101
|
da Silva AFM, Vital RDU, Martins DDL, da Rocha DR, Ferreira GB, Camargos Resende JAL, Lanznaster M. Investigation of cobalt(iii)–TPA complexes as potential bioreductively activated carriers for naphthoquinone-based drugs. NEW J CHEM 2017. [DOI: 10.1039/c7nj03072j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Differently from CH3, halogens as substituents foster incorporation of methoxide into NQs upon complexation.
Collapse
|
102
|
Green BP, Renfrew AK, Glenister A, Turner P, Hambley TW. The influence of the ancillary ligand on the potential of cobalt(iii) complexes to act as chaperones for hydroxamic acid-based drugs. Dalton Trans 2017; 46:15897-15907. [DOI: 10.1039/c7dt03645k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cobalt(iii) chaperone complexes can modulate the cytotoxicity and subcellular distribution of biologically active hydroxamic acids.
Collapse
Affiliation(s)
| | | | | | - Peter Turner
- School of Chemistry
- The University of Sydney
- Sydney
- Australia
| | | |
Collapse
|
103
|
Faes S, Duval AP, Planche A, Uldry E, Santoro T, Pythoud C, Stehle JC, Horlbeck J, Letovanec I, Riggi N, Demartines N, Dormond O. Acidic tumor microenvironment abrogates the efficacy of mTORC1 inhibitors. Mol Cancer 2016; 15:78. [PMID: 27919264 PMCID: PMC5139076 DOI: 10.1186/s12943-016-0562-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/28/2016] [Indexed: 11/24/2022] Open
Abstract
Background Blocking the mechanistic target of rapamycin complex-1 (mTORC1) with chemical inhibitors such as rapamycin has shown limited clinical efficacy in cancer. The tumor microenvironment is characterized by an acidic pH which interferes with cancer therapies. The consequences of acidity on the anti-cancer efficacy of mTORC1 inhibitors have not been characterized and are thus the focus of our study. Methods Cancer cell lines were treated with rapamycin in acidic or physiological conditions and cell proliferation was investigated. The effect of acidity on mTORC1 activity was determined by Western blot. The anticancer efficacy of rapamycin in combination with sodium bicarbonate to increase the intratumoral pH was tested in two different mouse models and compared to rapamycin treatment alone. Histological analysis was performed on tumor samples to evaluate proliferation, apoptosis and necrosis. Results Exposing cancer cells to acidic pH in vitro significantly reduced the anti-proliferative effect of rapamycin. At the molecular level, acidity significantly decreased mTORC1 activity, suggesting that cancer cell proliferation is independent of mTORC1 in acidic conditions. In contrast, the activation of mitogen-activated protein kinase (MAPK) or AKT were not affected by acidity, and blocking MAPK or AKT with a chemical inhibitor maintained an anti-proliferative effect at low pH. In tumor mouse models, the use of sodium bicarbonate increased mTORC1 activity in cancer cells and potentiated the anti-cancer efficacy of rapamycin. Combining sodium bicarbonate with rapamycin resulted in increased tumor necrosis, increased cancer cell apoptosis and decreased cancer cell proliferation as compared to single treatment. Conclusions Taken together, these results emphasize the inefficacy of mTORC1 inhibitors in acidic conditions. They further highlight the potential of combining sodium bicarbonate with mTORC1 inhibitors to improve their anti-tumoral efficacy.
Collapse
Affiliation(s)
- Seraina Faes
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Adrian P Duval
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland.,Current Address: Swiss Institute of Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland
| | - Anne Planche
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Emilie Uldry
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Tania Santoro
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Catherine Pythoud
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Jean-Christophe Stehle
- Mouse Pathology Facility, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Janine Horlbeck
- Mouse Pathology Facility, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Igor Letovanec
- Institute of Pathology, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Nicolo Riggi
- Institute of Pathology, Lausanne University Hospital CHUV and University of Lausanne, Lausanne, Switzerland
| | - Nicolas Demartines
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland
| | - Olivier Dormond
- Lausanne University Hospital CHUV and University of Lausanne, Pavillon 4, Av. de Beaumont, 1011, Lausanne, Switzerland.
| |
Collapse
|
104
|
Tang J, Ji H, Ren J, Li M, Zheng N, Wu L. Solid lipid nanoparticles with TPGS and Brij 78: A co-delivery vehicle of curcumin and piperine for reversing P-glycoprotein-mediated multidrug resistance in vitro. Oncol Lett 2016; 13:389-395. [PMID: 28123572 DOI: 10.3892/ol.2016.5421] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/24/2016] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance (MDR) is a main clinical hurdle for chemotherapy of cancer, and overexpression of P-glycoprotein (P-gp) is a key factor. In the present study, a new co-delivery system for reversing MDR was designed and developed. The system was composed of curcumin (Cur) and piperine (Pip) encapsulated in solid lipid nanoparticles (SLNs) with tocopheryl polyethylene glycol succinate (TPGS) and Brij 78 [(Cur+Pip)-SLNs]. TPGS and Brij 78 could sensitize MDR tumors by inhibiting the P-gp drug efflux system. The combination of Cur and Pip, when administered in SLNs formulations, resulted in a significant enhancement in cytotoxicity and allowed efficient intracellular delivery of the drugs in drug-resistant A2780/Taxol cells. This dual inhibitory strategy may have significant potential in the clinical management of MDR in cancer.
Collapse
Affiliation(s)
- Jingling Tang
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hongyu Ji
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratory of Medications Research, College of Heilongjiang Province, Harbin, Heilongjiang 150086, P.R. China
| | - Jinmei Ren
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China; Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratory of Medications Research, College of Heilongjiang Province, Harbin, Heilongjiang 150086, P.R. China
| | - Mengting Li
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China; Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratory of Medications Research, College of Heilongjiang Province, Harbin, Heilongjiang 150086, P.R. China
| | - Nannan Zheng
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Linhua Wu
- Department of Pharmaceutics, School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China; Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Key Laboratory of Medications Research, College of Heilongjiang Province, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
105
|
Drachman N, Kadlecek S, Pourfathi M, Xin Y, Profka H, Rizi R. In vivo pH mapping of injured lungs using hyperpolarized [1- 13 C]pyruvate. Magn Reson Med 2016; 78:1121-1130. [PMID: 27714832 DOI: 10.1002/mrm.26473] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE To optimize the production of hyperpolarized 13 C-bicarbonate from the decarboxylation of hyperpolarized [1-13 C]pyruvate and use it to image pH in the lungs and heart of rats with acute lung injury. METHODS Two forms of catalysis are compared calorimetrically to maximize the rate of decarboxylation and rapidly produce hyperpolarized bicarbonate from pyruvate while minimizing signal loss. Rats are injured using an acute lung injury model combining ventilator-induced lung injury and acid aspiration. Carbon images are obtained from both healthy (n = 4) and injured (n = 4) rats using a slice-selective chemical shift imaging sequence with low flip angle. pH is calculated from the relative HCO3- and CO2 signals using the Henderson-Hasselbalch equation. RESULTS It is demonstrated that base catalysis is more effective than metal-ion catalysis for this decarboxylation reaction. Bicarbonate polarizations up to 17.2% are achieved using the base-catalyzed reaction. A mean pH difference between lung and heart of 0.14 pH units is measured in the acute lung injury model. A significant pH difference between injured and uninjured lungs is also observed. CONCLUSION It is demonstrated that hyperpolarized 13 C-bicarbonate can be efficiently produced from the base-catalyzed decarboxylation of pyruvate. This method is used to obtain the first regional pH image of the lungs and heart of an animal. Magn Reson Med 78:1121-1130, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Nicholas Drachman
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen Kadlecek
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mehrdad Pourfathi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yi Xin
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harilla Profka
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rahim Rizi
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
106
|
Longo DL, Bartoli A, Consolino L, Bardini P, Arena F, Schwaiger M, Aime S. In Vivo Imaging of Tumor Metabolism and Acidosis by Combining PET and MRI-CEST pH Imaging. Cancer Res 2016; 76:6463-6470. [PMID: 27651313 DOI: 10.1158/0008-5472.can-16-0825] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/13/2016] [Accepted: 08/15/2016] [Indexed: 11/16/2022]
Abstract
The vast majority of cancers exhibit increased glucose uptake and glycolysis regardless of oxygen availability. This metabolic shift leads to an enhanced production of lactic acid that decreases extracellular pH (pHe), a hallmark of the tumor microenvironment. In this way, dysregulated tumor pHe and upregulated glucose metabolism are linked tightly and their relative assessment may be useful to gain understanding of the underlying biology. Here we investigated noninvasively the in vivo correlation between tumor 18F-FDG uptake and extracellular pH values in a murine model of HER2+ breast cancer. Tumor extracellular pH and perfusion were assessed by acquiring MRI-CEST (chemical exchange saturation transfer) images on a 3T scanner after intravenous administration of a pH-responsive contrast agent (iopamidol). Static PET images were recorded immediately after MRI acquisitions to quantify the extent of 18F-FDG uptake. We demonstrated the occurrence of tumor pHe changes that report on acidification of the interstitial fluid caused by an accelerated glycolysis. Combined PET and MRI-CEST images reported complementary spatial information of the altered glucose metabolism. Notably, a significant inverse correlation was found between extracellular tumor pH and 18F-FDG uptake, as a high 18F-FDG uptake corresponds to lower extracellular pH values. These results show how merging the information from 18F-FDG-uptake and extracellular pH measurements can improve characterization of the tumor microenvironment. Cancer Res; 76(22); 6463-70. ©2016 AACR.
