideration of physiological changes caused by preceding courses of treatment.
Collapse
Affiliation(s)
- E Jähde
- Institute of Cell Biology (Cancer Research), University of Essen Medical School, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Spoelstra EC, Westerhoff HV, Dekker H, Lankelma J. Kinetics of daunorubicin transport by P-glycoprotein of intact cancer cells. ACTA ACUST UNITED AC 1992; 207:567-79. [PMID: 1353020 DOI: 10.1111/j.1432-1033.1992.tb17083.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Drug permeation across the plasma membrane of multidrug-resistant cells depends on the kinetics of the P-glycoprotein-mediated pump activity as well as on the passive permeation of the drug. We here demonstrate a method to characterize kinetically the pump in intact cells. To this purpose, we examined the membrane-transport properties of daunorubicin in various sensitive cancer cell lines and in their multidrug resistant (MDR) counterparts. First, we determined the passive permeability coefficient for daunorubicin. Then, using a flow-through system, the drug flux into the cell was measured after inhibition of the P-glycoprotein-mediated efflux pump. Combining the two results allowed us to calculate the intracellular free concentration of the drug. In the steady-state, the pump rate must equal the net rate of passive diffusion of the drug and, therefore, the same experiments gave us the pumping rate of daunorubicin. These experiments were then repeated at various extracellular drug concentrations. By plotting the pumping rate versus the intracellular drug concentration, we then characterized the P-glycoprotein kinetically. Four independent methods were used to measure the passive permeability coefficient for the cell line A2780. Similar values were obtained. Maximal pump rates (Vmax) showed a good correlation with the amount of P-glycoprotein in the cell lines used. We obtained saturation curves for the variation of the pump rates with the intracellular daunorubicin concentrations. These curves were typical for positive cooperativity, which provides evidence that at least two binding sites for daunorubicin are present on the active transport system of daunorubicin. The apparent Km values for P-glycoprotein-mediated transport, the intracellular free cytosolic daunorubicin concentrations at half-maximal velocity for the cell lines used, were approximately 1.5 microM. Except for the cell lines with the highest amount of P-glycoprotein, the passive efflux rate of daunorubicin proved to be a substantial part of the total daunorubicin efflux rate for the cell lines used. In cell lines with relatively low levels of P-glycoprotein, passive daunorubicin efflux was even the main route of daunorubicin transport from the cells, determining the intracellular steady-state concentrations of daunorubicin.
Collapse
Affiliation(s)
- E C Spoelstra
- Department of Medical Oncology, Free University Hospital, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
42
|
Bonner JA, Christianson TJ, Lawrence TS. Interaction of buthionine sulfoximine and the stabilization of DNA-topoisomerase II complexes by doxorubicin. Int J Radiat Oncol Biol Phys 1992; 22:519-23. [PMID: 1310496 DOI: 10.1016/0360-3016(92)90866-g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although it has been shown previously that the depletion of cellular thiols increases doxorubicin cytotoxicity, the mechanism of sensitization is not clear. To study this question, the effect of D,L-buthionine-S,R-sulfoximine (BSO) on doxorubicin cytotoxicity and the stabilization of DNA-topoisomerase II complexes (cleavable complexes) was investigated in V79 cells. Incubations with BSO (10 mM) were for 5 hr beginning 4 hr prior to doxorubicin exposure since a 4 hr incubation with 10 mM BSO is known to decrease glutathione levels below 5% of control V79 cells. These BSO pre-treatment increased doxorubicin cytotoxicity. At doxorubicin concentrations of 5 micrograms/ml, BSO resulted in an 8-10 fold decrease in surviving cells, compared to cells exposed to doxorubicin alone. It was determined that BSO pre-treatments did not affect the accumulation of doxorubicin into the cell, the rate of cleavable complex stabilization by doxorubicin, or the rate of dissociation of stabilized cleavable complexes. These data suggest that BSO-induced doxorubicin sensitization occurs at a step following the stabilization of cleavable complexes or by an independent mechanism.
