1
|
Ramisetty BS, Yang S, Dorlo TPC, Wang MZ. Determining tissue distribution of the oral antileishmanial agent miltefosine: a physiologically-based pharmacokinetic modeling approach. Antimicrob Agents Chemother 2024; 68:e0032824. [PMID: 38842325 PMCID: PMC11232387 DOI: 10.1128/aac.00328-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024] Open
Abstract
Miltefosine (MTS) is the only approved oral drug for treating leishmaniasis caused by intracellular Leishmania parasites that localize in macrophages of the liver, spleen, skin, bone marrow, and lymph nodes. MTS is extensively distributed in tissues and has prolonged elimination half-lives due to its high plasma protein binding, slow metabolic clearance, and minimal urinary excretion. Thus, understanding and predicting the tissue distribution of MTS help assess therapeutic and toxicologic outcomes of MTS, especially in special populations, e.g., pediatrics. In this study, a whole-body physiologically-based pharmacokinetic (PBPK) model of MTS was built on mice and extrapolated to rats and humans. MTS plasma and tissue concentration data obtained by intravenous and oral administration to mice were fitted simultaneously to estimate model parameters. The resulting high tissue-to-plasma partition coefficient values corroborate extensive distribution in all major organs except the bone marrow. Sensitivity analysis suggests that plasma exposure is most susceptible to changes in fraction unbound in plasma. The murine oral-PBPK model was further validated by assessing overlay of simulations with plasma and tissue profiles obtained from an independent study. Subsequently, the murine PBPK model was extrapolated to rats and humans based on species-specific physiological and drug-related parameters, as well as allometrically scaled parameters. Fold errors for pharmacokinetic parameters were within acceptable range in both extrapolated models, except for a slight underprediction in the human plasma exposure. These animal and human PBPK models are expected to provide reliable estimates of MTS tissue distribution and assist dose regimen optimization in special populations.
Collapse
Affiliation(s)
| | - Sihyung Yang
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas, USA
| | - Thomas P. C. Dorlo
- Pharmacometrics Research Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Michael Zhuo Wang
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas, USA
| |
Collapse
|
2
|
Evaluation of release and pharmacokinetics of hexadecylphosphocholine (miltefosine) in phosphatidyldiglycerol-based thermosensitive liposomes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183698. [PMID: 34283999 DOI: 10.1016/j.bbamem.2021.183698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 12/18/2022]
Abstract
Hexadecylphosphocholine (HePC, Miltefosine) is a drug from the class of alkylphosphocholines with an antineoplastic and antiprotozoal activity. We previously reported that HePC uptake from thermosensitive liposomes (TSL) containing 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol (DPPG2) into cancer cells is accelerated at mild hyperthermia (HT) resulting in increased cytotoxicity. In this study, we compared HePC release of different TSL formulations in serum. HePC showed rapid but incomplete release below the transition temperature (Tm) of investigated TSL formulations in serum. Short heating (5 min) to 42 °C increased HePC release from DPPG2-TSL (Tm = 41 °C) by a factor of two in comparison to body temperature (37 °C). Bovine serum albumin (BSA) induced HePC release from DPPG2-TSL comparable to serum. Furthermore, multilamellar vesicles (MLV) were capable to extract HePC from DPPG2-TSL in a concentration- and temperature-dependent manner. Repetitive exposure of DPPG2-TSL to MLV at 37 °C led to a fast initial release of HePC which slowed down after subsequent extraction cycles finally reaching approx. 50% HePC release. A pharmacokinetic study in rats revealed a biphasic pattern with an immediate clearance of approx. 50% HePC whereas the remaining 50% HePC showed a prolonged circulation time. We speculate that HePC located in the external leaflet of DPPG2-TSL is rapidly released upon contact with suitable biological acceptors. As demonstrated by MLV transfer experiments, asymmetric incorporation of HePC into the internal leaflet of DPPG2-TSL might improve HePC retention in presence of complex biological media and still give rise to HT-induced HePC release.
Collapse
|
3
|
Gaillard B, Remy JS, Pons F, Lebeau L. Dual Gene Delivery Reagents From Antiproliferative Alkylphospholipids for Combined Antitumor Therapy. Front Chem 2020; 8:581260. [PMID: 33134279 PMCID: PMC7566913 DOI: 10.3389/fchem.2020.581260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/31/2020] [Indexed: 11/13/2022] Open
Abstract
Alkylphospholipids (APLs) have elicited great interest as antitumor agents due to their unique mode of action on cell membranes. However, their clinical applications have been limited so far by high hemolytic activity. Recently, cationic prodrugs of erufosine, a most promising APL, have been shown to mediate efficient intracellular gene delivery, while preserving the antiproliferative properties of the parent APL. Here, cationic prodrugs of the two APLs that are currently used in the clinic, miltefosine, and perifosine, are investigated and compared to the erufosine prodrugs. Their synthesis, stability, gene delivery and self-assembly properties, and hemolytic activity are discussed in detail. Finally, the potential of the pro-miltefosine and pro-perifosine compounds ME12 and PE12 in combined antitumor therapy is demonstrated using pUNO1-hTRAIL, a plasmid DNA encoding TRAIL, a member of the TNF superfamily. With these pro-APL compounds, we provide a proof of concept for a new promising strategy for cancer therapy combining gene therapy and APL-based chemotherapy.
Collapse
Affiliation(s)
- Boris Gaillard
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Jean-Serge Remy
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| |
Collapse
|
4
|
Gaillard B, Remy JS, Pons F, Lebeau L. Synthesis and Evaluation of Antitumor Alkylphospholipid Prodrugs. Pharm Res 2020; 37:106. [PMID: 32462253 DOI: 10.1007/s11095-020-02830-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/21/2020] [Indexed: 01/08/2023]
Abstract
PURPOSE Hemolysis is a serious side effect of antitumor alkylphospholipids (APLs) that limits dose levels and is a constraint in their use in therapeutic regimen. Nine prodrugs of promising APLs (miltefosine, perifosine, and erufosine) were synthesized so as to decrease their membrane activity and improve their toxicity profile while preserving their antineoplastic potency. METHODS The synthesis of the pro-APLs was straightforwardly achieved in one step starting from the parent APLs. The critical aggregation concentration of the prodrugs, their hydrolytic stability under various pH conditions, their blood compatibility and cytotoxicity in three different cell lines were determined and compared to those of the parent antitumor lipids. RESULTS The APL prodrugs display antitumor activity which is similar to that of the parent alkylphospholipids but without associated hemolytic toxicity. CONCLUSION The pro-APL compounds may be considered as intravenously injectable derivatives of APLs. They could thus address one of the major issues met in cancer therapies involving antitumor lipids and restricting their utilization to oral and topical administration because of limited maximum tolerated dose.