Collapse
Affiliation(s)
- Dario L Longo
- Institute of Biostructure and Bioimaging (CNR) c/o Molecular Biotechnologies Center, Torino, Italy.,Molecular Imaging Center, University of Torino, Torino, Italy
| | - Antonietta Bartoli
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lorena Consolino
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Paola Bardini
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Francesca Arena
- Molecular Imaging Center, University of Torino, Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universitat Munchen, Munich, Germany
| | - Silvio Aime
- Molecular Imaging Center, University of Torino, Torino, Italy. .,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
107
|
Bohloli M, Atashi A, Soleimani M, Kaviani S, Anbarlou A. Investigating Effects of Acidic pH on Proliferation, Invasion and Drug-Induced Apoptosis in Lymphoblastic Leukemia. CANCER MICROENVIRONMENT 2016; 9:119-126. [PMID: 27457339 DOI: 10.1007/s12307-016-0187-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022]
Abstract
Some studies have shown that extracellular pH in tumors, which results in tumor progression, is less than that in normal tissues. The aim of this study was to investigate the effects of extracellular acidic pH on proliferation, invasion, and drug-induced apoptosis in acute lymphoblastic cells. The cells were cultured in different pH (pH 6.6 and pH 7.4) for 12 days. Cell proliferation was assessed by MTT assay and cell invasion was assayed by invasion assay and gene expression analysis of MMP-9. Drug-induced apoptosis was evaluated after exposure to doxorubicin for 24 hours by annexin V/PI staining and gene expression analysis of BAX pro-apoptotic protein. The results indicated the enhanced growth and invasion of leukemic cells at pH 6.6 (P ≤ 0.05). Furthermore, the cells at pH 6.6 were resistant to apoptosis by doxorubicin (P ≤ 0.05). It can be concluded that acidic pH increases the proliferation, invasion and reduces the drug-induced apoptosis in acute lymphoblastic leukemia. Extracellular acidity can influence the behavior of leukemic cells and therefore, the manipulation of extracellular liquid can be selected as a therapeutic strategy for leukemia, especially for acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Mahbobeh Bohloli
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Atashi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeid Kaviani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Azadeh Anbarlou
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
108
|
Marchand V, Levêque P, Driesschaert B, Marchand-Brynaert J, Gallez B. In vivo EPR extracellular pH-metry in tumors using a triphosphonated trityl radical. Magn Reson Med 2016; 77:2438-2443. [PMID: 27364733 DOI: 10.1002/mrm.26316] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 01/19/2023]
Abstract
PURPOSE The ability to assess the extracellular pH (pHe) is an important issue in oncology, because extracellular acidification is associated with tumor aggressiveness and resistance to cytotoxic therapies. In this study, a stable triphosphonated triarylmethyl (TPTAM) radical was qualified as a pHe electron paramagnetic resonance (EPR) molecular reporter. METHODS Calibration of hyperfine splitting as a function of pH was performed using a 1.2-GHz EPR spectrometer. Gadolinium-diethylenetriamine pentaacetic acid (Gd-DTPA) was used as an extracellular paramagnetic broadening agent to assess the localization of TPTAM when incubated with cells. In vivo EPR pH-metry was performed in MDA, SiHa, and TLT tumor models and in muscle. Bicarbonate therapy was used to modulate the tumor pHe. EPR measurements were compared with microelectrode readouts. RESULTS The hyperfine splitting of TPTAM was strongly pH-dependent around the pKa of the probe (pKa = 6.99). Experiments with Gd-DTPA demonstrated that TPTAM remained in the extracellular compartment. pHe was found to be more acidic in the MDA, SiHa, and TLT tumor models compared with muscle. Treatment of animals by bicarbonate induced an increase in pHe in tumors: similar variations in pHe were found when using in vivo EPR or invasive microelectrodes measurements. CONCLUSION This study demonstrates the potential usefulness of TPTAM for monitoring pHe in tumors. Magn Reson Med 77:2438-2443, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Valérie Marchand
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université Catholique de Louvain, Brussels, Belgium
| | - Philippe Levêque
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université Catholique de Louvain, Brussels, Belgium
| | - Benoit Driesschaert
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université Catholique de Louvain, Brussels, Belgium.,Institute of Condensed Matter and Nanosciences, Molecules, Solids and Reactivity, Université Catholique de Louvain, Brussels, Belgium
| | - Jacqueline Marchand-Brynaert
- Institute of Condensed Matter and Nanosciences, Molecules, Solids and Reactivity, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Gallez
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
109
|
Nath K, Nelson DS, Putt ME, Leeper DB, Garman B, Nathanson KL, Glickson JD. Comparison of the Lonidamine Potentiated Effect of Nitrogen Mustard Alkylating Agents on the Systemic Treatment of DB-1 Human Melanoma Xenografts in Mice. PLoS One 2016; 11:e0157125. [PMID: 27285585 PMCID: PMC4902256 DOI: 10.1371/journal.pone.0157125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/25/2016] [Indexed: 11/18/2022] Open
Abstract
Previous NMR studies demonstrated that lonidamine (LND) selectively diminishes the intracellular pH (pHi) of DB-1 melanoma and mouse xenografts of a variety of other prevalent human cancers while decreasing their bioenergetic status (tumor βNTP/Pi ratio) and enhancing the activities of melphalan and doxorubicin in these cancer models. Since melphalan and doxorubicin are highly toxic agents, we have examined three other nitrogen (N)-mustards, chlorambucil, cyclophosphamide and bendamustine, to determine if they exhibit similar potentiation by LND. As single agents LND, melphalan and these N-mustards exhibited the following activities in DB-1 melanoma xenografts; LND: 100% tumor surviving fraction (SF); chlorambucil: 100% SF; cyclophosphamide: 100% SF; bendamustine: 79% SF; melphalan: 41% SF. When combined with LND administered 40 min prior to administration of the N-mustard (to maximize intracellular acidification) the following responses were obtained; chlorambucil: 62% SF; cyclophosphamide: 42% SF; bendamustine: 36% SF; melphalan: 10% SF. The effect of LND on the activities of these N-mustards is generally attributed to acid stabilization of the aziridinium active intermediate, acid inhibition of glutathione-S-transferase, which acts as a scavenger of aziridinium, and acid inhibition of DNA repair by O6-alkyltransferase. Depletion of ATP by LND may also decrease multidrug resistance and increase tumor response. At similar maximum tolerated doses, our data indicate that melphalan is the most effective N-mustard in combination with LND when treating DB-1 melanoma in mice, but the choice of N-mustard for coadministration with LND will also depend on the relative toxicities of these agents, and remains to be determined.
Collapse
Affiliation(s)
- Kavindra Nath
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - David S. Nelson
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mary E. Putt
- Biostatistics & Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Dennis B. Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Bradley Garman
- Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katherine L. Nathanson
- Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jerry D. Glickson
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
110
|
Federici C, Lugini L, Marino ML, Carta F, Iessi E, Azzarito T, Supuran CT, Fais S. Lansoprazole and carbonic anhydrase IX inhibitors sinergize against human melanoma cells. J Enzyme Inhib Med Chem 2016; 31:119-125. [PMID: 27142956 DOI: 10.1080/14756366.2016.1177525] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
CONTEXT Proton Pump Inhibitors (PPIs) reduce tumor acidity and therefore resistance of tumors to drugs. Carbonic Anhydrase IX (CA IX) inhibitors have proven to be effective against tumors, while tumor acidity might impair their full effectiveness. OBJECTIVE To analyze the effect of PPI/CA IX inhibitors combined treatment against human melanoma cells. METHODS The combination of Lansoprazole (LAN) and CA IX inhibitors (FC9-399A and S4) has been investigated in terms of cell proliferation inhibition and cell death in human melanoma cells. RESULTS The combination of these inhibitors was more effective than the single treatments in both inhibiting cell proliferation and in inducing cell death in human melanoma cells. DISCUSSION These results represent the first successful attempt in combining two different proton exchanger inhibitors. CONCLUSION This is the first evidence on the effectiveness of a new approach against tumors based on the combination of PPI and CA IX inhibitors, thus providing an alternative strategy against tumors.
Collapse
Affiliation(s)
- Cristina Federici
- a Department of Therapeutic Research and Medicine Evaluation , National Institute of Health , Rome , Italy and
| | - Luana Lugini
- a Department of Therapeutic Research and Medicine Evaluation , National Institute of Health , Rome , Italy and
| | - Maria Lucia Marino
- a Department of Therapeutic Research and Medicine Evaluation , National Institute of Health , Rome , Italy and
| | - Fabrizio Carta
- b NEUROFARBA Department, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze , Sesto Fiorentino (Florence) , Italy
| | - Elisabetta Iessi
- a Department of Therapeutic Research and Medicine Evaluation , National Institute of Health , Rome , Italy and
| | - Tommaso Azzarito
- a Department of Therapeutic Research and Medicine Evaluation , National Institute of Health , Rome , Italy and
| | - Claudiu T Supuran
- b NEUROFARBA Department, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze , Sesto Fiorentino (Florence) , Italy
| | - Stefano Fais
- a Department of Therapeutic Research and Medicine Evaluation , National Institute of Health , Rome , Italy and
| |
Collapse
|
111
|
Alfarouk KO. Tumor metabolism, cancer cell transporters, and microenvironmental resistance. J Enzyme Inhib Med Chem 2016; 31:859-66. [PMID: 26864256 DOI: 10.3109/14756366.2016.1140753] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Cancer cells reprogram their metabolic machineries to enter into permanent glycolytic pathways. The full reason for such reprogramming takes place is unclear. However, this metabolic switch is not made in vain for the lactate that is generated and exported outside cells is reused by other cells. This results in the generation of a pH gradient between the low extracellular pH that is acidic (pHe) and the higher cytosolic alkaline or near neutral pH (pHi) environments that are tightly regulated by the overexpression of several pumps and ion channels (e.g. NHE-1, MCT-1, V-ATPase, CA9, and CA12). The generation of this unique pH gradient serves as a determining factor in defining "tumor fitness". Tumor fitness is the capacity of the tumor to invade and metastasize due to its ability to reduce the efficiency of the immune system and confer resistance to chemotherapy. In this article, we highlight the importance of tumor microenvironment in mediating the failure of chemotherapeutic agents.