Collapse
Affiliation(s)
- J A Bonner
- Mayo Clinic, Radiation Oncology, Rochester, MN 55905
| | | | | |
Collapse
|
43
|
Carpentier Y, Gorisse MC, Desoize B. Evaluation of a method for detection of cells with reduced drug retention in solid tumours. ACTA ACUST UNITED AC 1992; 13:630-7. [PMID: 1360399 DOI: 10.1002/cyto.990130611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A method for detection of cells with reduced drug retention was evaluated in solid tumours. After a 1 h incubation with daunorubicin (DNR), the right angle scatter (RAS), forward angle scatter (FAS), and specific fluorescence (Fluo) were measured in sensitive and resistant cells; only Fluo was related qualitatively, but not quantitatively, to resistance. Various incubation conditions were examined. When the pH of the incubation medium increased, the DNR retention increased in sensitive and resistant cells. In contrast, when the cell concentration increased, the DNR retention decreased. Using sensitive and resistant cell lines, a proportion of resistant cells lower than 10% can be detected in a mixture. To analyse cells from solid tumours, the cells were dissociated by repeated fine needle aspirations. Tumours from 22 patients have been processed with this technique; 8 samples were classified as S (sensitive); 2 as R (resistant); and 12 as I (intermediate). Further experiments were run to study and improve the method. Another method of detection of dead cells was tested. The intra-assay variability of the technique was found to be less than 10%. When the study was performed with different fragments of the same tumour, the variation, corresponding to the tumour heterogeneity, rose to 21 to 36%. The inter-assay reproducibility was too bad, so a variant of this technique has been adapted, using verapamil or cyclosporin A, which is able to block DNR efflux; this new method allows tumour cells to be used as their own controls.
Collapse
Affiliation(s)
- Y Carpentier
- Institut Jean Godinot, GIBSA, BP 171, Reims, France
| | | | | |
Collapse
|
44
|
Ramu A, Ramu N, Gorodetsky R. Reduced ouabain-sensitive potassium entry as a possible mechanism of multidrug-resistance in P388 cells. Biochem Pharmacol 1991; 42:1699-704. [PMID: 1930295 DOI: 10.1016/0006-2952(91)90504-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multidrug-resistant P388 cells were found to be resistant also to a variety of ammonium, phosphonium and arsonium compounds. As previously shown for anthracyclines and vinca alkaloids, the resistance to the permanently charged lipophilic cationic compounds could be circumvented by verapamil. Relative to drug-sensitive cells, K+ uptake and plasma membrane Mg-ATPase activity in multidrug-resistant cells are ouabain resistant. The intracellular K+ concentration in drug-resistant cells is maintained at a normal level by increased activity of the furosemide sensitive transport system. It is suggested that the reduced activity of the electrogenic Na(+)-K+ pump in multidrug-resistant, cells could result in a lower transmembrane potential and therefore reduced accumulation of cationic lipophilic compounds.
Collapse
Affiliation(s)
- A Ramu
- Department of Oncology, Hadassah University Hospital, Jerusalem, Israel
| | | | | |
Collapse
|
45
|
Spoelstra EC, Dekker H, Schuurhuis GJ, Broxterman HJ, Lankelma J. P-glycoprotein drug efflux pump involved in the mechanisms of intrinsic drug resistance in various colon cancer cell lines. Evidence for a saturation of active daunorubicin transport. Biochem Pharmacol 1991; 41:349-59. [PMID: 1671638 DOI: 10.1016/0006-2952(91)90531-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We studied the resistance of colon tumors to anticancer agents in vitro. Using daunorubicin (DN), a number of cellular parameters which normally indicate acquired or multidrug resistance (MDR), were compared for several human wild-type colon cell lines, i.e. HT29, SW1116 and COLO 320, and the murine colon cell line C-26. The sensitive/MDR human ovarian cancer cell line couple A2780/2780AD was used as a reference. The amount of P-glycoprotein (P-gp) was in the order HT29, A2780 less than or equal to SW1116 less than C26 less than or equal to COLO 320 less than 2780AD. The MDR modifiers verapamil, Cremophor EL, cyclosporin A and Ro 11-2933/001 had significant effects on DN cytotoxicity, total DN accumulation and efflux, only if P-gp was present. A flow-through system was used to study the mechanism of DN transport. For the first time, evidence for saturation of an active transport of DN from the cells is reported. We discussed the possible presence of cooperative activity between at least two binding sites on the protein responsible for DN efflux, likely to be P-gp.