Collapse
Affiliation(s)
- Boris Gaillard
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS - Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin - BP 60024, 67401, Illkirch, France
| | - Jean-Serge Remy
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS - Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin - BP 60024, 67401, Illkirch, France
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS - Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin - BP 60024, 67401, Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS - Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin - BP 60024, 67401, Illkirch, France.
| |
Collapse
|
5
|
Eleftheriou K, Kaminari A, Panagiotaki KN, Sideratou Z, Zachariadis M, Anastassopoulou J, Tsiourvas D. A combination drug delivery system employing thermosensitive liposomes for enhanced cell penetration and improved in vitro efficacy. Int J Pharm 2020; 574:118912. [PMID: 31809858 DOI: 10.1016/j.ijpharm.2019.118912] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/15/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023]
Abstract
Drug-loaded thermosensitive liposomes are investigated as drug delivery systems in combination with local mild hyperthermia therapy due to their capacity to release their cargo at a specific temperature range (40-42 °C). Additional benefit can be achieved by the development of such systems that combine two different anticancer drugs, have cell penetration properties and, when heated, release their drug payload in a controlled fashion. To this end, liposomes were developed incorporating at low concentration (5 mol%) a number of monoalkylether phosphatidylcholine lipids, encompassing the platelet activating factor, PAF, and its analogues that induce thermoresponsiveness and have anticancer biological activity. These thermoresponsive liposomes were efficiently (>90%) loaded with doxorubicin (DOX), and their thermal properties, stability and drug release were investigated both at 37 ◦C and at elevated temperatures. In vitro studies of the most advantageous liposomal formulation containing the methylated PAF derivative (methyl-PAF, edelfosine), an established antitumor agent, were performed on human prostate cancer cell lines. This system exhibits controlled release of DOX at 40-42 °C, enhanced cell uptake due to the presence of methyl-PAF, and improved cell viability inhibition due to the combined action of both medications.
Collapse
Affiliation(s)
- Kleopatra Eleftheriou
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Katerina N Panagiotaki
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Zili Sideratou
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Michael Zachariadis
- Institute of Biosciences and Applications, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece
| | - Jane Anastassopoulou
- Radiation Chemistry and Biospectroscopy, School of Chemical Engineering, National Technical University of Athens, Athens, Greece
| | - Dimitris Tsiourvas
- Institute of Nanoscience and Nanotechnology, NCSR ''Demokritos", 15310 Aghia Paraskevi, Greece.
| |
Collapse
|
6
|
Gaillard B, Seguin C, Remy JS, Pons F, Lebeau L. Erufosine (ErPC3) Cationic Prodrugs as Dual Gene Delivery Reagents for Combined Antitumor Therapy. Chemistry 2019; 25:15662-15679. [PMID: 31549752 DOI: 10.1002/chem.201903976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/23/2019] [Indexed: 12/14/2022]
Abstract
Sixteen cationic prodrugs of the antitumor alkylphospholipid (APL) erufosine were rationally synthesized to provide original gene delivery reagents with improved cytotoxicity profile. The DNA complexation properties of these cationic lipids were determined and associated transfection rates were measured. Furthermore, the self-assembly properties of the pro-erufosine compounds were investigated and their critical aggregation concentration was determined. Their hydrolytic stability under pH conditions mimicking the extracellular environment and the late endosome milieu was measured. Hemolytic activity and cytotoxicity of the compounds were investigated. The results obtained in various cell lines demonstrate that the prodrugs of erufosine display antineoplastic activity similar to that of the parent antitumor drug but are not associated with hemolytic toxicity, which is a dose-limiting side effect of APLs and a major obstacle to their use in anticancer therapeutic regimen. Furthermore, by using lipoplexes prepared from a prodrug of erufosine and a plasmid DNA encoding a pro-apoptotic protein (TRAIL), evidence was provided for selective cytotoxicity towards tumor cells while nontumor cells were resistant. This study demonstrates that the combination approach involving well tolerated erufosine cationic prodrugs and cancer gene therapy holds significant promise in tumor therapy.
Collapse
Affiliation(s)
- Boris Gaillard
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Cendrine Seguin
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Jean-Serge Remy
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin-BP 60024, 67401, Illkirch, France
| |
Collapse
|
7
|
Rodríguez-Nogales C, Sebastián V, Irusta S, Desmaële D, Couvreur P, Blanco-Prieto MJ. A unique multidrug nanomedicine made of squalenoyl-gemcitabine and alkyl-lysophospholipid edelfosine. Eur J Pharm Biopharm 2019; 144:165-173. [PMID: 31546021 DOI: 10.1016/j.ejpb.2019.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 02/04/2023]
Abstract
Among anticancer nanomedicines, squalenoyl nanocomposites have obtained encouraging outcomes in a great variety of tumors. The prodrug squalenoyl-gemcitabine has been chosen in this study to construct a novel multidrug nanosystem in combination with edelfosine, an alkyl-lysophopholipid with proven anticancer activity. Given their amphiphilic nature, it was hypothesized that both anticancer compounds, with complementary molecular targets, could lead to the formation of a new multitherapy nanomedicine. Nanoassemblies were formulated by the nanoprecipitation method and characterized by dynamic light scattering, transmission electron microscopy and X-ray photoelectron spectroscopy. Because free edelfosine is highly hemolytic, hemolysis experiments were performed using human blood erythrocytes and nanoassemblies efficacy was evaluated in a patient-derived metastatic pediatric osteosarcoma cell line. It was observed that these molecules spontaneously self-assembled as stable and monodisperse nanoassemblies of 51 ± 1 nm in a surfactant/polymer free-aqueous suspension. Compared to squalenoyl-gemcitabine nanoassemblies, the combination of squalenoyl-gemcitabine with edelfosine resulted in smaller particle size and a new supramolecular conformation, with higher stability and drug content, and ameliorated antitumor profile.
Collapse
Affiliation(s)
- C Rodríguez-Nogales
- Chemistry and Pharmaceutical Technology Department, Universidad de Navarra, Pamplona 31008, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - V Sebastián
- Department of Chemical and Environmental Engineering & Institute of Nanoscience of Aragon (INA), University of Zaragoza, Zaragoza 50018, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain
| | - S Irusta
- Department of Chemical and Environmental Engineering & Institute of Nanoscience of Aragon (INA), University of Zaragoza, Zaragoza 50018, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid 28029, Spain
| | - D Desmaële
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry Cedex 92290, France
| | - P Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry Cedex 92290, France.
| | - M J Blanco-Prieto
- Chemistry and Pharmaceutical Technology Department, Universidad de Navarra, Pamplona 31008, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain.
| |
Collapse
|
8
|
Voak AA, Standing JF, Sepúlveda N, Harris A, Croft SL, Seifert K. Pharmacodynamics and cellular accumulation of amphotericin B and miltefosine in Leishmania donovani-infected primary macrophages. J Antimicrob Chemother 2019; 73:1314-1323. [PMID: 29506127 PMCID: PMC5909632 DOI: 10.1093/jac/dky014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022] Open
Abstract
Objectives We examined the in vitro pharmacodynamics and cellular accumulation of the standard anti-leishmanial drugs amphotericin B and miltefosine in intracellular Leishmania donovani amastigote-macrophage drug assays. Methods Primary mouse macrophages were infected with L. donovani amastigotes. In time-kill assays infected macrophages were exposed to at least six different concentrations of serially diluted drugs and the percentage of infected macrophages was determined after 6, 12, 24, 48, 72 and 120 h of exposure. Cellular drug accumulation was measured following exposure to highly effective drug concentrations for 1, 6, 24, 48 and 72 h. Data were analysed through a mathematical model, relating drug concentration to the percentage of infected cells over time. Host cell membrane damage was evaluated through measurement of lactate dehydrogenase release. The effect of varying the serum and albumin concentrations in medium on the cellular accumulation levels of miltefosine was measured. Results Amphotericin B was more potent than miltefosine (EC50 values of 0.65 and 1.26 μM, respectively) and displayed a wider therapeutic window in vitro. The kinetics of the cellular accumulation of amphotericin B was concentration- and formulation-dependent. At an extracellular concentration of 10 μM miltefosine maximum cellular drug levels preceded maximum anti-leishmanial kill. Miltefosine induced membrane damage in a concentration-, time- and serum-dependent manner. Its cellular accumulation levels increased with decreasing amounts of protein in assay medium. Conclusions We have developed a novel approach to investigate the cellular pharmacology of anti-leishmanial drugs that serves as a model for the characterization of new drug candidates.