Collapse
Affiliation(s)
- Khalid O Alfarouk
- a Department of Pharmacology , Faculty of Pharmacy, AL-Neelain University , Khartoum , Sudan
| |
Collapse
|
112
|
Flavell RR, Truillet C, Regan MK, Ganguly T, Blecha JE, Kurhanewicz J, VanBrocklin HF, Keshari KR, Chang CJ, Evans MJ, Wilson DM. Caged [(18)F]FDG Glycosylamines for Imaging Acidic Tumor Microenvironments Using Positron Emission Tomography. Bioconjug Chem 2015; 27:170-8. [PMID: 26649808 DOI: 10.1021/acs.bioconjchem.5b00584] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Solid tumors are hypoxic with altered metabolism, resulting in secretion of acids into the extracellular matrix and lower relative pH, a feature associated with local invasion and metastasis. Therapeutic and diagnostic agents responsive to this microenvironment may improve tumor-specific delivery. Therefore, we pursued a general strategy whereby caged small-molecule drugs or imaging agents liberate their parent compounds in regions of low interstitial pH. In this manuscript, we present a new acid-labile prodrug method based on the glycosylamine linkage, and its application to a class of positron emission tomography (PET) imaging tracers, termed [(18)F]FDG amines. [(18)F]FDG amines operate via a proposed two-step mechanism, in which an acid-labile precursor decomposes to form the common radiotracer 2-deoxy-2-[(18)F]fluoro-d-glucose, which is subsequently accumulated by glucose avid cells. The rate of decomposition of [(18)F]FDG amines is tunable in a systematic fashion, tracking the pKa of the parent amine. In vivo, a 4-phenylbenzylamine [(18)F]FDG amine congener showed greater relative accumulation in tumors over benign tissue, which could be attenuated upon tumor alkalinization using previously validated models, including sodium bicarbonate treatment, or overexpression of carbonic anhydrase. This new class of PET tracer represents a viable approach for imaging acidic interstitial pH with potential for clinical translation.
Collapse
Affiliation(s)
- Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - Charles Truillet
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - Melanie K Regan
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - Tanushree Ganguly
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - Joseph E Blecha
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - Kayvan R Keshari
- Department of Radiology and Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center , New York, New York 10065, United States
| | - Christopher J Chang
- Departments of Chemistry and Molecular and Cell Biology and the Howard Hughes Medical Institute, University of California , Berkeley, California 94720, United States
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California , San Francisco, California 94158, United States
| |
Collapse
|
113
|
Yu F, Chen Z, Wang B, Jin Z, Hou Y, Ma S, Liu X. The role of lysosome in cell death regulation. Tumour Biol 2015; 37:1427-36. [DOI: 10.1007/s13277-015-4516-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/25/2015] [Indexed: 02/01/2023] Open
|
114
|
Amith SR, Wilkinson JM, Baksh S, Fliegel L. The Na⁺/H⁺ exchanger (NHE1) as a novel co-adjuvant target in paclitaxel therapy of triple-negative breast cancer cells. Oncotarget 2015; 6:1262-75. [PMID: 25514463 PMCID: PMC4359231 DOI: 10.18632/oncotarget.2860] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/25/2014] [Indexed: 02/04/2023] Open
Abstract
Dysregulation of Na+ /H+ exchanger isoform one (NHE1) activity is a hallmark of cells undergoing tumorigenesis and metastasis, the leading cause of patient mortality. The acidic tumor microenvironment is thought to facilitate the development of resistance to chemotherapy drugs and to promote extracellular matrix remodeling leading to metastasis. Here, we investigated NHE1 as a co-adjuvant target in paclitaxel chemotherapy of metastatic breast cancer. We generated a stable NHE1-knockout of the highly invasive, triple-negative, MDA-MB-231 breast cancer cells. The NHE1-knockout cells proliferated comparably to parental cells, but had markedly lower rates of migration and invasion in vitro. In vivo xenograft tumor growth in athymic nude mice was also dramatically decreased compared to parental MDA-MB-231 cells. Loss of NHE1 expression also increased the susceptibility of knockout cells to paclitaxel-mediated cell death. NHE1 inhibition, in combination with paclitaxel, resulted in a dramatic decrease in viability, and migratory and invasive potential of triple-negative breast cancer cells, but not in hormone receptor-positive, luminal MCF7 cells. Our data suggest that NHE1 is critical in triple-negative breast cancer metastasis, and its chemical inhibition boosts the efficacy of paclitaxel in vitro, highlighting NHE1 as a novel, potential co-adjuvant target in breast cancer chemotherapy.
Collapse
Affiliation(s)
- Schammim Ray Amith
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Shairaz Baksh
- Department of Pediatrics, Biochemistry and Oncology, Alberta Inflammatory Bowel Disease Consortium, University of Alberta, Edmonton, Alberta, Canada
| | - Larry Fliegel
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
115
|
Gilson RC, Tang R, Som A, Klajer C, Sarder P, Sudlow GP, Akers WJ, Achilefu S. Protonation and Trapping of a Small pH-Sensitive Near-Infrared Fluorescent Molecule in the Acidic Tumor Environment Delineate Diverse Tumors in Vivo. Mol Pharm 2015; 12:4237-46. [PMID: 26488921 DOI: 10.1021/acs.molpharmaceut.5b00430] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enhanced glycolysis and poor perfusion in most solid malignant tumors create an acidic extracellular environment, which enhances tumor growth, invasion, and metastasis. Complex molecular systems have been explored for imaging and treating these tumors. Here, we report the development of a small molecule, LS662, that emits near-infrared (NIR) fluorescence upon protonation by the extracellular acidic pH environment of diverse solid tumors. Protonation of LS662 induces selective internalization into tumor cells and retention in the tumor microenvironment. Noninvasive NIR imaging demonstrates selective retention of the pH sensor in diverse tumors, and two-photon microscopy of ex vivo tumors reveals significant retention of LS662 in tumor cells and the acid tumor microenvironment. Passive and active internalization processes combine to enhance NIR fluorescence in tumors over time. The low background fluorescence allows tumors to be detected with high sensitivity, as well as dead or dying cells to be delineated from healthy cells. In addition to demonstrating the feasibility of using small molecule pH sensors to image multiple aggressive solid tumor types via a protonation-induced internalization and retention pathway, the study reveals the potential of using LS662 to monitor treatment response and tumor-targeted drug delivery.
Collapse
Affiliation(s)
- Rebecca C Gilson
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Rui Tang
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Avik Som
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Chloe Klajer
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Pinaki Sarder
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Gail P Sudlow
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Walter J Akers
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| | - Samuel Achilefu
- Departments of †Radiology, ‡Biomedical Engineering, and §Biochemistry & Molecular Biophysics, Washington University in St. Louis , St. Louis 63110, United States
| |
Collapse
|
116
|
Gallagher FA, Sladen H, Kettunen MI, Serrao EM, Rodrigues TB, Wright A, Gill AB, McGuire S, Booth TC, Boren J, McIntyre A, Miller JL, Lee SH, Honess D, Day SE, Hu DE, Howat WJ, Harris AL, Brindle KM. Carbonic Anhydrase Activity Monitored In Vivo by Hyperpolarized 13C-Magnetic Resonance Spectroscopy Demonstrates Its Importance for pH Regulation in Tumors. Cancer Res 2015; 75:4109-18. [PMID: 26249175 PMCID: PMC4594768 DOI: 10.1158/0008-5472.can-15-0857] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/12/2015] [Indexed: 11/16/2022]
Abstract
Carbonic anhydrase buffers tissue pH by catalyzing the rapid interconversion of carbon dioxide (CO2) and bicarbonate (HCO3 (-)). We assessed the functional activity of CAIX in two colorectal tumor models, expressing different levels of the enzyme, by measuring the rate of exchange of hyperpolarized (13)C label between bicarbonate (H(13)CO3(-)) and carbon dioxide ((13)CO2), following injection of hyperpolarized H(13)CO3(-), using (13)C-magnetic resonance spectroscopy ((13)C-MRS) magnetization transfer measurements. (31)P-MRS measurements of the chemical shift of the pH probe, 3-aminopropylphosphonate, and (13)C-MRS measurements of the H(13)CO3(-)/(13)CO2 peak intensity ratio showed that CAIX overexpression lowered extracellular pH in these tumors. However, the (13)C measurements overestimated pH due to incomplete equilibration of the hyperpolarized (13)C label between the H(13)CO3(-) and (13)CO2 pools. Paradoxically, tumors overexpressing CAIX showed lower enzyme activity using magnetization transfer measurements, which can be explained by the more acidic extracellular pH in these tumors and the decreased activity of the enzyme at low pH. This explanation was confirmed by administration of bicarbonate in the drinking water, which elevated tumor extracellular pH and restored enzyme activity to control levels. These results suggest that CAIX expression is increased in hypoxia to compensate for the decrease in its activity produced by a low extracellular pH and supports the hypothesis that a major function of CAIX is to lower the extracellular pH.
Collapse
Affiliation(s)
- Ferdia A Gallagher
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom. Department of Radiology, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | - Helen Sladen
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Mikko I Kettunen
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Eva M Serrao
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Tiago B Rodrigues
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Alan Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Andrew B Gill
- Department of Radiology, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Sarah McGuire
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Thomas C Booth
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Joan Boren
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Alan McIntyre
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jodi L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Shen-Han Lee
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Davina Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Sam E Day
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - De-En Hu
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - William J Howat
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Adrian L Harris
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| |
Collapse
|
117
|
Xing Y, Zhao S, Zhou BP, Mi J. Metabolic reprogramming of the tumour microenvironment. FEBS J 2015; 282:3892-8. [DOI: 10.1111/febs.13402] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/18/2015] [Accepted: 08/05/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Yazhi Xing
- Department of Biochemistry and Molecular Cell Biology Shanghai Key Laboratory of Tumor Microenvironment and Inflammation Shanghai Jiao Tong University School of Medicine China
| | - Shimin Zhao
- School of Life Sciences Fudan University Shanghai China
| | - Binhua P. Zhou
- Markey Cancer Center University of Kentucky Lexington USA
| | - Jun Mi
- Department of Biochemistry and Molecular Cell Biology Shanghai Key Laboratory of Tumor Microenvironment and Inflammation Shanghai Jiao Tong University School of Medicine China
| |
Collapse
|
118
|
Wang BY, Zhang J, Wang JL, Sun S, Wang ZH, Wang LP, Zhang QL, Lv FF, Cao EY, Shao ZM, Fais S, Hu XC. Intermittent high dose proton pump inhibitor enhances the antitumor effects of chemotherapy in metastatic breast cancer. J Exp Clin Cancer Res 2015; 34:85. [PMID: 26297142 PMCID: PMC4546346 DOI: 10.1186/s13046-015-0194-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/20/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Acidity is a hallmark of malignant tumor, representing a very efficient mechanism of chemoresistance. Proton pump inhibitors (PPI) at high dosage have been shown to sensitize chemoresistant human tumor cells and tumors to cytotoxic molecules. The aim of this pilot study was to investigate the efficacy of PPI in improving the clinical outcome of docetaxel + cisplatin regimen in patients with metastatic breast cancer (MBC). METHODS Patients enrolled were randomly assigned to three arms: Arm A, docetaxel 75 mg/m(2) followed by cisplatin 75 mg/m(2) on d4, repeated every 21 days with a maximum of 6 cycles; Arm B, the same chemotherapy preceded by three days esomeprazole (ESOM) 80 mg p.o. bid, beginning on d1 repeated weekly. Weekly intermittent administration of ESOM (3 days on 4 days off) was maintained up to maximum 66 weeks; Arm C, the same as Arm B with the only difference being dose of ESOM at 100 mg p.o. bid. The primary endpoint was response rate. RESULTS Ninety-four patients were randomly assigned and underwent at least one injection of chemotherapy. Response rates for arm A, B and C were 46.9, 71.0, and 64.5 %, respectively. Median TTP for arm A (n = 32), B (n = 31), C (n = 31) were 8.7, 9.4, and 9.7 months, respectively. A significant difference was observed between patients who had taken PPI and who not with ORR (67.7 % vs. 46.9 %, p = 0.049) and median TTP (9.7 months vs. 8.7 months, p = 0.045) [corrected]. Exploratory analysis showed that among 15 patients with triple negative breast cancer (TNBC), this difference was bigger with median TTP of 10.7 and 5.8 months, respectively (p = 0.011). PPI combination showed a marked effect on OS as well, while with a borderline significance (29.9 vs. 19.2 months, p = 0.090). No additional toxicity was observed with PPI. CONCLUSIONS The results of this pilot clinical trial showed that intermittent high dose PPI enhance the antitumor effects of chemotherapy in MBC patients without evidence of additional toxicity, which requires urgent validation in a multicenter, randomized, phase III trial. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT01069081 .
