1
|
Arias-Ramos N, Vieira C, Pérez-Carro R, López-Larrubia P. Integrative Magnetic Resonance Imaging and Metabolomic Characterization of a Glioblastoma Rat Model. Brain Sci 2024; 14:409. [PMID: 38790388 PMCID: PMC11118082 DOI: 10.3390/brainsci14050409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Glioblastoma (GBM) stands as the most prevalent and lethal malignant brain tumor, characterized by its highly infiltrative nature. This study aimed to identify additional MRI and metabolomic biomarkers of GBM and its impact on healthy tissue using an advanced-stage C6 glioma rat model. Wistar rats underwent a stereotactic injection of C6 cells (GBM group, n = 10) or cell medium (sham group, n = 4). A multiparametric MRI, including anatomical T2W and T1W images, relaxometry maps (T2, T2*, and T1), the magnetization transfer ratio (MTR), and diffusion tensor imaging (DTI), was performed. Additionally, ex vivo magnetic resonance spectroscopy (MRS) HRMAS spectra were acquired. The MRI analysis revealed significant differences in the T2 maps, T1 maps, MTR, and mean diffusivity parameters between the GBM tumor and the rest of the studied regions, which were the contralateral areas of the GBM rats and both regions of the sham rats (the ipsilateral and contralateral). The ex vivo spectra revealed markers of neuronal loss, apoptosis, and higher glucose uptake by the tumor. Notably, the myo-inositol and phosphocholine levels were elevated in both the tumor and the contralateral regions of the GBM rats compared to the sham rats, suggesting the effects of the tumor on the healthy tissue. The MRI parameters related to inflammation, cellularity, and tissue integrity, along with MRS-detected metabolites, serve as potential biomarkers for the tumor evolution, treatment response, and impact on healthy tissue. These techniques can be potent tools for evaluating new drugs and treatment targets.
Collapse
Affiliation(s)
| | | | | | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain; (N.A.-R.)
| |
Collapse
|
2
|
Deborne J, Benkhaled I, Bouchaud V, Pinaud N, Crémillieux Y. Implantable theranostic device for in vivo real-time NMR evaluation of drug impact in brain tumors. Sci Rep 2024; 14:4541. [PMID: 38402370 PMCID: PMC10894190 DOI: 10.1038/s41598-024-55269-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
The evaluation of the efficacy of a drug is a fundamental step in the development of new treatments or in personalized therapeutic strategies and patient management. Ideally, this evaluation should be rapid, possibly in real time, easy to perform and reliable. In addition, it should be associated with as few adverse effects as possible for the patient. In this study, we present a device designed to meet these goals for assessing therapeutic response. This theranostic device is based on the use of magnetic resonance imaging and spectroscopy for the diagnostic aspect and on the application of the convection-enhanced delivery technique for the therapeutic aspect. The miniaturized device is implantable and can be used in vivo in a target tissue. In this study, the device was applied to rodent glioma models with local administration of choline kinase inhibitor and acquisition of magnetic resonance images and spectra at 7 Tesla. The variations in the concentration of key metabolites measured by the device during the administration of the molecules demonstrate the relevance of the approach and the potential of the device.
Collapse
Affiliation(s)
- Justine Deborne
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France
| | - Imad Benkhaled
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France
| | - Véronique Bouchaud
- Centre de Résonance Magnétique des Systèmes Biologiques, Université de Bordeaux, UMR 5536, Bordeaux, France
| | - Noël Pinaud
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France
| | - Yannick Crémillieux
- Institut des Sciences Moléculaires, Université de Bordeaux, UMR 5255, Bordeaux, France.