Collapse
Affiliation(s)
- Andrew A Voak
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Joseph F Standing
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Nuno Sepúlveda
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK.,Centre for Statistics and Applications of University of Lisbon, Lisbon, Portugal
| | - Andy Harris
- Pharmidex, 3rd Floor, 14 Hanover Street, London W1S 1YH, UK
| | - Simon L Croft
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Karin Seifert
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
9
|
Kip AE, Schellens JHM, Beijnen JH, Dorlo TPC. Clinical Pharmacokinetics of Systemically Administered Antileishmanial Drugs. Clin Pharmacokinet 2019; 57:151-176. [PMID: 28756612 PMCID: PMC5784002 DOI: 10.1007/s40262-017-0570-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review describes the pharmacokinetic properties of the systemically administered antileishmanial drugs pentavalent antimony, paromomycin, pentamidine, miltefosine and amphotericin B (AMB), including their absorption, distribution, metabolism and excretion and potential drug–drug interactions. This overview provides an understanding of their clinical pharmacokinetics, which could assist in rationalising and optimising treatment regimens, especially in combining multiple antileishmanial drugs in an attempt to increase efficacy and shorten treatment duration. Pentavalent antimony pharmacokinetics are characterised by rapid renal excretion of unchanged drug and a long terminal half-life, potentially due to intracellular conversion to trivalent antimony. Pentamidine is the only antileishmanial drug metabolised by cytochrome P450 enzymes. Paromomycin is excreted by the kidneys unchanged and is eliminated fastest of all antileishmanial drugs. Miltefosine pharmacokinetics are characterized by a long terminal half-life and extensive accumulation during treatment. AMB pharmacokinetics differ per drug formulation, with a fast renal and faecal excretion of AMB deoxylate but a much slower clearance of liposomal AMB resulting in an approximately ten-fold higher exposure. AMB and pentamidine pharmacokinetics have never been evaluated in leishmaniasis patients. Studies linking exposure to effect would be required to define target exposure levels in dose optimisation but have only been performed for miltefosine. Limited research has been conducted on exposure at the drug’s site of action, such as skin exposure in cutaneous leishmaniasis patients after systemic administration. Pharmacokinetic data on special patient populations such as HIV co-infected patients are mostly lacking. More research in these areas will help improve clinical outcomes by informed dosing and combination of drugs.
Collapse
Affiliation(s)
- Anke E Kip
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital/MC Slotervaart, Amsterdam, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Jan H M Schellens
- Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- Department of Clinical Pharmacology, Antoni van Leeuwenhoek Hospital/The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital/MC Slotervaart, Amsterdam, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
- Department of Clinical Pharmacology, Antoni van Leeuwenhoek Hospital/The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thomas P C Dorlo
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital/MC Slotervaart, Amsterdam, The Netherlands.
- Pharmacometrics Research Group, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Miltefosine Reduces the Cytolytic Activity and Virulence of Acinetobacter baumannii. Antimicrob Agents Chemother 2018; 63:AAC.01409-18. [PMID: 30373804 DOI: 10.1128/aac.01409-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/23/2018] [Indexed: 12/31/2022] Open
Abstract
Stagnation in antimicrobial development has led to a serious threat to public health because some Acinetobacter baumannii infections have become untreatable. New therapeutics with alternative mechanisms of action to combat A. baumannii are therefore necessary to treat these infections. To this end, the virulence of A. baumannii isolates with various antimicrobial susceptibilities was assessed when the isolates were treated with miltefosine, a phospholipase C inhibitor. Phospholipase C activity is a contributor to A. baumannii virulence associated with hemolysis, cytolysis of A549 human alveolar epithelial cells, and increased mortality in the Galleria mellonella experimental infection model. While the effects on bacterial growth were variable among strains, miltefosine treatment significantly reduced both the hemolytic and cytolytic activity of all treated A. baumannii strains. Additionally, scanning electron microscopy of polarized A549 cells infected with bacteria of the A. baumannii ATCC 19606T strain or the AB5075 multidrug-resistant isolate showed a decrease in A549 cell damage with a concomitant increase in the presence of A549 surfactant upon administration of miltefosine. The therapeutic ability of miltefosine was further supported by the results of G. mellonella infections, wherein miltefosine treatment of animals infected with ATCC 19606T significantly decreased mortality. These data demonstrate that inhibition of phospholipase C activity results in the overall reduction of A. baumannii virulence in both in vitro and in vivo models, making miltefosine a viable option for the treatment of A. baumannii infections, particularly those caused by multidrug-resistant isolates.
Collapse
|
11
|
Validation and Clinical Evaluation of a Novel Method To Measure Miltefosine in Leishmaniasis Patients Using Dried Blood Spot Sample Collection. Antimicrob Agents Chemother 2016; 60:2081-9. [PMID: 26787691 PMCID: PMC4808221 DOI: 10.1128/aac.02976-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/09/2016] [Indexed: 12/31/2022] Open
Abstract
To facilitate future pharmacokinetic studies of combination treatments against leishmaniasis in remote regions in which the disease is endemic, a simple cheap sampling method is required for miltefosine quantification. The aims of this study were to validate a liquid chromatography-tandem mass spectrometry method to quantify miltefosine in dried blood spot (DBS) samples and to validate its use with Ethiopian patients with visceral leishmaniasis (VL). Since hematocrit (Ht) levels are typically severely decreased in VL patients, returning to normal during treatment, the method was evaluated over a range of clinically relevant Ht values. Miltefosine was extracted from DBS samples using a simple method of pretreatment with methanol, resulting in >97% recovery. The method was validated over a calibration range of 10 to 2,000 ng/ml, and accuracy and precision were within ±11.2% and ≤7.0% (≤19.1% at the lower limit of quantification), respectively. The method was accurate and precise for blood spot volumes between 10 and 30 μl and for Ht levels of 20 to 35%, although a linear effect of Ht levels on miltefosine quantification was observed in the bioanalytical validation. DBS samples were stable for at least 162 days at 37°C. Clinical validation of the method using paired DBS and plasma samples from 16 VL patients showed a median observed DBS/plasma miltefosine concentration ratio of 0.99, with good correlation (Pearson's r = 0.946). Correcting for patient-specific Ht levels did not further improve the concordance between the sampling methods. This successfully validated method to quantify miltefosine in DBS samples was demonstrated to be a valid and practical alternative to venous blood sampling that can be applied in future miltefosine pharmacokinetic studies with leishmaniasis patients, without Ht correction.