Collapse
Affiliation(s)
- Bi-Yun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Lei Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Si Sun
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong-Hua Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei-Ping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qun-Ling Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Fang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - En-Ying Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Min Shao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Stefano Fais
- Anti-Tumour Drugs Section, Department of Therapeutic Research and Medicines Evaluation, National Institute of Health, Rome, Italy.
| | - Xi-Chun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
119
|
Pacheco-Torres J, Mukherjee N, Walko M, López-Larrubia P, Ballesteros P, Cerdan S, Kocer A. Image guided drug release from pH-sensitive Ion channel-functionalized stealth liposomes into an in vivo glioblastoma model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1345-54. [DOI: 10.1016/j.nano.2015.03.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 03/03/2015] [Accepted: 03/29/2015] [Indexed: 01/11/2023]
|
120
|
Evaluating pH in the Extracellular Tumor Microenvironment Using CEST MRI and Other Imaging Methods. ACTA ACUST UNITED AC 2015; 2015. [PMID: 27761517 DOI: 10.1155/2015/206405] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor acidosis is a consequence of altered metabolism, which can lead to chemoresistance and can be a target of alkalinizing therapies. Noninvasive measurements of the extracellular pH (pHe) of the tumor microenvironment can improve diagnoses and treatment decisions. A variety of noninvasive imaging methods have been developed for measuring tumor pHe. This review provides a detailed description of the advantages and limitations of each method, providing many examples from previous research reports. A substantial emphasis is placed on methods that use MR spectroscopy and MR imaging, including recently developed methods that use chemical exchange saturation transfer MRI that combines some advantages of MR spectroscopy and imaging. Together, this review provides a comprehensive overview of methods for measuring tumor pHe, which may facilitate additional creative approaches in this research field.
Collapse
|
121
|
Abstract
Cancers progress through a series of events that can be characterized as "somatic evolution." A central premise of Darwinian evolutionary theory is that the environment imparts pressure to select for species that are most fit within that particular microenvironmental context. Furthermore, the rate of evolution is proportional to both (1) the strength of the environmental selection and (2) the phenotypic variance of the selected population. It is notable that, during the progression of cancers from carcinogenesis to local invasion to metastasis, the selective landscape continuously changes, and throughout this process, there is increased selection for cells that have altered metabolic phenotypes: implying that these phenotypes impart a selective advantage during the process of environmental selection. One of the most prevalent selected phenotypes is that of aerobic glycolysis, that is, the continued fermentation of glucose even in the presence of adequate oxygen. The mechanisms of this so-called "Warburg effect" have been well studied, and there are multiple models to explain how this occurs at the molecular level. Herein, we propose that unifying insights can be gained by evaluating the environmental context within which this phenotype arises. In other words, we focus not on the "how" but the "why" do cancer cells exhibit high aerobic glycolysis. This is best approached by examining the sequelae of aerobic glycolysis that may impart a selective advantage. Many of these have been considered, including generation of anabolic substrates, response rates of glycolysis vis-à-vis respiration, and generation of antioxidants. A further sequeala considered here is that aerobic glycolysis results in a high rate of lactic acid production; resulting in acidification of the extracellular space. Indeed, it has been shown that a low extracellular pH promotes local invasion, promotes metastasis, and inhibits antitumor immunity. In naturally occurring cancers, low extracellular pH is a strong negative prognostic indicator of metastasis-free survival. Furthermore, it has been shown that inhibition of extracellular acidosis can inhibit metastasis and promote antitumor immunity. Hence, we propose that excess acid production confers a selective advantage for cells during the somatic evolution of cancers.
Collapse
Affiliation(s)
- Robert J Gillies
- From the Departments of Cancer Imaging and Metabolism and Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | |
Collapse
|
122
|
Sarkar A, Karmakar S, Bhattacharyya S, Purkait K, Mukherjee A. Nitric oxide release by N-(2-chloroethyl)-N-nitrosoureas: a rarely discussed mechanistic path towards their anticancer activity. RSC Adv 2015. [DOI: 10.1039/c4ra11137k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Our work shows that NO release is a feasible pathway of action for aromatic and heterocyclic N-(2-chloroethyl)-N-nitrosoureas and faster NO release may not lead to higher cytotoxicity.
Collapse
Affiliation(s)
- Amrita Sarkar
- Department of Chemical Sciences
- Indian Institute of Science Education & Research Kolkata
- Nadia
- India
| | - Subhendu Karmakar
- Department of Chemical Sciences
- Indian Institute of Science Education & Research Kolkata
- Nadia
- India
| | - Sudipta Bhattacharyya
- Department of Chemical Sciences
- Indian Institute of Science Education & Research Kolkata
- Nadia
- India
| | - Kallol Purkait
- Department of Chemical Sciences
- Indian Institute of Science Education & Research Kolkata
- Nadia
- India
| | - Arindam Mukherjee
- Department of Chemical Sciences
- Indian Institute of Science Education & Research Kolkata
- Nadia
- India
| |
Collapse
|
123
|
Choi SYC, Lin D, Gout PW, Collins CC, Xu Y, Wang Y. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Adv Drug Deliv Rev 2014; 79-80:222-37. [PMID: 25305336 DOI: 10.1016/j.addr.2014.09.009] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 09/02/2014] [Accepted: 09/23/2014] [Indexed: 12/21/2022]
Abstract
The development of novel cancer therapeutics is often plagued by discrepancies between drug efficacies obtained in preclinical studies and outcomes of clinical trials. The inconsistencies can be attributed to a lack of clinical relevance of the cancer models used for drug testing. While commonly used in vitro culture systems are advantageous for addressing specific experimental questions, they are often gross, fidelity-lacking simplifications that largely ignore the heterogeneity of cancers as well as the complexity of the tumor microenvironment. Factors such as tumor architecture, interactions among cancer cells and between cancer and stromal cells, and an acidic tumor microenvironment are critical characteristics observed in patient-derived cancer xenograft models and in the clinic. By mimicking these crucial in vivo characteristics through use of 3D cultures, co-culture systems and acidic culture conditions, an in vitro cancer model/microenvironment that is more physiologically relevant may be engineered to produce results more readily applicable to the clinic.
Collapse
Affiliation(s)
- Stephen Yiu Chuen Choi
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Peter W Gout
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| | - Colin C Collins
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Yong Xu
- Department of Urology, Second Affiliated Hospital of Tianjin Medical University, Tianjin, P.R. China.
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| |
Collapse
|
124
|
Acid-mediated tumor proteolysis: contribution of cysteine cathepsins. Neoplasia 2014; 15:1125-37. [PMID: 24204192 DOI: 10.1593/neo.13946] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/28/2013] [Accepted: 08/28/2013] [Indexed: 12/21/2022] Open
Abstract
One of the noncellular microenvironmental factors that contribute to malignancy of solid tumors is acidic peritumoral pH. We have previously demonstrated that extracellular acidosis leads to localization of the cysteine pro-tease cathepsin B on the tumor cell membrane and its secretion. The objective of the present study was to determine if an acidic extracellular pH such as that observed in vivo (i.e., pHe 6.8) affects the activity of proteases, e.g., cathepsin B, that contribute to degradation of collagen IV by tumor cells when grown in biologically relevant three-dimensional (3D) cultures. For these studies, we used 1) 3D reconstituted basement membrane overlay cultures of human carcinomas, 2) live cell imaging assays to assess proteolysis, and 3) in vivo imaging of active tumor proteases. At pHe 6.8, there were increases in pericellular active cysteine cathepsins and in degradation of dye-quenched collagen IV, which was partially blocked by a cathepsin B inhibitor. Imaging probes for active cysteine cathepsins localized to tumors in vivo. The amount of bound probe decreased in tumors in bicarbonate-treated mice, a treatment previously shown to increase peritumoral pHe and reduce local invasion of the tumors. Our results are consistent with the acid-mediated invasion hypothesis and with a role for cathepsin B in promoting degradation of a basement membrane protein substrate, i.e., type IV collagen, in an acidic peritumoral environment.