| |
Collapse
|
3
|
Louise Kelly C, Wydrzynska M, Phelan MM, Osharovich S, Delikatny EJ, Sée V, Poptani H. Inhibition of glioblastoma cell proliferation and invasion by the choline-kinase inhibitor JAS239 varies with cell type and hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576078. [PMID: 38293093 PMCID: PMC10827177 DOI: 10.1101/2024.01.17.576078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background Elevated choline kinase alpha (ChoK) is observed in most solid tumours including glioblastomas (GBM), yet until recently, inhibitors of ChoK have demonstrated limited efficacy in GBM models. Given that hypoxia is associated with GBM therapy resistance, we hypothesised that tumour hypoxia could be responsible for such limitations. We therefore evaluated in GBM cells, the effect of hypoxia on the function of JAS239, a potent ChoK inhibitor. Methods Rodent (F98 and 9L) and human (U-87 MG and U-251 MG) GBM cell lines were subjected to 72 hours of hypoxia conditioning and treated with JAS239 for 24 hours. NMR metabolomic measurements and analyses were performed to evaluate the signalling pathways involved. In addition, cell proliferation, cell cycle progression and cell invasion were measured in cell monolayers and 3D spheroids, with or without JAS239 treatment in normoxic or hypoxic cells to assess how hypoxia affects JAS239 function. Results Hypoxia and JAS239 treatment led to significant changes in the cellular metabolic pathways, specifically the phospholipid and glycolytic pathways associated with a reduction in cell proliferation via induced cell cycle arrest. Interestingly, JAS239 also impaired GBM invasion. However, JAS239 effects were variable depending on the cell line, reflecting the inherent heterogeneity observed in GBMs. Conclusion Our findings indicate that JAS239 and hypoxia can deregulate cellular metabolism, inhibit proliferation and alter cell invasion. These results may be useful for the design of new therapeutic strategies based on ChoK inhibition that can act on multiple pro-tumorigenic features.
Collapse
Affiliation(s)
- Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Martyna Wydrzynska
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- High field NMR facility, Department of Biochemistry & Systems Biology, University of Liverpool, UK
| | - Sofya Osharovich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Violaine Sée
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
4
|
Bhaduri S, Kelly CL, Lesbats C, Sharkey J, Ressel L, Mukherjee S, Platt MD, Delikatny EJ, Poptani H. Metabolic changes in glioblastomas in response to choline kinase inhibition: In vivo MRS in rodent models. NMR IN BIOMEDICINE 2023; 36:e4855. [PMID: 36269130 PMCID: PMC10078495 DOI: 10.1002/nbm.4855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Changes in glioblastoma (GBM) metabolism was investigated in response to JAS239, a choline kinase inhibitor, using MRS. In addition to the inhibition of phosphocholine synthesis, we investigated changes in other key metabolic pathways associated with GBM progression and treatment response. Three syngeneic rodent models of GBM were used: F98 (N = 12) and 9L (N = 8) models in rats and GL261 (N = 10) in mice. Rodents were intracranially injected with GBM cells in the right cortex and tumor growth was monitored using T2 -weighted images. Animals were treated once daily with intraperitoneal injections of 4 mg/kg JAS239 (F98 rats, n = 6; 9L rats, n = 6; GL261 mice, n = 5) or saline (control group, F98 rats, n = 6; 9L rats, n = 2; GL261 mice, n = 5) for five consecutive days. Single voxel spectra were acquired on Days 0 (T0, baseline) and 6 (T6, end of treatment) from the tumor as well as the contralateral normal brain using a PRESS sequence. Changes in metabolite ratios (tCho/tCr, tCho/NAA, mI/tCr, Glx/tCr and (Lip + Lac)/Cr) were used to assess metabolic