Collapse
|
12
|
Morozova NG, Timofeev GA, Timakova AA, Shmendel EV, Kubasova TS, Tyutyunnik LL, Markova AA, Maslov MA, Shtil AA. Synthesis and antitumor activity of new alkyl glycoglycerolipids. MENDELEEV COMMUNICATIONS 2015. [DOI: 10.1016/j.mencom.2015.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Abramowski P, Steinbach K, Zander AR, Martin R. Immunomodulatory effects of the ether phospholipid edelfosine in experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 274:111-24. [PMID: 25086877 DOI: 10.1016/j.jneuroim.2014.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 01/24/2023]
Abstract
The 2-lysophosphatidylcholine analog edelfosine induces apoptosis in highly proliferating cells, e.g. activated immune cells. We examined mechanisms of action of edelfosine on immune functions in experimental autoimmune encephalomyelitis, a well-accepted animal model for multiple sclerosis. We observed activated caspase-3 expression in lymphoid organs and the central nervous system; however, edelfosine did not induce global apoptosis. Edelfosine improved the disease course and led to reduced frequencies of CD4(+) T cells infiltrating into the central nervous system. Our data suggest edelfosine as an interesting treatment candidate for multiple sclerosis.
Collapse
Affiliation(s)
- Pierre Abramowski
- Institute for Neuroimmunology and Clinical MS Research (inims), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Karin Steinbach
- Institute for Neuroimmunology and Clinical MS Research (inims), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Axel R Zander
- Department for Stem Cell Transplantation, University Cancer Center Hamburg (UCCH), Martinistr. 52, 20246 Hamburg, Germany
| | - Roland Martin
- Institute for Neuroimmunology and Clinical MS Research (inims), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany; Neuroimmunology and MS Research (nims), Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland.
| |
Collapse
|
14
|
Anticancer mechanisms and clinical application of alkylphospholipids. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:663-74. [PMID: 23137567 DOI: 10.1016/j.bbalip.2012.10.008] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 10/28/2012] [Accepted: 10/29/2012] [Indexed: 11/20/2022]
Abstract
Synthetic alkylphospholipids (ALPs), such as edelfosine, miltefosine, perifosine, erucylphosphocholine and erufosine, represent a relatively new class of structurally related antitumor agents that act on cell membranes rather than on DNA. They selectively target proliferating (tumor) cells, inducing growth arrest and apoptosis, and are potent sensitizers of conventional chemo- and radiotherapy. ALPs easily insert in the outer leaflet of the plasma membrane and cross the membrane via an ATP-dependent CDC50a-containing 'flippase' complex (in carcinoma cells), or are internalized by lipid raft-dependent endocytosis (in lymphoma/leukemic cells). ALPs resist catabolic degradation, therefore accumulate in the cell and interfere with lipid-dependent survival signaling pathways, notably PI3K-Akt and Raf-Erk1/2, and de novo phospholipid biosynthesis. At the same time, stress pathways (e.g. stress-activated protein kinase/JNK) are activated to promote apoptosis. In many preclinical and clinical studies, perifosine was the most effective ALP, mainly because it inhibits Akt activity potently and consistently, also in vivo. This property is successfully exploited clinically in highly malignant tumors, such as multiple myeloma and neuroblastoma, in which a tyrosine kinase receptor/Akt pathway is amplified. In such cases, perifosine therapy is most effective in combination with conventional anticancer regimens or with rapamycin-type mTOR inhibitors, and may overcome resistance to these agents. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metabolism.
Collapse
|
15
|
Estella-Hermoso de Mendoza A, Campanero MA, Lana H, Villa-Pulgarin JA, de la Iglesia-Vicente J, Mollinedo F, Blanco-Prieto MJ. Complete inhibition of extranodal dissemination of lymphoma by edelfosine-loaded lipid nanoparticles. Nanomedicine (Lond) 2012; 7:679-90. [DOI: 10.2217/nnm.11.134] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Lipid nanoparticles (LNs) made of synthetic lipids Compritol® 888 ATO and Precirol® ATO 5 were developed with an average size of 110.4 ± 2.1 and 103.1 ± 2.9 nm, and an encapsulation efficiency above 85% for both type of lipids. These LNs decrease the hemolytic toxicity of the drug by 90%. Materials & methods: Pharmacokinetic and biodistribution profiles of the drug were studied after intravenous and oral administration of edelfosine-containing LNs. Results: This provided an increase in relative oral bioavailability of 1500% after a single oral administration of drug-loaded LNs, maintaining edelfosine plasma levels over 7 days in contrast to a single oral administration of edelfosine solution, which presented a relative oral bioavailability of 10%. Moreover, edelfosine-loaded LNs showed a high accumulation of the drug in lymph nodes and resulted in slower tumor growth than the free drug in a murine lymphoma xenograft model, as well as potent extranodal dissemination inhibition. Original submitted 5 April 2011; Revised submitted 5 July 2011
Collapse
Affiliation(s)
| | - Miguel A Campanero
- Clinical Pharmacology Service, University of Navarra Clinic, E-31008, Spain
| | - Hugo Lana
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, E-31008, Spain
| | - Janny A Villa-Pulgarin
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Janis de la Iglesia-Vicente
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, E-37007 Salamanca, Spain
| | - María J Blanco-Prieto
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, E-31008, Spain
| |
Collapse
|
16
|
Bagley RG, Kurtzberg L, Rouleau C, Yao M, Teicher BA. Erufosine, an alkylphosphocholine, with differential toxicity to human cancer cells and bone marrow cells. Cancer Chemother Pharmacol 2011; 68:1537-46. [PMID: 21526352 DOI: 10.1007/s00280-011-1658-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 04/14/2011] [Indexed: 11/27/2022]
Abstract
PURPOSE To investigate the activity and myeloprotective properties of erufosine, a novel alkylphosphocholine (APC), on human malignant cells and normal bone marrow cells. METHODS Human or mouse bone marrow cells were exposed to erufosine, miltefosine, perifosine, or edelfosine in CFU-GM assays. Human MDA-MB-231 breast carcinoma, Panc-1 pancreatic carcinoma, and RPMI8226 multiple myeloma cells were exposed to erufosine in colony formation assays. Colony formation of Panc-1 tumor cells and mouse bone marrow cells ex vivo were quantified following intravenous administration of erufosine to tumor-bearing mice. Western blotting methods were applied to human U87 glioblastoma cells exposed to erufosine to investigate Akt inhibition. RESULTS Erufosine was less toxic to human and mouse bone marrow cells than perifosine, miltefosine, and edelfosine and was equally toxic to human and mouse CFU-GM. The human cancer cells MDA-MB-231 breast, Panc-1 pancreatic, and RPMI8226 MM cells were more sensitive to erufosine in a colony formation assay than were human bone marrow cells generating an approximately tenfold differential in IC(90) values. Erufosine injected intravenously significantly reduced Panc-1 tumor cell colony formation ex vivo but not mouse bone marrow CFU-GM. Erufosine inhibited Akt phosphorylation in human U87 glioblastoma cells. CONCLUSIONS Erufosine offers potential as a novel therapeutic for cancer with a reduced toxicity profile to bone marrow cells compared with other agents in this class. Human cancer cells were more sensitive to erufosine than human or mouse bone marrow cells indicating a favorable therapeutic window for erufosine.