Collapse
|
125
|
Scholz DJ, Janich MA, Köllisch U, Schulte RF, Ardenkjaer-Larsen JH, Frank A, Haase A, Schwaiger M, Menzel MI. Quantified pH imaging with hyperpolarized (13) C-bicarbonate. Magn Reson Med 2014; 73:2274-82. [PMID: 25046867 DOI: 10.1002/mrm.25357] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/18/2014] [Accepted: 06/18/2014] [Indexed: 11/07/2022]
Abstract
PURPOSE Because pH plays a crucial role in several diseases, it is desirable to measure pH in vivo noninvasively and in a spatially localized manner. Spatial maps of pH were quantified in vitro, with a focus on method-based errors, and applied in vivo. METHODS In vitro and in vivo (13) C mapping were performed for various flip angles for bicarbonate (BiC) and CO2 with spectral-spatial excitation and spiral readout in healthy Lewis rats in five slices. Acute subcutaneous sterile inflammation was induced with Concanavalin A in the right leg of Buffalo rats. pH and proton images were measured 2 h after induction. RESULTS After optimizing the signal to noise ratio of the hyperpolarized (13) C-bicarbonate, error estimation of the spectral-spatial excited spectrum reveals that the method covers the biologically relevant pH range of 6 to 8 with low pH error (< 0.2). Quantification of pH maps shows negligible impact of the residual bicarbonate signal. pH maps reflect the induction of acute metabolic alkalosis. Inflamed, infected regions exhibit lower pH. CONCLUSION Hyperpolarized (13) C-bicarbonate pH mapping was shown to be sensitive in the biologically relevant pH range. The mapping of pH was applied to healthy in vivo organs and interpreted within inflammation and acute metabolic alkalosis models.
Collapse
Affiliation(s)
| | | | - Ulrich Köllisch
- Technische Universität München, Institute of Medical Engineering, Munich, Germany
| | | | - Jan H Ardenkjaer-Larsen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Denmark.,Department of Electrical Engineering, Technical University of Denmark, Kgs Lyngby, Denmark.,GE Healthcare, Broendby, Denmark
| | - Annette Frank
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Axel Haase
- Technische Universität München, Institute of Medical Engineering, Munich, Germany
| | - Markus Schwaiger
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | |
Collapse
|
126
|
Swietach P, Vaughan-Jones RD, Harris AL, Hulikova A. The chemistry, physiology and pathology of pH in cancer. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130099. [PMID: 24493747 PMCID: PMC3917353 DOI: 10.1098/rstb.2013.0099] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell survival is conditional on the maintenance of a favourable acid-base balance (pH). Owing to intensive respiratory CO2 and lactic acid production, cancer cells are exposed continuously to large acid-base fluxes, which would disturb pH if uncorrected. The large cellular reservoir of H(+)-binding sites can buffer pH changes but, on its own, is inadequate to regulate intracellular pH. To stabilize intracellular pH at a favourable level, cells control trans-membrane traffic of H(+)-ions (or their chemical equivalents, e.g. ) using specialized transporter proteins sensitive to pH. In poorly perfused tumours, additional diffusion-reaction mechanisms, involving carbonic anhydrase (CA) enzymes, fine-tune control extracellular pH. The ability of H(+)-ions to change the ionization state of proteins underlies the exquisite pH sensitivity of cellular behaviour, including key processes in cancer formation and metastasis (proliferation, cell cycle, transformation, migration). Elevated metabolism, weakened cell-to-capillary diffusive coupling, and adaptations involving H(+)/H(+)-equivalent transporters and extracellular-facing CAs give cancer cells the means to manipulate micro-environmental acidity, a cancer hallmark. Through genetic instability, the cellular apparatus for regulating and sensing pH is able to adapt to extracellular acidity, driving disease progression. The therapeutic potential of disturbing this sequence by targeting H(+)/H(+)-equivalent transporters, buffering or CAs is being investigated, using monoclonal antibodies and small-molecule inhibitors.
Collapse
Affiliation(s)
- Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Adrian L. Harris
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
127
|
Bustamante FL, Miranda FS, Castro FA, Resende JA, Pereira MD, Lanznaster M. A study on the properties and reactivity of naphthoquinone–cobalt(III) prototypes for bioreductive prodrugs. J Inorg Biochem 2014; 132:37-44. [DOI: 10.1016/j.jinorgbio.2013.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/14/2013] [Accepted: 11/19/2013] [Indexed: 12/17/2022]
|
128
|
Pilot study of Iopamidol-based quantitative pH imaging on a clinical 3T MR scanner. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2014; 27:477-85. [DOI: 10.1007/s10334-014-0433-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 12/22/2022]
|
129
|
Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin. PLoS One 2014; 9:e88193. [PMID: 24516610 PMCID: PMC3916404 DOI: 10.1371/journal.pone.0088193] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 01/05/2014] [Indexed: 12/02/2022] Open
Abstract
Intrinsic resistance to cytotoxic drugs has been a main issue in cancer therapy for decades. Microenvironmental acidity is a simple while highly efficient mechanism of chemoresistance, exploited through impairment of drug delivery. The latter is achieved by extracellular protonation and/or sequestration into acidic vesicles. This study investigates the importance of extracellular acidosis and nanovesicle (exosome) release in the resistance of human tumour cell to cisplatin (CisPt); in parallel to proton pump inhibitors (PPI) ability of interfering with these tumour cell features. The results showed that CisPt uptake by human tumour cells was markedly impaired by low pH conditions. Moreover, exosomes purified from supernatants of these cell cultures contained various amounts of CisPt, which correlated to the pH conditions of the culture medium. HPLC-Q-ICP-MS analysis revealed that exosome purified from tumour cell culture supernatants contained CisPt in its native form. PPI pre-treatment increased cellular uptake of CisPt, as compared to untreated cells, in an acidic-depend manner. Furthermore, it induced a clear inhibition of exosome release by tumour cells. Human tumours obtained from xenografts pretreated with PPI contained more CisPt as compared to tumours from xenografts treated with CisPt alone. Further analysis showed that in vivo PPI treatment induced a clear reduction in the plasmatic levels of tumour-derived exosomes which also contained lower level of CisPt. Altogether, these findings point to the identification of a double mechanism that human malignant melanoma use in resisting to a dreadful cellular poison such as cisplatin. This framework of resistance includes both low pH-dependent extracellular sequestration and an exosome-mediated elimination. Both mechanisms are markedly impaired by proton pump inhibition, leading to an increased CisPt-dependent cytotoxicity.
Collapse
|
130
|
Abstract
The metabolism of cancer cells differs substantially from normal cells, including ion transport. Although this phenomenon has been long recognized, ion transporters have not been viewed as suitable therapeutic targets. However, the acidic pH values present in tumours which are well outside of normal limits are now becoming recognized as an important therapeutic target. Carbonic anhydrase IX (CAIX) is fundamental to tumour pH regulation. CAIX is commonly expressed in cancer, but lowly expressed in normal tissues and that presents an attractive target. Here, we discuss the possibilities of exploiting the acidic, hypoxic tumour environment as possible target for therapy. Additionally, clinical experience with CAIX targeting in cancer patients is discussed.
Collapse
Affiliation(s)
- E Oosterwijk
- Department of Urology, 267 Experimental Urology, Radboud University Medical Center, , PO Box 9101, Nijmegen 6500 HB, The Netherlands
| | | |
Collapse
|
131
|
Pellegrini P, Strambi A, Zipoli C, Hägg-Olofsson M, Buoncervello M, Linder S, De Milito A. Acidic extracellular pH neutralizes the autophagy-inhibiting activity of chloroquine: implications for cancer therapies. Autophagy 2014; 10:562-71. [PMID: 24492472 DOI: 10.4161/auto.27901] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acidic pH is an important feature of tumor microenvironment and a major determinant of tumor progression. We reported that cancer cells upregulate autophagy as a survival mechanism to acidic stress. Inhibition of autophagy by administration of chloroquine (CQ) in combination anticancer therapies is currently evaluated in clinical trials. We observed in 3 different human cancer cell lines cultured at acidic pH that autophagic flux is not blocked by CQ. This was consistent with a complete resistance to CQ toxicity in cells cultured in acidic conditions. Conversely, the autophagy-inhibiting activity of Lys-01, a novel CQ derivative, was still detectable at low pH. The lack of CQ activity was likely dependent on a dramatically reduced cellular uptake at acidic pH. Using cell lines stably adapted to chronic acidosis we could confirm that CQ lack of activity was merely caused by acidic pH. Moreover, unlike CQ, Lys-01 was able to kill low pH-adapted cell lines, although higher concentrations were required as compared with cells cultured at normal pH conditions. Notably, buffering medium pH in low pH-adapted cell lines reverted CQ resistance. In vivo analysis of tumors treated with CQ showed that accumulation of strong LC3 signals was observed only in normoxic areas but not in hypoxic/acidic regions. Our observations suggest that targeting autophagy in the tumor environment by CQ may be limited to well-perfused regions but not achieved in acidic regions, predicting possible limitations in efficacy of CQ in antitumor therapies.
Collapse
Affiliation(s)
- Paola Pellegrini
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| | - Angela Strambi
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| | - Chiara Zipoli
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| | - Maria Hägg-Olofsson
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| | - Maria Buoncervello
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| | - Stig Linder
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| | - Angelo De Milito
- Cancer Center Karolinska; Department of Oncology-Pathology; Karolinska Institute; Stockholm, Sweden
| |
Collapse
|
132
|
Vaughan RA, Gannon NP, Garcia-Smith R, Licon-Munoz Y, Barberena MA, Bisoffi M, Trujillo KA. β-alanine suppresses malignant breast epithelial cell aggressiveness through alterations in metabolism and cellular acidity in vitro. Mol Cancer 2014; 13:14. [PMID: 24460609 PMCID: PMC3937146 DOI: 10.1186/1476-4598-13-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 01/21/2014] [Indexed: 01/15/2023] Open
Abstract
Background Deregulated energetics is a property of most cancer cells. This phenomenon, known as the Warburg Effect or aerobic glycolysis, is characterized by increased glucose uptake, lactate export and extracellular acidification, even in the presence of oxygen. β-alanine is a non-essential amino acid that has previously been shown to be metabolized into carnosine, which functions as an intracellular buffer. Because of this buffering capacity, we investigated the effects of β-alanine on the metabolic cancerous phenotype. Methods Non-malignant MCF-10a and malignant MCF-7 breast epithelial cells were treated with β-alanine at 100 mM for 24 hours. Aerobic glycolysis was quantified by measuring extracellular acidification rate (ECAR) and oxidative metabolism was quantified by measuring oxygen consumption rate (OCR). mRNA of metabolism-related genes was quantified by qRT-PCR with corresponding protein expression quantified by immunoblotting, or by flow cytometry which was verified by confocal microscopy. Mitochondrial content was quantified using a mitochondria-specific dye and measured by flow cytometry. Results Cells treated with β-alanine displayed significantly suppressed basal and peak ECAR (aerobic glycolysis), with simultaneous increase in glucose transporter 1 (GLUT1). Additionally, cells treated with β-alanine exhibited significantly reduced basal and peak OCR (oxidative metabolism), which was accompanied by reduction in mitochondrial content with subsequent suppression of genes which promote mitochondrial biosynthesis. Suppression of glycolytic and oxidative metabolism by β-alanine resulted in the reduction of total metabolic rate, although cell viability was not affected. Because β-alanine treatment reduces extracellular acidity, a constituent of the invasive microenvironment that promotes progression, we investigated the effect of β-alanine on breast cell viability and migration. β-alanine was shown to reduce both cell migration and proliferation without acting in a cytotoxic fashion. Moreover, β-alanine significantly increased malignant cell sensitivity to doxorubicin, suggesting a potential role as a co-therapeutic agent. Conclusion Taken together, our results suggest that β-alanine may elicit several anti-tumor effects. Our observations support the need for further investigation into the mechanism(s) of action and specificity of β-alanine as a co-therapeutic agent in the treatment of breast tumors.