Collapse
Affiliation(s)
- E C Spoelstra
- Department of Medical Oncology, Free University Hospital, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
46
|
Bouchelouche P, Friche E, Sehested M, Jensen PB, Skovsgaard T. Cytosolic free Ca2+ in daunorubicin and vincristine resistant Ehrlich ascites tumor cells. Drug accumulation is independent of intracellular Ca2+ changes. Biochem Pharmacol 1991; 41:243-53. [PMID: 1899193 DOI: 10.1016/0006-2952(91)90483-l] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The possible role of intracellular calcium on daunorubicin (DNR) accumulation in wild-type (EHR2) and multi-drug resistant (MDR) Ehrlich ascites tumor cell subline was investigated. DNR accumulation was not enhanced either by increasing the concentration of cellular calcium with the calcium ionophore ionomycin nor by chelating the cytosolic free Ca2+ by the membrane permeable Ca2(+)-buffering agents BAPTA or MAPTAM. No effect was observed in the presence of extremely low extracellular calcium concentration that prevent transmembrane calcium influx or when the cells were calcium depleted using EGTA and ionomycin. Using the fluorescent Ca2+ indicator fura-2 it is further shown that both drug-resistant daunorubicin (EHR2/DNR+) and vincristine (EHR/VCR+) sublines had lower (50-80 nM) concentration of cytosolic free calcium ([Ca2+]i) compared to their corresponding wild-type parenteral tumors (140-180 nM). In calcium free medium, however, no significant difference was found, all cell lines having a [Ca2+]i of 60-80 nM. Furthermore, the total amount of Ca2+ released to the cytosol with 10 microM ionomycin and 5 mM EGTA was 3-4-fold higher in EHR2 than in EHR2/DNR+ or EHR2/VCR+. Mobilization of Ca2+ with 1 microM ionomycin was almost identical in the presence and absence of Ca2+ in the extracellular medium in EHR2 as well as in EHR2/DNR+ suggesting that the increase in [Ca2+]i is mainly due to discharge of Ca2+ from intracellular stores. Furthermore, the total cell calcium [Ca2+]t concentration was slightly higher in EHR2/DNR+ and EHR2/VCR+ cells compared to EHR2. Incubation of the cells with the Ca2(+)-channel blocker verapamil or the intracellular Ca2(+)-antagonist TMB-8 causes depression of the Ca2(+)-response in terms of rise in [Ca2+]i caused by ionomycin. Sorcin, a major calcium-binding protein (Mr 22 kDa), is shown to be overproduced in EHR2/DNR+ cells. The overproduction of this protein in resistant cells may be related to the difference in the intracellular calcium observed in this study. Thus, though handling of Ca2+ is different in wild-type and MDR cell lines, our data suggest that calcium is not involved directly in drug transport processes and the level of Ca2+ per se have no influence on drug accumulation.
Collapse
Affiliation(s)
- P Bouchelouche
- Dept. of Clinical Chemistry, Herlev University Hospital, Denmark
| | | | | | | | | |
Collapse
|
47
|
Demant EJ, Sehested M, Jensen PB. A model for computer simulation of P-glycoprotein and transmembrane delta pH-mediated anthracycline transport in multidrug-resistant tumor cells. BIOCHIMICA ET BIOPHYSICA ACTA 1990; 1055:117-25. [PMID: 2242381 DOI: 10.1016/0167-4889(90)90111-p] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Anthracycline resistance in multidrug-resistant (MDR) tumor cells is due in part to a reduced cellular drug accumulation. Using a simple kinetic model and numerical computer simulations, we have analyzed mathematically the following possible mechanisms controlling fluxes of the membrane permeable anthracyclines in MDR cells: (1) active outward transport via a specific drug transporter (P-glycoprotein), (2) exocytotic drug export via the endosomal vesicle system, and (3) pH-gradients across the plasma membrane. Model calculations were based on morphometric and kinetic data previously presented in the literature for daunorubicin transport in wild-type Ehrlich ascites tumor cells (EHR2) and the corresponding daunorubicin (DNR)-resistant cell line EHR2/DNR+. The results confirm the possible importance of the cell-surface pH in controlling DNR accumulation in the cells. With P-glycoprotein as the main efflux pump, a catalytic constant of the protein greater than 40 mol DNR transported/mol protein per min is predicted by the model calculations. Changes in the drug binding affinity of P-glycoprotein (Km = 10(-9)-10(-6) M) is of little importance in influencing its effectiveness to reduce DNR accumulation, which could explain the broad substrate specificity of the MDR efflux pump system. The conditions to evaluate unidirectional fluxes of DNR across the plasma membrane in cells with active P-glycoprotein are defined. An efflux mechanism which relies solely on pH-dependent drug trapping in a pH 5 endosomal compartment by a simple diffusion process followed by exocytosis, appears inadequate to account for the high rate of DNR efflux in EHR2/DNR+ cells.