Collapse
Affiliation(s)
- Rebecca G Bagley
- Genzyme Corporation, 49 New York Ave, Framingham, MA 01701-9322, USA.
| | | | | | | | | |
Collapse
|
17
|
Comparative study of A HPLC–MS assay versus an UHPLC–MS/MS for anti-tumoral alkyl lysophospholipid edelfosine determination in both biological samples and in lipid nanoparticulate systems. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877:4035-41. [DOI: 10.1016/j.jchromb.2009.10.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 10/06/2009] [Accepted: 10/19/2009] [Indexed: 11/20/2022]
|
18
|
Henke G, Lindner LH, Vogeser M, Eibl HJ, Wörner J, Müller AC, Bamberg M, Wachholz K, Belka C, Jendrossek V. Pharmacokinetics and biodistribution of Erufosine in nude mice--implications for combination with radiotherapy. Radiat Oncol 2009; 4:46. [PMID: 19852786 PMCID: PMC2773776 DOI: 10.1186/1748-717x-4-46] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 10/23/2009] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Alkylphosphocholines represent promising antineoplastic drugs that induce cell death in tumor cells by primary interaction with the cell membrane. Recently we could show that a combination of radiotherapy with Erufosine, a paradigmatic intravenously applicable alkylphosphocholine, in vitro leads to a clear increase of irradiation-induced cell death. In view of a possible combination of Erufosine and radiotherapy in vivo we determined the pharmacokinetics and bioavailability as well as the tolerability of Erufosine in nude mice. METHODS NMRI (nu/nu) nude mice were treated by intraperitoneal or subcutaneous injections of 5 to 40 mg/kg body weight Erufosine every 48 h for one to three weeks. Erufosine-concentrations were measured in brain, lungs, liver, small intestine, colon, spleen, kidney, stomach, adipoid tissue, and muscle by tandem-mass spectroscopy. Weight course, blood cell count and clinical chemistry were analyzed to evaluate general toxicity. RESULTS Intraperitoneal injections were generally well tolerated in all dose groups but led to a transient loss of the bodyweight (<10%) in a dose dependent manner. Subcutaneous injections of high-dose Erufosine caused local reactions at the injection site. Therefore, this regimen at 40 mg/kg body weight Erufosine was stopped after 14 days. No gross changes were observed in organ weight, clinical chemistry and white blood cell count in treated compared to untreated controls except for a moderate increase in lactate dehydrogenase and aspartate-aminotransferase after intensive treatment. Repeated Erufosine injections resulted in drug-accumulation in different organs with maximum concentrations of about 1000 nmol/g in spleen, kidney and lungs. CONCLUSION Erufosine was well tolerated and organ-concentrations surpassed the cytotoxic drug concentrations in vitro. Our investigations establish the basis for a future efficacy testing of Erufosine in xenograft tumor models in nude mice alone and in combination with chemo- or radiotherapy.
Collapse
Affiliation(s)
- Guido Henke
- Department of Radiooncology, University Hospital Tübingen, Hoppe-Seyler-Str 3, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
de Mendoza AEH, Campanero MA, de la Iglesia-Vicente J, Gajate C, Mollinedo F, Blanco-Prieto MJ. Antitumor Alkyl Ether Lipid Edelfosine: Tissue Distribution and Pharmacokinetic Behavior in Healthy and Tumor-Bearing Immunosuppressed Mice. Clin Cancer Res 2009; 15:858-64. [DOI: 10.1158/1078-0432.ccr-08-1654] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Lindner LH, Hossann M, Vogeser M, Teichert N, Wachholz K, Eibl H, Hiddemann W, Issels RD. Dual role of hexadecylphosphocholine (miltefosine) in thermosensitive liposomes: Active ingredient and mediator of drug release. J Control Release 2008; 125:112-20. [DOI: 10.1016/j.jconrel.2007.10.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 10/07/2007] [Accepted: 10/15/2007] [Indexed: 11/28/2022]
|
21
|
Bakht O, Delgado J, Amat-Guerri F, Acuña AU, London E. The phenyltetraene lysophospholipid analog PTE-ET-18-OMe as a fluorescent anisotropy probe of liquid ordered membrane domains (lipid rafts) and ceramide-rich membrane domains. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1768:2213-21. [PMID: 17573036 PMCID: PMC2034443 DOI: 10.1016/j.bbamem.2007.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 04/13/2007] [Accepted: 05/07/2007] [Indexed: 12/29/2022]
Abstract
The conjugated phenyltetraene PTE-ET-18-OMe (all-(E)-1-O-(15'-phenylpentadeca-8',10',12',14'-tetraenyl)-2-O-methyl-rac-glycero-3-phosphocholine) is a recently developed fluorescent lysophospholipid analog of edelfosine, (Quesada et al. (2004) J. Med. Chem. 47, 5333-5335). We investigated the use of this analog as a probe of membrane structure. PTE-ET-18-OMe was found to have several properties that are favorable for fluorescence anisotropy (polarization) experiments in membranes, including low fluorescence in water and moderately strong association with lipid bilayers. PTE-ET-18-OMe has absorbance and fluorescence properties similar to those of diphenylhexatriene (DPH) probes, with about as large a difference between its fluorescence anisotropy in liquid disordered (Ld) and ordered states (gel and Lo) as observed for DPH. Also like DPH, PTE-ET-18-OMe has a moderate affinity for both gel state ordered domains and Lo state ordered domains (rafts). However, unlike fluorescent sterols or DPH (Megha and London (2004) J. Biol. Chem. 279, 9997-10004), PTE-ET-18-OMe is not displaced from ordered domains by ceramide. Also unlike DPH, PTE-ET-18-OMe shows only slow exchange between the inner and outer leaflets of membrane bilayers, and can thus be used to examine anisotropy of an individual leaflet of a lipid bilayer. Since PTE-ET-18-OMe is a zwitterionic molecule, it should not be as influenced by electrostatic interactions as are other probes that do not cross the lipid bilayer but have a net charge. We conclude that PTE-ET-18-OMe has some unique properties that should make it a useful fluorescence probe of membrane structure.
Collapse
Affiliation(s)
- Omar Bakht
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | | | | | | | | |
Collapse
|
22
|
Taylor LA, Arends J, Hodina AK, Unger C, Massing U. Plasma lyso-phosphatidylcholine concentration is decreased in cancer patients with weight loss and activated inflammatory status. Lipids Health Dis 2007; 6:17. [PMID: 17623088 PMCID: PMC1939842 DOI: 10.1186/1476-511x-6-17] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 07/10/2007] [Indexed: 11/25/2022] Open
Abstract
Background It has been observed that ras-transformed cell lines in culture have a higher phosphatidylcholine (PC) biosynthesis rate as well as higher PC-degradation rate (increased PC-turnover) than normal cells. In correspondence to these findings, the concentrations of the PC-degradation product lyso-phosphatidylcholine (LPC) in cancer patients were found to be decreased. Our objective was the systematic investigation of the relationship between LPC and inflammatory and nutritional parameters in cancer patients. Therefore, plasma LPC concentrations were assessed in 59 cancer patients and related to nutritional and inflammatory parameters. To determine LPC in blood plasma we developed and validated a HPTLC method. Results Average plasma LPC concentration was 207 ± 59 μM which corresponds to the lower limit of the reported range in healthy subjects. No correlation between LPC and age, performance status, body mass index (BMI) or fat mass could be seen. However, LPC correlated inversely with plasma C-reactive protein (CRP) and whole blood hydrogen peroxides (HPO). Further, a negative correlation could be observed between LPC and whole body extra cellular fluid volume (ECF) as well as with relative change in body weight since cancer diagnosis. Conclusion In conclusion, LPC concentrations were decreased in cancer patients. LPC plasma concentrations correlated with weight loss and inflammatory parameters and, therefore, might be a general indicator of severity of malignant disease.