Collapse
Affiliation(s)
- Roger A Vaughan
- Department of Health, Exercise and Sports Science, University of New Mexico, Albuquerque, NM 87131, USA.
| | | | | | | | | | | | | |
Collapse
|
133
|
Han L, Liu M, Ye D, Zhang N, Lim E, Lu J, Jiang C. Tumor cell membrane-targeting pH-dependent electron donor-acceptor fluorescence systems with low background signals. Biomaterials 2014; 35:2952-60. [PMID: 24388814 DOI: 10.1016/j.biomaterials.2013.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 12/11/2013] [Indexed: 11/25/2022]
Abstract
Minimizing the background signal is crucial for developing tumor-imaging techniques with sufficient specificity and sensitivity. Here we use pH difference between healthy tissues and tumor and tumor targeting delivery to achieve this goal. We synthesize fluorophore-dopamine conjugate as pH-dependent electron donor-acceptor fluorescence system. Fluorophores are highly sensitive to electron-transfer processes, which can alter their optical properties. The intrinsic redox properties of dopamine are oxidation of hydroquinone to quinone at basic pH and reduction of quinone to hydroquinone at acidic pH. Quinone can accept electron then quench fluorescence. We design tumor cell membrane-targeting carrier for delivery. We demonstrate quenched fluorophore-quinone can be specially transferred to tumor extracellular environment and tumor-accumulated fluorophore can be activated by acidic pH. These tumor-targeting pH-dependent electron donor-acceptor fluorescence systems may offer new opportunity for developing tumor-imaging techniques.
Collapse
Affiliation(s)
- Liang Han
- Key Laboratory of Molecular Engineering of Polymers of Ministry of Education, Fudan University, Shanghai 201203, China; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mingming Liu
- Department of Medical Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Deyong Ye
- Department of Medical Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ning Zhang
- In Vivo Imaging R&D, Caliper Life Sciences, A Perkin Elmer Company, Alameda, CA 94501, USA
| | - Ed Lim
- In Vivo Imaging R&D, Caliper Life Sciences, A Perkin Elmer Company, Alameda, CA 94501, USA
| | - Jing Lu
- In Vivo Imaging R&D, Caliper Life Sciences, A Perkin Elmer Company, Alameda, CA 94501, USA
| | - Chen Jiang
- Key Laboratory of Molecular Engineering of Polymers of Ministry of Education, Fudan University, Shanghai 201203, China; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
134
|
Tafreshi NK, Lloyd MC, Bui MM, Gillies RJ, Morse DL. Carbonic anhydrase IX as an imaging and therapeutic target for tumors and metastases. Subcell Biochem 2014; 75:221-54. [PMID: 24146382 DOI: 10.1007/978-94-007-7359-2_12] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Carbonic anhydrase IX (CAIX) which is a zinc containing metalloprotein, efficiently catalyzes the reversible hydration of carbon dioxide. It is constitutively up-regulated in several cancer types and has an important role in tumor progression, acidification and metastasis. High expression of CAIX generally correlates with poor prognosis and is related to a decrease in the disease-free interval following successful therapy. Therefore, it is considered as a prognostic indicator in oncology.In this review, we describe CAIX regulation and its role in tumor hypoxia, acidification and metastasis. In addition, the molecular imaging of CAIX and its potential for use in cancer detection, diagnosis, staging, and for use in following therapy response is discussed. Both antibodies and small molecular weight compounds have been used for targeted imaging of CAIX expression. The use of CAIX expression as an attractive and promising candidate marker for systemic anticancer therapy is also discussed.
Collapse
Affiliation(s)
- Narges K Tafreshi
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| | | | | | | | | |
Collapse
|
135
|
Han L, Guo Y, Ma H, He X, Kuang Y, Zhang N, Lim E, Zhou W, Jiang C. Acid active receptor-specific peptide ligand for in vivo tumor-targeted delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3647-3658. [PMID: 23649993 DOI: 10.1002/smll.201300279] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Indexed: 06/02/2023]
Abstract
Targeting therapy of tumors in their early stages is crucial to increase the survival rate of cancer patients. Currently most drug-delivery systems target the neoplasia through the tumor-associated receptors overexpressed on the cancer cell membrane. However, the expression of these receptors on normal cells and tissues is inevitable, which leads to unwanted accumulation and side effects. Characteristics of the tumor microenvironment, such as acidosis, are pervasive in almost all solid tumors and can be easily accessed. It is shown that the different extracellular pH value can be used to activate/inactivate the receptor-mediated endocytosis on tumor/normal cells. This idea is implemented by conjugating a shielding molecule at the terminus of a receptor-specific ligand via a pH-sensitive hydrazone bond. The acid-activated detachment of the shielding molecule and enhanced tumor/background accumulation ratio are demonstrated. These results suggest that acid active receptor-specific peptide ligand-modified tumor-targeting delivery systems have potential use in the treatment of tumors.
Collapse
Affiliation(s)
- Liang Han
- Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Harguindey S, Arranz JL, Polo Orozco JD, Rauch C, Fais S, Cardone RA, Reshkin SJ. Cariporide and other new and powerful NHE1 inhibitors as potentially selective anticancer drugs--an integral molecular/biochemical/metabolic/clinical approach after one hundred years of cancer research. J Transl Med 2013; 11:282. [PMID: 24195657 PMCID: PMC3826530 DOI: 10.1186/1479-5876-11-282] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/25/2013] [Indexed: 02/04/2023] Open
Abstract
In recent years an increasing number of publications have emphasized the growing importance of hydrogen ion dynamics in modern cancer research, from etiopathogenesis and treatment. A proton [H+]-related mechanism underlying the initiation and progression of the neoplastic process has been recently described by different research groups as a new paradigm in which all cancer cells and tissues, regardless of their origin and genetic background, have a pivotal energetic and homeostatic disturbance of their metabolism that is completely different from all normal tissues: an aberrant regulation of hydrogen ion dynamics leading to a reversal of the pH gradient in cancer cells and tissues (↑pHi/↓pHe, or “proton reversal”). Tumor cells survive their hostile microenvironment due to membrane-bound proton pumps and transporters, and their main defensive strategy is to never allow internal acidification because that could lead to their death through apoptosis. In this context, one of the primary and best studied regulators of both pHi and pHe in tumors is the Na+/H+ exchanger isoform 1 (NHE1). An elevated NHE1 activity can be correlated with both an increase in cell pH and a decrease in the extracellular pH of tumors, and such proton reversal is associated with the origin, local growth, activation and further progression of the metastatic process. Consequently, NHE1 pharmaceutical inhibition by new and potent NHE1 inhibitors represents a potential and highly selective target in anticancer therapy. Cariporide, being one of the better studied specific and powerful NHE1 inhibitors, has proven to be well tolerated by humans in the cardiological context, however some side-effects, mainly related to drug accumulation and cerebrovascular complications were reported. Thus, cariporide could become a new, slightly toxic and effective anticancer agent in different human malignancies.
Collapse
Affiliation(s)
- Salvador Harguindey
- Instituto de Biología Clínica y Metabolismo (IBCM), Postas 13-01004, Vitoria, Spain.
| | | | | | | | | | | | | |
Collapse
|
137
|
Han L, Ma H, Guo Y, Kuang Y, He X, Jiang C. pH-controlled delivery of nanoparticles into tumor cells. Adv Healthc Mater 2013; 2:1435-9. [PMID: 23564477 DOI: 10.1002/adhm.201300013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Indexed: 12/31/2022]
Abstract
Nanoparticles target tumor cells by pH-controlled means. Nanoparticles carry three synergistic delivery functions: 1) tumor tissue targeting by the EPR effect; 2) tumor cell targeting by pHLIP-mediated membrane-localization; and 3) tumor cell uptake by adsorptive-mediated endocytosis.
Collapse
Affiliation(s)
- Liang Han
- Key Laboratory of Smart Drug Delivery, Ministry of Education& PLA, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | | | | | | | | | | |
Collapse
|
138
|
Patel KJ, Lee C, Tan Q, Tannock IF. Use of the proton pump inhibitor pantoprazole to modify the distribution and activity of doxorubicin: a potential strategy to improve the therapy of solid tumors. Clin Cancer Res 2013; 19:6766-76. [PMID: 24141627 DOI: 10.1158/1078-0432.ccr-13-0128] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Limited drug distribution within solid tumors is an important cause of drug resistance. Basic drugs (e.g., doxorubicin) may be sequestered in acidic organelles, thereby limiting drug distribution to distal cells and diverting drugs from their target DNA. Here we investigate the effects of pantoprazole, a proton pump inhibitor, on doxorubicin uptake, and doxorubicin distribution and activity using in vitro and murine models. EXPERIMENTAL DESIGN Murine EMT-6 and human MCF-7 cells were treated with pantoprazole to evaluate changes in endosomal pH using fluorescence spectroscopy, and uptake of doxorubicin using flow cytometry. Effects of pantoprazole on tissue penetration of doxorubicin were evaluated in multilayered cell cultures (MCC), and in solid tumors using immunohistochemistry. Effects of pantoprazole to influence tumor growth delay and toxicity because of doxorubicin were evaluated in mice. RESULTS Pantoprazole (>200 μmol/L) increased endosomal pH in cells, and also increased nuclear uptake of doxorubicin. Pretreatment with pantoprazole increased tissue penetration of doxorubicin in MCCs. Pantoprazole improved doxorubicin distribution from blood vessels in solid tumors. Pantoprazole given before doxorubicin led to increased growth delay when given as single or multiple doses to mice bearing MCF7 xenografts. CONCLUSIONS Use of pantoprazole to enhance the distribution and cytotoxicity of anticancer drugs in solid tumors might be a novel treatment strategy to improve their therapeutic index.