pathway alterations in response to JAS239. Tumor growth arrest was noted in all models in response to JAS239 treatment compared with saline-treated animals, with a significant reduction (p < 0.05) in the F98 model. A reduction in tCho/tCr was observed with JAS239 treatment in all GBM models, indicating reduced phospholipid metabolism, with the highest reduction in 9L followed by GL261 and F98 tumors. A significant reduction (p < 0.05) in the tCho/NAA ratio was observed in the 9L model. A significant reduction in mI/tCr (p < 0.05) was found in JAS239-treated F98 tumors compared with the saline-treated animals. A non-significant trend of reduction in Glx/tCr was observed only in F98 and 9L tumors. JAS239-treated F98 tumors also showed a significant increase in Lip + Lac (p < 0.05), indicating increased cell death. This study demonstrated the utility of MRS in assessing metabolic changes in GBM in response to choline kinase inhibition.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Lorenzo Ressel
- Department of Veterinary Anatomy Physiology and PathologyUniversity of LiverpoolChesterUK
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Mark David Platt
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
5
|
Miriam Jose A, Rasool M. Choline kinase: An underappreciated rheumatoid arthritis therapeutic target. Life Sci 2022; 309:121031. [DOI: 10.1016/j.lfs.2022.121031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022]
|
6
|
Zhao D, Grist JT, Rose HEL, Davies NP, Wilson M, MacPherson L, Abernethy LJ, Avula S, Pizer B, Gutierrez DR, Jaspan T, Morgan PS, Mitra D, Bailey S, Sawlani V, Arvanitis TN, Sun Y, Peet AC. Metabolite selection for machine learning in childhood brain tumour classification. NMR IN BIOMEDICINE 2022; 35:e4673. [PMID: 35088473 DOI: 10.1002/nbm.4673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
MRS can provide high accuracy in the diagnosis of childhood brain tumours when combined with machine learning. A feature selection method such as principal component analysis is commonly used to reduce the dimensionality of metabolite profiles prior to classification. However, an alternative approach of identifying the optimal set of metabolites has not been fully evaluated, possibly due to the challenges of defining this for a multi-class problem. This study aims to investigate metabolite selection from in vivo MRS for childhood brain tumour classification. Multi-site 1.5 T and 3 T cohorts of patients with a brain tumour and histological diagnosis of ependymoma, medulloblastoma and pilocytic astrocytoma were retrospectively evaluated. Dimensionality reduction was undertaken by selecting metabolite concentrations through multi-class receiver operating characteristics and compared with principal component analysis. Classification accuracy was determined through leave-one-out and k-fold cross-validation. Metabolites identified as crucial in tumour classification include myo-inositol (P < 0.05, AUC = 0 . 81 ± 0 . 01 ), total lipids and macromolecules at 0.9 ppm (P < 0.05, AUC = 0 . 78 ± 0 . 01 ) and total creatine (P < 0.05, AUC = 0 . 77 ± 0 . 01 ) for the 1.5 T cohort, and glycine (P < 0.05, AUC = 0 . 79 ± 0 . 01 ), total N-acetylaspartate (P < 0.05, AUC = 0 . 79 ± 0 . 01 ) and total choline (P < 0.05, AUC = 0 . 75 ± 0 . 01 ) for the 3 T cohort. Compared with the principal components, the selected metabolites were able to provide significantly improved discrimination between the tumours through most classifiers (P < 0.05). The highest balanced classification accuracy determined through leave-one-out cross-validation was 85% for 1.5 T 1 H-MRS through support vector machine and 75% for 3 T 1 H-MRS through linear discriminant analysis after oversampling the minority. The study suggests that a group of crucial metabolites helps to achieve better discrimination between childhood brain tumours.