Collapse
Affiliation(s)
- Lenka A Taylor
- Tumor Biology Center, Dept. of Clinical Research, Breisacher Straße 117, D-79106 Freiburg, Germany
| | - Jann Arends
- Tumor Biology Center, Dept. of Clinical Research, Breisacher Straße 117, D-79106 Freiburg, Germany
| | - Arwen K Hodina
- Tumor Biology Center, Dept. of Clinical Research, Breisacher Straße 117, D-79106 Freiburg, Germany
| | - Clemens Unger
- Tumor Biology Center, Dept. of Clinical Research, Breisacher Straße 117, D-79106 Freiburg, Germany
| | - Ulrich Massing
- Tumor Biology Center, Dept. of Clinical Research, Breisacher Straße 117, D-79106 Freiburg, Germany
| |
Collapse
|
23
|
Ménez C, Buyse M, Farinotti R, Barratt G. Inward Translocation of the Phospholipid Analogue Miltefosine across Caco-2 Cell Membranes Exhibits Characteristics of a Carrier-mediated Process. Lipids 2007; 42:229-40. [PMID: 17393228 DOI: 10.1007/s11745-007-3026-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Accepted: 01/09/2007] [Indexed: 10/23/2022]
Abstract
Miltefosine (hexadecylphosphocholine, HePC) is the first effective oral agent for the treatment of visceral leishmaniasis. The characteristics of HePC incorporation into the human intestinal epithelial cell line Caco-2 were investigated in order to understand its oral absorption mechanism. The results provide evidence for the involvement of a carrier-mediated mechanism, since the association of HePC at the apical pole of Caco-2 cells was (1) saturable as a function of time with a rapid initial incorporation over 5 min followed by a more gradual increase; (2) saturable as a function of concentration over the range studied (2-200 microM) with a saturable component which followed Michaelis-Menten kinetics (apparent K (m) 15.7 micromol/L, V (max) 39.2 nmol/mg protein/h) and a nonspecific diffusion component; (3) partially inhibited by low temperature and ATP depletion, indicating the temperature and energy-dependence of the uptake process. Moreover, we demonstrated, by an albumin back-extraction method, that HePC is internalized via translocation from the outer to the inner leaflet of the plasma membrane and that HePC may preferentially diffuse through intact raft microdomains. In conclusion, our results suggest that incorporation of HePC at the apical membrane of Caco-2 cells may occur through a passive diffusion followed by a translocation in the inner membrane leaflet through an active carrier-mediated mechanism.
Collapse
Affiliation(s)
- Cécile Ménez
- Laboratoire de Physico-chimie, Pharmacotechnie et Biopharmacie, UMR CNRS 8612 Faculté de Pharmacie, Univ. Paris-Sud 11, IFR 141, Tour D5, 2éme étage, 5 rue J.B. Clément, Châtenay-Malabry, Cedex 92296, France
| | | | | | | |
Collapse
|
24
|
Rationale and clinical application of alkylphospholipid analogues in combination with radiotherapy. Cancer Treat Rev 2007; 33:191-202. [PMID: 17287087 DOI: 10.1016/j.ctrv.2006.12.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Revised: 12/01/2006] [Accepted: 12/04/2006] [Indexed: 12/29/2022]
Abstract
Concurrent treatment with radiotherapy and chemotherapy has emerged as an effective strategy to improve clinical outcome of cancer. In addition to combining radiation with classical anticancer agents, several new biological response modifiers are under investigation in pre-clinical and clinical studies. Synthetic alkylphospholipids are anticancer agents that in contrast to most anticancer drugs, do not target DNA, but insert in the plasma membrane and subsequently induce a broad range of biological effects, ultimately leading to cell death. Alkylphospholipids kill tumor cells directly by induction of both apoptotic and non-apoptotic cell death, and indirectly by interference with critical signal transduction pathways involved in phospholipid metabolism and survival. Due to their distinct mode of action, these drugs are considered as attractive candidates to combine with radiotherapy. In this review, we will discuss several alkylphospholipids that reached clinical application. These include first-generation alkyl-lysophospholipids edelfosine and ilmofosine, second-generation alkylphosphocholine-prototype miltefosine and more recently developed analogues perifosine and erucylphosphocholine. We focus on mechanisms of action and the rationale to combine these agents with radiotherapy. The preclinical results on molecular targeting underlying this approach will be reviewed, concluded with first clinical data on combined treatment of radiotherapy with perifosine.
Collapse
|
25
|
Ménez C, Buyse M, Dugave C, Farinotti R, Barratt G. Intestinal Absorption of Miltefosine: Contribution of Passive Paracellular Transport. Pharm Res 2007; 24:546-54. [PMID: 17252190 DOI: 10.1007/s11095-006-9170-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2006] [Accepted: 09/20/2006] [Indexed: 11/27/2022]
Abstract
PURPOSE This study aimed to characterize the transepithelial transport of miltefosine (HePC), the first orally effective drug against visceral leishmaniasis, across the intestinal barrier to further understand its oral absorption mechanism. MATERIALS AND METHODS Caco-2 cell monolayers were used as an in vitro model of the human intestinal barrier. The roles of active and passive mechanisms in HePC intestinal transport were investigated and the relative contributions of the transcellular and paracellular routes were estimated. RESULTS HePC transport was observed to be pH-independent, partially temperature-dependent, linear as a function of time and non-saturable as a function of concentration. The magnitude of HePC transport was quite similar to that of the paracellular marker mannitol, and EDTA treatment led to an increase in HePC transport. Furthermore, HePC transport was found to be similar in the apical-to-basolateral and basolateral-to-apical directions, strongly suggesting that HePC exhibits non-polarized transport and that no MDR-mediated efflux was involved. CONCLUSIONS These results demonstrate that HePC crosses the intestinal epithelium by a non-specific passive pathway and provide evidence supporting a concentration-dependent paracellular transport mechanism, although some transcellular diffusion cannot be ruled out. Considering that HePC opens epithelial tight junctions, this study shows that HePC may promote its own permeation across the intestinal barrier.
Collapse
Affiliation(s)
- Cécile Ménez
- Laboratoire de Physico-chimie, Pharmacotechnie et Biopharmacie, UMR CNRS 8612, IFR 141, University Paris-Sud 11, Faculté de Pharmacie, 5 rue J.B. Clément, Châtenay-Malabry Cedex, F-92296, France
| | | | | | | | | |
Collapse
|
26
|
Vink SR, Lagerwerf S, Mesman E, Schellens JHM, Begg AC, van Blitterswijk WJ, Verheij M. Radiosensitization of squamous cell carcinoma by the alkylphospholipid perifosine in cell culture and xenografts. Clin Cancer Res 2006; 12:1615-22. [PMID: 16533789 DOI: 10.1158/1078-0432.ccr-05-2033] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Combined modality treatment has improved outcome in various solid tumors. Besides classic anticancer drugs, a new generation of biological response modifiers has emerged that increases the efficacy of radiation. Here, we have investigated whether perifosine, an orally applicable, membrane-targeted alkylphospholipid, enhances the antitumor effect of radiation in vitro and in vivo. EXPERIMENTAL DESIGN Several long-term and short-term in vitro assays (clonogenic survival, sulforhodamine B cytotoxicity, apoptosis, and cell cycle analysis) were used to assess the cytotoxic effect of perifosine in combination with radiation. In vivo, the response of human KB squamous cell carcinoma xenografts was measured after treatment with perifosine, irradiation, and the combination. Radiolabeled perifosine was used to determine drug disposition in tumor and normal tissues. At various intervals after treatment, tumor specimens were collected to document histopathologic changes. RESULTS In vitro, perifosine reduced clonogenic survival, enhanced apoptosis, and increased cell cycle arrest after radiation. In vivo, radiation and perifosine alone induced a dose-dependent tumor growth delay. When combining multiple perifosine administrations with single or split doses of radiation, complete and sustained tumor regression was observed. Histopathologic analysis of tumor specimens revealed a prominent apoptotic response after combined treatment with radiation and perifosine. Radiation-enhanced tumor response was observed at clinically relevant plasma perifosine concentrations and accumulating drug disposition of >100 microg/g in tumor tissue. CONCLUSIONS Perifosine enhances radiation-induced cytotoxicity, as evidenced by reduced clonogenic survival and increased apoptosis induction in vitro and by complete tumor regression in vivo. These data provide strong support for further development of this combination in clinical studies.