Collapse
Affiliation(s)
- Krupa J Patel
- Authors' Affiliations: Department of Medical Biophysics and Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
139
|
Nwe K, Huang CH, Tsourkas A. Gd-labeled glycol chitosan as a pH-responsive magnetic resonance imaging agent for detecting acidic tumor microenvironments. J Med Chem 2013; 56:7862-9. [PMID: 24044414 DOI: 10.1021/jm4012565] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neoplastic lesions can create a hostile tumor microenvironment with low extracellular pH. It is commonly believed that these conditions can contribute to tumor progression as well as resistance to therapy. We report the development and characterization of a pH-responsive magnetic resonance imaging contrast agent for imaging the acidic tumor microenvironment. The preparation included the conjugation of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid 1-(2,5-dioxo-1-pyrrolidinyl) ester (DOTA-NHS) to the surface of a water-soluble glycol chitosan (GC) polymer, which contains pH-titrable primary amines, followed by gadolinium complexation (GC-NH2-GdDOTA). GC-NH2-GdDOTA had a chelate-to-polymer ratio of approximately1:24 and a molar relaxivity of 9.1 mM(-1) s(-1). GC-NH2-GdDOTA demonstrated pH-dependent cellular association in vitro compared to the control. It also generated a 2.4-fold enhancement in signal in tumor-bearing mice 2 h postinjection. These findings suggest that glycol chitosan coupled with contrast agents can provide important diagnostic information about the tumor microenvironment.
Collapse
Affiliation(s)
- Kido Nwe
- Department of Bioengineering, University of Pennsylvania , 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia 19104, Pennsylvania, United States
| | | | | |
Collapse
|
140
|
Ganguly A, Chakraborty P, Banerjee K, Choudhuri SK. The role of a Schiff base scaffold, N-(2-hydroxy acetophenone) glycinate-in overcoming multidrug resistance in cancer. Eur J Pharm Sci 2013; 51:96-109. [PMID: 24044945 DOI: 10.1016/j.ejps.2013.09.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/16/2013] [Accepted: 09/03/2013] [Indexed: 01/01/2023]
Abstract
Drug resistance is a problem that hinders the numerous successes of chemotherapeutic intervention of cancer and continues to be a major obstacle for cures. Till date, several attempts have been made to develop suitable multidrug resistance (MDR) reversing agents. But, throughout the clinical development of MDR reversing agents, patients repeatedly suffer from toxicities. So far, some anticancer activity of Schiff bases which are the condensation products of carbonyl compounds and primary amines and their metal complexes has been described. But, overcoming multidrug resistance, by the use of such small molecules still remain unexplored. Under this backdrop, in search of less toxic and more effective MDR reversing agents our laboratory has developed the different metal chelates of Schiff base N-(2-hydroxy acetophenone)glycinate (NG) which is structurally similar to azatyrosine [L-β-(5-hydroxy-2-pyridyl)-alanine] that inhibits tumor formation by deactivating the c-Raf-1 kinase and c-Ha-ras signalling pathway. A decade-long research proposes possible strategies to overcome MDR by exploiting the chemical nature of such metal chelates. In this review we have catalogued the success of metal chelates of NG to overcome MDR in cancer. The review depict that the problem of MDR can be circumvent by synchronized activation of immunogenic cell death pathways that utilize the components of a host's immune system to kill cancer cells in combination with other conventional strategies. The current wealth of preclinical information promises better understanding of the cellular processes underlying MDR reversing activity of metal derivatives of NG and thus exposes several cellular targets for rational designing of new generation of Schiff base metal chelates as MDR reversing agents.
Collapse
Affiliation(s)
- Avishek Ganguly
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, Kolkata, India
| | | | | | | |
Collapse
|
141
|
Kato Y, Ozawa S, Miyamoto C, Maehata Y, Suzuki A, Maeda T, Baba Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int 2013; 13:89. [PMID: 24004445 PMCID: PMC3849184 DOI: 10.1186/1475-2867-13-89] [Citation(s) in RCA: 885] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/29/2013] [Indexed: 12/18/2022] Open
Abstract
Acidic extracellular pH is a major feature of tumor tissue, extracellular acidification being primarily considered to be due to lactate secretion from anaerobic glycolysis. Clinicopathological evidence shows that transporters and pumps contribute to H+ secretion, such as the Na+/H+ exchanger, the H+-lactate co-transporter, monocarboxylate transporters, and the proton pump (H+-ATPase); these may also be associated with tumor metastasis. An acidic extracellular pH not only activates secreted lysosomal enzymes that have an optimal pH in the acidic range, but induces the expression of certain genes of pro-metastatic factors through an intracellular signaling cascade that is different from hypoxia. In addition to lactate, CO2 from the pentose phosphate pathway is an alternative source of acidity, showing that hypoxia and extracellular acidity are, while being independent from each other, deeply associated with the cellular microenvironment. In this article, the importance of an acidic extracellular pH as a microenvironmental factor participating in tumor progression is reviewed.
Collapse
Affiliation(s)
- Yasumasa Kato
- Department of Oral Function and Molecular Biology, Ohu University School of Dentistry, 963-8611, Koriyama, Japan.
| | | | | | | | | | | | | |
Collapse
|
142
|
Maxwell SA, Mousavi-Fard S. Non-Hodgkin's B-cell lymphoma: advances in molecular strategies targeting drug resistance. Exp Biol Med (Maywood) 2013; 238:971-90. [PMID: 23986223 DOI: 10.1177/1535370213498985] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Non-Hodgkin's lymphoma (NHL) is a heterogeneous class of cancers displaying a diverse range of biological phenotypes, clinical behaviours and prognoses. Standard treatments for B-cell NHL are anthracycline-based combinatorial chemotherapy regimens composed of cyclophosphamide, doxorubicin, vincristine and prednisolone. Even though complete response rates of 40-50% with chemotherapy can be attained, a substantial proportion of patients relapse, resulting in 3-year overall survival rates of about 30%. Relapsed lymphomas are refractory to subsequent treatments with the initial chemotherapy regimen and can exhibit cross-resistance to a wide variety of anticancer drugs. The emergence of acquired chemoresistance thus poses a challenge in the clinic preventing the successful treatment and cure of disseminated B-cell lymphomas. Gene-expression analyses have increased our understanding of the molecular basis of chemotherapy resistance and identified rational targets for drug interventions to prevent and treat relapsed/refractory diffuse large B-cell lymphoma. Acquisition of drug resistance in lymphoma is in part driven by the inherent genetic heterogeneity and instability of the tumour cells. Due to the genetic heterogeneity of B-cell NHL, many different pathways leading to drug resistance have been identified. Successful treatment of chemoresistant NHL will thus require the rational design of combinatorial drugs targeting multiple pathways specific to different subtypes of B-cell NHL as well as the development of personalized approaches to address patient-to-patient genetic heterogeneity. This review highlights the new insights into the molecular basis of chemorefractory B-cell NHL that are facilitating the rational design of novel strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Steve A Maxwell
- Texas A&M Health Science Center, College Station, TX 77843-1114, USA
| | | |
Collapse
|
143
|
Expression and/or activity of the SVCT2 ascorbate transporter may be decreased in many aggressive cancers, suggesting potential utility for sodium bicarbonate and dehydroascorbic acid in cancer therapy. Med Hypotheses 2013; 81:664-70. [PMID: 23916956 DOI: 10.1016/j.mehy.2013.07.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 07/08/2013] [Accepted: 07/13/2013] [Indexed: 12/22/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a heterodimer transcription factor whose elevated activity in many cancers helps them to survive under hypoxic conditions and enhances their capacity to grow invasively, establish metastases, and survive chemo- or radiotherapy. Optimal intracellular levels of ascorbate suppress the level and transcriptional activity of HIF-1under normoxic or mildly hypoxic conditions by supporting the activity of proly and asparagyl hydroxylases that target HIF-1alpha. High intracellular ascorbate can also work in various ways to down-regulate activation of NF-kappaB which, like HIF-1 is constitutively active in many cancers and promotes aggressive behavior - in part by promoting transcription of HIF-1alpha. Yet recent evidence suggests that, even in the context of adequate ascorbate nutrition, the intracellular ascorbate content of many aggressive cancers may be supoptimal for effective HIF-1 control. This likely reflects low expression or activity of the SVCT2 ascorbate transporter. The expression of SVCT2 in cancers has so far received little study; but the extracellular acidity characteristic of many tumors would be expected to reduce the activity of this transporter, which has a mildly alkaline pH optimum. Unfortunately, since SVCT2 has a high affinity for ascorbate, and its activity is nearly saturated at normal healthy serum levels of this vitamin, increased oral administration of ascorbate would be unlikely to have much impact on the intracellular ascorbate content of tumors. However, cancers in which HIF-1 is active express high levels of glucose transporters such as GLUT-1, and these transporters can promote influx of dehydroascorbic acid (DHA) via facilitated diffusion; once inside the cell, DHA is rapidly reduced to ascorbate, which effectively is "trapped" within the cell. Hence, episodic intravenous infusions of modest doses of DHA may have potential for optimizing the intracellular ascorbate content of cancers, potentially rendering them less aggressive. Indeed, several published studies have concluded that parenteral DHA--sometimes in quite modest doses--can retard the growth of transplanted tumors in rodents. As an alternative or adjunctive strategy, oral administration of sodium bicarbonate, by normalizing the extracellular pH of tumors, has the potential to boost the activity of SCTV2 in tumor cells, thereby promoting increased ascorbate uptake. Indeed, the utility of oral sodium bicarbonate for suppressing metastasis formation in nude mice xenografted with a human breast cancer has been reported. Hence, oral sodium bicarbonate and intravenous DHA may have the potential to blunt the aggressiveness of certain cancers in which suboptimal intracellular ascorbate levels contribute to elevated HIF-1 activity.