Collapse
Affiliation(s)
- E J Demant
- Department of Biochemistry C, Panum Institute, University of Copenhagen, Denmark
| | | | | |
Collapse
|
48
|
Sehested M, Skovsgaard T, Jensen PB, Demant EJ, Friche E, Bindslev N. Transport of the multidrug resistance modulators verapamil and azidopine in wild type and daunorubicin resistant Ehrlich ascites tumour cells. Br J Cancer 1990; 62:37-41. [PMID: 1975202 PMCID: PMC1971743 DOI: 10.1038/bjc.1990.225] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Verapamil has been proposed to modulate the multidrug resistance phenotype by competitive inhibition of an energy dependent efflux of cytotoxic drug. However, the accumulation of both 14C-verapamil and 3H-verapamil was similar in wild type EHR2 and multidrug resistant EHR2/DNR+ Ehrlich ascites cells, and was much less in both cell lines in energy deprived medium than in medium containing glucose. Azidopine accumulation was also similar in both EHR2 and EHR2/DNR+ cells but, in contrast to verapamil, did not differ significantly with changes in cellular energy levels. Azidopine photolabelled a 170 kDa protein in EHR2/DHR+ plasma membrane vesicles which was immunoprecipitated by monoclonal antibody towards P-glycoprotein. Azidopine increased daunorubicin accumulation and modulated vincristine resistance in EHR2/DNR+ cells in a similar fashion to verapamil. Azidopine photolabelling was inhibited by vincristine and verapamil, but not by daunorubicin. Vincristine, but not daunorubicin, was able to increase both azidopine and verapamil accumulation in EHR2/DNR+ cells only. Finally, though both verapamil and azidopine are a substrate for P-glycoprotein in EHR2/DNR+ cells, they do not themselves appear to be transported by the multidrug resistance efflux mechanism to any significant extent in these cells.
Collapse
|
49
|
Jähde E, Glüsenkamp KH, Rajewsky MF. Protection of cultured malignant cells from mitoxantrone cytotoxicity by low extracellular pH: a possible mechanism for chemoresistance in vivo. Eur J Cancer 1990; 26:101-6. [PMID: 2138903 DOI: 10.1016/0277-5379(90)90290-a] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In malignant tumors the distribution of pH values is shifted to lower values (range, pH 5.8-7.4) as compared to normal tissues (range, pH 6.9-7.4) or peripheral blood (pH 7.35-7.45). We have investigated whether the cytotoxic effect of the anthracenedione anti-cancer drug mitoxantrone (MX) on malignant cells in culture is dependent on changes of extracellular pH. The clonogenic fraction of M1R rat mammary carcinoma cells was measured after exposure to MX at an extracellular pH (pHe) of 6.5-7.4. At pHe 6.8 (approximately the average pH measured in a number of malignant tumors in vivo) the clonogenic fraction of M1R cells exposed to MX (0.1 microgram/ml) only decreased to 1 X 10(-1) as compared to 2.5 X 10(-4) at pHe 7.4, corresponding to a 400-fold inhibition of MX cytotoxicity at reduced environmental pH. The H+ ion-mediated resistance of M1R cells to MX could be partially reversed by verapamil, suggesting that a reduced microenvironmental pH possibly interferes with intracellular MX accumulation. Therefore, drugs like MX may not be effective in the elimination of cells in acidic tumor areas. Moreover, investigations on anti-cancer drug activity in vitro at what is frequently referred to as 'physiological pH' may be irrelevant in terms of the cytotoxic effects of the respective agents at the pH values prevailing in malignant tissues in vivo.
Collapse
Affiliation(s)
- E Jähde
- Medizinische Universitätsklinik, Abteilung Innere Medizin II, Tübingen, F.R.G
| | | | | |
Collapse
|
50
|
Abstract
The mechanism that underlies doxorubicin-induced radiosensitization was investigated in a cell culture system. When V79 hamster cells were treated with doxorubicin, radiosensitization occurred as illustrated by an increase in alpha and a reduction in the mean inactivation dose (D) of the radiation survival curve. Under control conditions the radiation survival curve showed an alpha of 0.010 +/- 0.02 and a D of 4.4 +/- 0.2. After exposure to doxorubicin (1.5 micrograms/ml for 1 h prior to irradiation), alpha increased to 0.28 +/- 0.04 and D decreased to 3.0 +/- 0.3. Similar mean inactivation doses were found for doxorubicin doses of 0.75, 1.5 and 3.0 micrograms/ml, indicating that enhancement was not dose-dependent in the dose range studied. The influence of doxorubicin on radiation-induced DNA double-strand breaks (dsb) was then assessed under the same conditions as were used in the cell survival studies. Pretreatment with doxorubicin minimally affected radiation-induced dsb. However, doxorubicin increased dsb by 1.25 +/- 0.07 under conditions in which repair was allowed to proceed for 45 min. These results show that doxorubicin inhibits the repair of radiation-induced dsb. Doxorubicin is known to change DNA conformation by the stabilization of DNA topoisomerase II complexes. It is possible that these changes alter the ability of repair enzymes to recognize and correct radiation-induced damage.
Collapse
Affiliation(s)
- J A Bonner
- University of Michigan Medical Center, Department of Radiation Oncology, Ann Arbor 48109
| | | |
Collapse
|