Collapse
Affiliation(s)
- Dadi Zhao
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - James T Grist
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Heather E L Rose
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
| | - Nigel P Davies
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
- Imaging and Medical Physics, University Hospitals Birmingham, Birmingham, UK
| | - Martin Wilson
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | | | | | | | - Barry Pizer
- Paediatric Oncology, Alder Hey Children's Hospital, Liverpool, UK
| | - Daniel R Gutierrez
- Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
- Medical Physics, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Tim Jaspan
- Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
- Neuroradiology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Paul S Morgan
- Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
- Medical Physics, Nottingham University Hospitals NHS Trust, Nottingham, UK
- Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Dipayan Mitra
- Neuroradiology, The Newcastle upon Tyne Hospitals, Newcastle upon Tyne, UK
| | - Simon Bailey
- Paediatric Oncology, Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Vijay Sawlani
- Radiology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
- School of Psychology, University of Birmingham, Birmingham, UK
| | - Theodoros N Arvanitis
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
- Institute of Digital Healthcare, WMG, University of Warwick, Coventry, UK
| | - Yu Sun
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
- University of Birmingham and Southeast University Joint Research Centre for Biomedical Engineering, Suzhou, China
| | - Andrew C Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Oncology, Birmingham Children's Hospital, Birmingham, UK
| |
Collapse
|
7
|
Saito RDF, Andrade LNDS, Bustos SO, Chammas R. Phosphatidylcholine-Derived Lipid Mediators: The Crosstalk Between Cancer Cells and Immune Cells. Front Immunol 2022; 13:768606. [PMID: 35250970 PMCID: PMC8889569 DOI: 10.3389/fimmu.2022.768606] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/13/2022] [Indexed: 01/16/2023] Open
Abstract
To become resistant, cancer cells need to activate and maintain molecular defense mechanisms that depend on an energy trade-off between resistance and essential functions. Metabolic reprogramming has been shown to fuel cell growth and contribute to cancer drug resistance. Recently, changes in lipid metabolism have emerged as an important driver of resistance to anticancer agents. In this review, we highlight the role of choline metabolism with a focus on the phosphatidylcholine cycle in the regulation of resistance to therapy. We analyze the contribution of phosphatidylcholine and its metabolites to intracellular processes of cancer cells, both as the major cell membrane constituents and source of energy. We further extended our discussion about the role of phosphatidylcholine-derived lipid mediators in cellular communication between cancer and immune cells within the tumor microenvironment, as well as their pivotal role in the immune regulation of therapeutic failure. Changes in phosphatidylcholine metabolism are part of an adaptive program activated in response to stress conditions that contribute to cancer therapy resistance and open therapeutic opportunities for treating drug-resistant cancers.
Collapse
Affiliation(s)
- Renata de Freitas Saito
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Luciana Nogueira de Sousa Andrade
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Silvina Odete Bustos
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Kumar M, Nanga RPR, Verma G, Wilson N, Brisset JC, Nath K, Chawla S. Emerging MR Imaging and Spectroscopic Methods to Study Brain Tumor Metabolism. Front Neurol 2022; 13:789355. [PMID: 35370872 PMCID: PMC8967433 DOI: 10.3389/fneur.2022.789355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) provides a non-invasive biochemical profile of brain tumors. The conventional 1H-MRS methods present a few challenges mainly related to limited spatial coverage and low spatial and spectral resolutions. In the recent past, the advent and development of more sophisticated metabolic imaging and spectroscopic sequences have revolutionized the field of neuro-oncologic metabolomics. In this review article, we will briefly describe the scientific premises of three-dimensional echoplanar spectroscopic imaging (3D-EPSI), two-dimensional correlation spectroscopy (2D-COSY), and chemical exchange saturation technique (CEST) MRI techniques. Several published studies have shown how these emerging techniques can significantly impact the management of patients with glioma by determining histologic grades, molecular profiles, planning treatment strategies, and assessing the therapeutic responses. The purpose of this review article is to summarize the potential clinical applications of these techniques in studying brain tumor metabolism.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Ravi Prakash Reddy Nanga
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Gaurav Verma
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Neil Wilson
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | | | - Kavindra Nath
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Sanjeev Chawla
| |
Collapse
|
9
|