Collapse
|
144
|
Investigating mechanisms of alkalinization for reducing primary breast tumor invasion. BIOMED RESEARCH INTERNATIONAL 2013; 2013:485196. [PMID: 23936808 PMCID: PMC3722989 DOI: 10.1155/2013/485196] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 06/16/2013] [Indexed: 01/15/2023]
Abstract
The extracellular pH (pHe) of many solid tumors is acidic as a result of glycolytic metabolism and poor perfusion. Acidity promotes invasion and enhances metastatic potential. Tumor acidity can be buffered by systemic administration of an alkaline agent such as sodium bicarbonate. Tumor-bearing mice maintained on sodium bicarbonate drinking water exhibit fewer metastases and survive longer than untreated controls. We predict this effect is due to inhibition of tumor invasion. Reducing tumor invasion should result in fewer circulating tumor cells (CTCs). We report that bicarbonate-treated MDA-MB-231 tumor-bearing mice exhibited significantly lower numbers of CTCs than untreated mice (P < 0.01). Tumor pHe buffering may reduce optimal conditions for enzymes involved in tumor invasion such as cathepsins and matrix metalloproteases (MMPs). To address this, we tested the effect of transient alkalinization on cathepsin and MMP activity using enzyme activatable fluorescence agents in mice bearing MDA-MB-231 mammary xenografts. Transient alkalinization significantly reduced the fluorescent signal of protease-specific activatable agents in vivo (P ≤ 0.003). Alkalinization, however, did not affect expression of carbonic anhydrase IX (CAIX). The findings suggest a possible mechanism in a live model system for breast cancer where systemic alkalinization slows the rate of invasion.
Collapse
|
145
|
Du X, Lei NY, Hu P, Lei Z, Ong DHC, Ge X, Zhang Z, Lam MHW. In vivo imaging of the morphology and changes in pH along the gastrointestinal tract of Japanese medaka by photonic band-gap hydrogel microspheres. Anal Chim Acta 2013; 787:193-202. [PMID: 23830439 DOI: 10.1016/j.aca.2013.05.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/27/2022]
Abstract
Colloidal crystalline microspheres with photonic band-gap properties responsive to media pH have been developed for in vivo imaging purposes. These colloidal crystalline microspheres were constructed from monodispersed core-shell nano-size particles with poly(styrene-co-acrylic acid) (PS-co-PAA) cores and poly(acrylic acid-co-N-isopropylacrylamide) (PAA-co-PNIPAM) hydrogel shells cross-linked by N,N'-methylenebisacrylamide. A significant shift in the photonic band-gap properties of these colloidal crystalline microspheres was observed in the pH range of 4-5. This was caused by the discontinuous volume phase transition of the hydrogel coating, due to the protonation/deprotonation of its acrylic acid moieties, on the core-shell nano-sized particles within the microspheres. The in vivo imaging capability of these pH-responsive photonic microspheres was demonstrated on a test organism - Japanese medaka, Oryzia latipes - in which the morphology and change in pH along their gastrointestinal (GI) tracts were revealed under an ordinary optical microscope. This work illustrates the potential of stimuli-responsive photonic band-gap materials in tissue-/organ-level in vivo bio-imaging.
Collapse
Affiliation(s)
- Xuemin Du
- Advanced Laboratory for Environmental Research & Technology, USTC-CityU, Suzhou 215123, China
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Macholl S, Morrison MS, Iveson P, Arbo BE, Andreev OA, Reshetnyak YK, Engelman DM, Johannesen E. In vivo pH imaging with (99m)Tc-pHLIP. Mol Imaging Biol 2013; 14:725-34. [PMID: 22371188 DOI: 10.1007/s11307-012-0549-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE A novel molecular imaging agent has been developed recently, which stains tissues of low extracellular pH [pH (low) insertion peptide, pHLIP(®)]. A pH-dependent process of peptide folding and insertion into cell membranes has been found in vitro. Targeting of acidic solid tumours has been demonstrated in vivo using fluorescence and PET labels. Here, we present proof of feasibility studies of pHLIP with a single-photon emission computed tomography (SPECT) label, (99m)Tc-AH114567, with focus on preclinical efficacy and imageability. PROCEDURES Lewis lung carcinoma, lymph node carcinoma of the prostate and prostate adenocarcinoma tumour xenografts were grown in mice and characterised by the angiogenesis marker (99m)Tc-NC100692 and by extracellular pH measurements with (31)P-MRS of 3-aminopropyl phosphonate. Biodistribution was assessed and CT/SPECT imaging performed. Oral administration of bicarbonate served as control. RESULTS AND CONCLUSION Tc-AH114567 can be obtained via a robust synthesis with good radiolabelling profile and improved formulation. The tracer retains the pH-dependent ability to insert into membranes and to target tumours with similar pharmacokinetics and efficacy that had been demonstrated earlier for pHLIP with optical or (64)Cu PET labels. Despite the inherent challenges of SPECT compared to optical and PET imaging, e.g., in terms of lower sensitivity, (99m)Tc-AH114567 shows adequate image quality and contrast. The main development need for transitioning SPECT labelled pHLIP into the clinic is more rapid background signal reduction, which will be the focus of a subsequent optimisation study.
Collapse
Affiliation(s)
- Sven Macholl
- Medical Diagnostics, The Grove Centre, GE Healthcare, GC/18, Amersham HP7 9LL, UK.
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Yamamoto N, Renfrew AK, Kim BJ, Bryce NS, Hambley TW. Dual targeting of hypoxic and acidic tumor environments with a cobalt(III) chaperone complex. J Med Chem 2012. [PMID: 23199008 DOI: 10.1021/jm3014713] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The rational design of prodrugs for selective accumulation and activation in tumor microenvironments is one of the most promising strategies for minimizing the toxicity of anticancer drugs. Manipulation of the charge of the prodrug represents a potential mechanism to selectively deliver the prodrug to the acidic tumor microenvironment. Here we present delivery of a fluorescent coumarin using a cobalt(III) chaperone to target hypoxic regions, and charged ligands for pH selectivity. Protonation or deprotonation of the complexes over a physiologically relevant pH range resulted in pH dependent accumulation of the fluorophore in colon cancer cells. Furthermore, in a spheroid solid tumor model, the anionic complexes exhibited preferential release of the fluorophore in the acidic/hypoxic region. By fine-tuning the physicochemical properties of the cobalt-chaperone moiety, we have demonstrated selective drug release in the acidic and hypoxic tumor microenvironment.
Collapse
Affiliation(s)
- Natsuho Yamamoto
- School of Chemistry, University of Sydney, 412C F11, Sydney 2006, New South Wales, Australia
| | | | | | | | | |
Collapse
|
148
|
Zheng D, Duan C, Zhang D, Jia L, Liu G, Liu Y, Wang F, Li C, Guo H, Zhang Q. Galactosylated chitosan nanoparticles for hepatocyte-targeted delivery of oridonin. Int J Pharm 2012; 436:379-86. [DOI: 10.1016/j.ijpharm.2012.06.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 06/04/2012] [Accepted: 06/15/2012] [Indexed: 12/21/2022]
|
149
|
Yu JX, Cui W, Bourke VA, Mason RP. 6-Trifluoromethylpyridoxine: novel (19)F NMR pH indicator for in vivo detection. J Med Chem 2012; 55:6814-21. [PMID: 22775397 PMCID: PMC3430128 DOI: 10.1021/jm300520q] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
pH plays an important role in tumor proliferation, angiogenesis, metabolic control, and the efficacy of cytotoxic therapy, and accurate noninvasive assessment of tumor pH promises to provide insight into developmental processes and prognostic information. In this paper, we report the design, synthesis, and characterization of two novel pH indicators 6-trifluoromethylpyridoxine 8 and α(4),α(5)-di-O-[3'-O-(β-d-glucopyranosyl)propyl]-6-trifluoromethylpyridoxine 17 and demonstrate 8 as an extracellular (19)F NMR pH probe to assess pH(e) of various tumors in vivo.
Collapse
Affiliation(s)
- Jian-Xin Yu
- Advanced Radiological Sciences, Department of Radiology, The University of Texas Southwestern Medical Center at Dallas , 5323 Harry Hines Boulevard, Dallas, Texas 75390-9058, United States.
| | | | | | | |
Collapse
|
150
|
Wang L, Ma W, Zhu L, Ye D, Li Y, Liu S, Li H, Zuo W, Li B, Ye W, Chen L. ClC-3 is a candidate of the channel proteins mediating acid-activated chloride currents in nasopharyngeal carcinoma cells. Am J Physiol Cell Physiol 2012; 303:C14-23. [DOI: 10.1152/ajpcell.00145.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acid-activated chloride currents have been reported in several cell types and may play important roles in regulation of cell function. However, the molecular identities of the channels that mediate the currents are not defined. In this study, activation of the acid-induced chloride current and the possible candidates of the acid-activated chloride channel were investigated in human nasopharyngeal carcinoma cells (CNE-2Z). A chloride current was activated when extracellular pH was reduced to 6.6 from 7.4. However, a further decrease of extracellular pH to 5.8 inhibited the current. The current was weakly outward-rectified and was suppressed by hypertonicity-induced cell shrinkage and by the chloride channel blockers 5-nitro-2–3-phenylpropylamino benzoic acid (NPPB), tamoxifen, and 4,4′-diisothiocyanatostilbene-2,2′-disulfonic acid disodium salt hydrate (DIDS). The permeability sequence of the channel to anions was I− > Br− > Cl− > gluconate−. Among the ClC chloride channels, ClC-3 and ClC-7 were strongly expressed in CNE-2Z cells. Knockdown of ClC-3 expression with ClC-3 small interfering (si)RNA prevented the activation of the acid-induced current, but silence of ClC-7 expression with ClC-7 siRNA did not significantly affect the current. The results suggest that the chloride channel mediating the acid-induced chloride current was volume sensitive. ClC-3 is a candidate of the channel proteins that mediate or regulate the acid-activated chloride current in nasopharyngeal carcinoma cells.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Wenbo Ma
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Linyan Zhu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China; and
| | - Dong Ye
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Yuan Li
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China; and
| | - Shanwen Liu
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Huarong Li
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Wanhong Zuo
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Bingxue Li
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China; and
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Lixin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China; and
| |
Collapse
|