Collapse
Affiliation(s)
- Stefan R Vink
- Division of Experimental Therapy and Cellular Biochemistry, the Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
27
|
Oberle C, Massing U, Krug HF. On the mechanism of alkylphosphocholine (APC)-induced apoptosis in tumour cells. Biol Chem 2005; 386:237-45. [PMID: 15843169 DOI: 10.1515/bc.2005.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alkylphosphocholines (APCs) represent a new and very encouraging class of antitumour agents that have also been shown to induce apoptosis in tumour cells, but their exact mode of action has still not been elucidated. The APC compound presented here, S-1-O-phosphocholine-2- N-acetyl-octadecane (S-NC-2) induces apoptosis in a variety of cancer cells. To define the molecular requirements for S-NC-2-induced apoptosis, activation of caspase-8 and -3 and the cleavage of death substrates, such as poly(ADP-ribose) polymerase (PARP), were investigated in Jurkat, BJAB, SKW6.4 and K562 cells. The signalling pathway seems to be initiated at the death receptor level. Cells that are defective in Fas-receptor signalling (e.g., FADDdn BJAB), as well as cells lacking the Fas receptor (K562), were resistant to S-NC-2 treatment. Furthermore, the treatment of Jurkat cells with S-NC-2 resulted in the clustering of death receptor molecules and co-localisation of the Fas receptor with caveolin, a marker for lipid rafts. In addition, the involvement of mitochondria was detected, since S-NC-2 induces the breakdown of the mitochondrial membrane potential. Overexpression of the anti-apoptotic protein Bcl-2 prevented the loss of delta psi(m) in type II (Jurkat) but not in type I cells (SKW6.4). Moreover, cleavage of Bid was found, which points to a possible linkage between the receptor-dependent and the mitochondrial pathways.
Collapse
Affiliation(s)
- Carolin Oberle
- Forschungszentrum Karlsruhe GmbH, Institut für Toxikologie und Genetik, P.O. Box 3640, D-76021 Karlsruhe, Germany
| | | | | |
Collapse
|
28
|
Vink SR, Schellens JHM, van Blitterswijk WJ, Verheij M. Tumor and normal tissue pharmacokinetics of perifosine, an oral anti-cancer alkylphospholipid. Invest New Drugs 2005; 23:279-86. [PMID: 16012787 DOI: 10.1007/s10637-005-1436-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Clinical use of anti-cancer alkylphospholipids is limited by gastrointestinal toxicity. However, new interest has emerged since it was shown that these drugs enhance the cytotoxic effect of conventional chemotherapy and radiotherapy in preclinical models. The aim of this study was to characterize the pharmacokinetic profile of perifosine, an oral analog of alkylphosphocholine (APC), and to compare in vitro drug uptake with in vivo drug accumulation in three human-derived squamous cell carcinomas (A431, HNXOE and KB). In vitro, KB cells showed a remarkably high uptake and sensitivity for perifosine compared with A431 and HNXOE cells. In vivo, perifosine reached a clinically relevant plasma concentration in mice after a single oral dose of 40 mg/kg. Perifosine was not metabolized and displayed slow elimination, with a terminal half-life of 137 (+/- 20) hours and an apparent volume of distribution of 11.3 l/kg. Comparable tumor accumulation was observed for A431 and HNXOE tumors, whereas perifosine uptake by KB xenografts was substantially higher. Tissue distribution occurred throughout the whole body reaching high perifosine levels in the gastro-intestinal tract, while heart and brain tissue contained relatively low levels. Based on its stability and relatively high tumor uptake in vivo, perifosine is an attractive candidate for further evaluation, e.g. as radiosensitizer.
Collapse
Affiliation(s)
- Stefan R Vink
- Division of Experimental Therapy, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
29
|
Arndt D, Zeisig R, Fichtner I, Teppke AD, Fahr A. Pharmacokinetics of sterically stabilized hexadecylphosphocholine liposomes versus conventional liposomes and free hexadecylphosphocholine in tumor-free and human breast carcinoma bearing mice. Breast Cancer Res Treat 1999; 58:71-80. [PMID: 10634520 DOI: 10.1023/a:1006224611505] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The pharmacokinetics of free and different liposomal formulations of hexadecylphosphocholine (HPC) was investigated in tumor-bearing (human mammary tumor MaTu) and tumor-free mice after intravenous and intraperitoneal administration. The levels of HPC were evaluated at different times in serum, normal tissues, and tumor. The purpose was to test the hypothesis that the enhanced therapeutic efficacy of sterically stabilized HPC liposomes in comparison to conventional vesicles and free HPC is due to its pharmacokinetics. Conventional non-compartmental pharmacokinetic analysis and an elaborate three- and four-compartmental model were used for explaining the experimental data. The serum levels of HPC obtained with sterically stabilized liposomes were only consistently higher in comparison to conventional vesicles and free HPC in the first 4 h. In the xenografted MaTu carcinoma, the differences of the HPC content between the different groups are unexpectedly low and do not reflect the high therapeutic activity [5] of sterically stabilized HPC liposomes. Detailed analysis shows that the liposomally encapsulated drug displays a modified pharmacokinetic behavior, which may also involve lymphatic absorption of the liposomal drug.
Collapse
Affiliation(s)
- D Arndt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| | | | | | | | | |
Collapse
|
30
|
Mayhew E, Ahmad I, Bhatia S, Dause R, Filep J, Janoff AS, Kaisheva E, Perkins WR, Zha Y, Franklin JC. Stability of association of 1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphocholine with liposomes is composition dependent. BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1329:139-48. [PMID: 9370251 DOI: 10.1016/s0005-2736(97)00102-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The ether lipid, 1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphocholine (ET-18-OCH3), has anticancer activity, but it has serious side-effects, including hemolysis, which prevent its optimal use. We surmised if ET-18-OCH3 could be stably associated with liposomes, less free ET-18-OCH3 would be available for lytic interaction with red cells. Liposome composition variables investigated included acyl chain saturation, phospholipid head group and mole ratio of Chol and ET-18-OCH3. It was found that attenuation of hemolysis was strongly liposome composition dependent. Some ET-18-OCH3 liposome compositions were minimally hemolytic. For example, whereas the HI5 (drug concentration required to cause 5% human red cell lysis) was 5-6 microM for free ET-18-OCH3, it was approximately 250 microM for DOPC (dioleoylphosphatidylcholine):Chol (cholesterol):DOPE-GA (glutaric acid derivatized DOPE):ET-18-OCH3, (4:3:1:2) and 640 microM for DOPE (dioleyolphosphatidylethanolamine):Chol:DOPE-GA:ET-18-OCH3 (4:3:1:2) liposomes. Efflux of carboxyfluorescein (CF) from liposomes and Langmuir trough determinations of mean molecular area of lipids in monolayers (MMAM) were used as indicators of membrane packing and stability. Incorporation of ET-18-OCH3 in liposomes reduced the MMAM. Reduction in CF permeation was correlated with reduction in hemolysis. The most stable liposomes included components, such as cholesterol, DOPC and DOPE, which have complementary shapes to ET-18-OCH3.
Collapse
Affiliation(s)
- E Mayhew
- The Liposome Company Inc., Princeton Forrestal Center, NJ 08540-6619, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Perkins WR, Dause RB, Li X, Franklin JC, Cabral-Lilly DJ, Zha Y, Dank EH, Mayhew E, Janoff AS. Combination of antitumor ether lipid with lipids of complementary molecular shape reduces its hemolytic activity. BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1327:61-8. [PMID: 9247167 DOI: 10.1016/s0005-2736(97)00043-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Because the therapeutic use of the antitumor ether lipid 1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphorylcholine (ET-18-OCH3) is restricted by its hemolytic activity we explored the use of lipid packing parameters to reduce this toxicity by creating structurally optimized ET-18-OCH3 liposomes. We postulated that combination of ET-18-OCH3, which is similar in structure to lysophosphatidylcholine, with lipid molecules of complementary molecular shape (opposite headgroup/chain volume) would likely yield a stable lamellar phase from which ET-18-OCH3 exchange to red blood cell membranes would be curtailed. To quantitate the degree of shape complementarity, we used a Langmuir trough and measured the mean molecular area per molecule (MMAM) for monolayers comprised of ET-18-OCH3, the host lipids, and binary mixtures of varying mole percentage ET-18-OCH3. The degree of complementarity was taken as the reduction in MMAM from the value expected based on simple additivity of the individual components. The greatest degree of shape complementarity was observed with cholesterol: the order of complementarity for the ET-18-OCH3-lipid mixtures examined was cholesterol >> DOPE > POPC approximately DOPC. Phosphorus NMR and TLC analysis of aqueous suspensions of ET-18-OCH3 (40 mol%) with the host lipids revealed them to all be lamellar phase. For ET-18-OCH3 at 40 mol% in liposomes, the hemolytic activity followed the trend of the reduction in MMAM and was least for the ET-18-OCH3/cholesterol system (H50 = 661 microM ET-18-OCH3) followed by ET-18-OCH3/DOPE (H50 = 91 microM) and mixtures with POPC and DOPC which were comparable at H50 = 26 microM and 38 microM, respectively: the H50 concentration for free ET-18-OCH3 was 16 microM. This experimental strategy for designing optimized liposomes with a reduction in exchange, and hence toxicity, may be useful for other amphipathic/lipophilic drugs that are dimensionally compatible with lipid bilayers.
Collapse
Affiliation(s)
- W R Perkins
- The Liposome Company, Inc., Princeton, New Jersey 08540, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Small GW, Strum JC, Daniel LW. Characterization of an HL-60 cell variant resistant to the antineoplastic ether lipid 1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine. Lipids 1997; 32:715-23. [PMID: 9252959 DOI: 10.1007/s11745-997-0091-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A resistant cell line (HL-60R) was selected by incubating HL-60 cells with increasing concentrations of 1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine (ET-18-OCH3) and used to examine the mechanism of resistance to the antineoplastic ether-linked lipid. The HL-60R cells exhibited a > 10-fold increase in resistance when measured by [3H]-thymidine incorporation in comparison to the HL-60 cell line. ET-18-OCH3 binding occurred at 4 degrees C and was not saturable at the concentrations tested (1-100 microM), indicating that the binding was receptor-independent. At 4 degrees C, association of ET-18-OCH3 was low for each cell line. AT 37 degrees C, uptake in the HL-60 cells was approximately 5-fold greater in comparison to HL-60R cells at each concentration tested. However, when the cellular content of ET-18-OCH3 was equal, both cell lines experienced similar declines in cell growth. Cellular incorporation of ether lipid was determined using serum-free media and in the presence of serum albumin or lipoproteins. Reduced uptake by the resistant cell line was observed only in the presence of albumin. A greater proportion of ether lipid could be removed from prelabeled HL-60R cells than from HL-60 cells, by an albumin wash procedure, indicating an increased rate of internalization and retention by the sensitive cell line. ET-18-OCH3 uptake in the HL-60 cell line was also more sensitive to treatment with endocytic (chloroquine, monensin) or metabolic (NaF, KCN) inhibitors. These results suggest that uptake is the principal determinant influencing sensitivity of the resistant cell line and consists of receptor-independent binding followed by internalization. Differential uptake requires the presence of serum albumin and is dependent on the energy-dependent endocytosis of the ether lipid.
Collapse
Affiliation(s)
- G W Small
- Department of Biochemistry, Bowman Gray School of Medicine, Wake Forest University, Winston-Salem, North Carolina 27157-1016, USA
| | | | | |
Collapse
|
33
|
Brinkmeier H, Schneider M, Rüdel R. Ether lipid-induced cell damage of neuroblastoma cells is only weakly correlated with increased intracellular Ca2+ levels. Cell Calcium 1996; 19:383-90. [PMID: 8793178 DOI: 10.1016/s0143-4160(96)90111-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The cell-damaging action of the ether lipid ET-18-OCH3 was studied in single NH15-CA2 neuroblastoma x glioma hybrid cells using light microscopy, and correlated with changes of the free intracellular calcium concentration, [Ca2+]i, as measured with the fluorescent Ca2+ indicator Fura-2. Addition of 3-100 microM ET-18-OCH3 to the cultures caused disintegration of neurites, cell rounding and detachment of the cells from the bottom of the culture dish. The effects occurred within 30-240 min, the faster, the higher the ET-18-OCH3 concentration. Also [Ca2+]i increased in a concentration-dependent manner, however, within seconds, and stayed high during the recording time of 20 min. The presence of 50 microM lanthanum or gadolinium ions prevented the ET-18-OCH3-induced increases of [Ca2+]i, but had no effect on neurite destruction and cell rounding. Preincubation with 1 mM diisopropylfluorphosphate or 100 microM leupeptin, both membrane-permeant inhibitors of intracellular proteases, did not prevent the effects. Nor was neurite destruction prohibited in the presence of 10 microM of the actin-stabilizing agent phalloidin or 2 microM taxol, a microtubule-stabilizer. We conclude that [Ca2+]i, although being increased during ET-18-OCH3-induced cell damage, is not the key factor of cell destruction.
Collapse
Affiliation(s)
- H Brinkmeier
- Department of General Physiology, University of Ulm, Germany.
| | | | | |
Collapse
|
34
|
Affiliation(s)
- W J Houlihan
- Charles A. Dana Research Institute for Scientists Emeriti, Drew University, Madison, New Jersey 07940-4000, USA
| | | | | | | |
Collapse
|