Bhaduri S, Lesbats C, Sharkey J, Kelly CL, Mukherjee S, Taylor A, Delikatny EJ, Kim SG, Poptani H. Assessing Tumour Haemodynamic Heterogeneity and Response to Choline Kinase Inhibition Using Clustered Dynamic Contrast Enhanced MRI Parameters in Rodent Models of Glioblastoma. Cancers (Basel) 2022; 14:cancers14051223. [PMID: 35267531 PMCID: PMC8909848 DOI: 10.3390/cancers14051223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 12/04/2022] Open
Abstract
To investigate the utility of DCE-MRI derived pharmacokinetic parameters in evaluating tumour haemodynamic heterogeneity and treatment response in rodent models of glioblastoma, imaging was performed on intracranial F98 and GL261 glioblastoma bearing rodents. Clustering of the DCE-MRI-based parametric maps (using Tofts, extended Tofts, shutter speed, two-compartment, and the second generation shutter speed models) was performed using a hierarchical clustering algorithm, resulting in areas with poor fit (reflecting necrosis), low, medium, and high valued pixels representing parameters Ktrans, ve, Kep, vp, τi and Fp. There was a significant increase in the number of necrotic pixels with increasing tumour volume and a significant correlation between ve and tumour volume suggesting increased extracellular volume in larger tumours. In terms of therapeutic response in F98 rat GBMs, a sustained decrease in permeability and perfusion and a reduced cell density was observed during treatment with JAS239 based on Ktrans, Fp and ve as compared to control animals. No significant differences in these parameters were found for the GL261 tumour, indicating that this model may be less sensitive to JAS239 treatment regarding changes in vascular parameters. This study demonstrates that region-based clustered pharmacokinetic parameters derived from DCE-MRI may be useful in assessing tumour haemodynamic heterogeneity with the potential for assessing therapeutic response.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
| | - Arthur Taylor
- Department of Molecular Physiology & Cell Signalling, University of Liverpool, Liverpool L69 3BX, UK;
| | - Edward J. Delikatny
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Sungheon G. Kim
- Department of Radiology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (S.B.); (C.L.); (J.S.); (C.L.K.); (S.M.)
- Correspondence:
| |
Collapse
|
10
|
Quartieri F, Nesi M, Avanzi NR, Borghi D, Casale E, Corti E, Cucchi U, Donati D, Fasolini M, Felder ER, Galvani A, Giorgini ML, Lomolino A, Menichincheri M, Orrenius C, Perrera C, Re Depaolini S, Riccardi-Sirtori F, Salsi E, Isacchi A, Gnocchi P. Identification of unprecedented ATP-competitive choline kinase inhibitors. Bioorg Med Chem Lett 2021; 51:128310. [PMID: 34416377 DOI: 10.1016/j.bmcl.2021.128310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/22/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
In this article we describe the identification of unprecedented ATP-competitive ChoKα inhibitors starting from initial hit NMS-P830 that binds to ChoKα in an ATP concentration-dependent manner. This result is confirmed by the co-crystal structure of NMS-P830 in complex with Δ75-ChoKα. NMS-P830 is able to inhibit ChoKα in cells resulting in the reduction of intracellular phosphocholine formation. A structure-based medicinal chemistry program resulted in the identification of selective compounds that have good biochemical activity, solubility and metabolic stability and are suitable for further optimization. The ChoKα inhibitors disclosed in this article demonstrate for the first time the possibility to inhibit ChoKα with ATP-competitive compounds.
Collapse
Affiliation(s)
- Francesca Quartieri
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy.
| | - Marcella Nesi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Nilla R Avanzi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Daniela Borghi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Elena Casale
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Emiliana Corti
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Ulisse Cucchi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Daniele Donati
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Marina Fasolini
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Eduard R Felder
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Arturo Galvani
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Maria L Giorgini
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Antonio Lomolino
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | | | - Christian Orrenius
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Claudia Perrera
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | | | | | - Enea Salsi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Antonella Isacchi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| | - Paola Gnocchi
- Nerviano Medical Sciences Srl, Viale Pasteur 10, 20014 Nerviano (MI), Italy
| |
Collapse
|
11
|
Rubtsova NI, Hart MC, Arroyo AD, Osharovich SA, Liebov BK, Miller J, Yuan M, Cochran JM, Chong S, Yodh AG, Busch TM, Delikatny EJ, Anikeeva N, Popov AV. NIR Fluorescent Imaging and Photodynamic Therapy with a Novel Theranostic Phospholipid Probe for Triple-Negative Breast Cancer Cells. Bioconjug Chem 2021; 32:1852-1863. [PMID: 34139845 DOI: 10.1021/acs.bioconjchem.1c00295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
New exogenous probes are needed for both imaging diagnostics and therapeutics. Here, we introduce a novel nanocomposite near-infrared (NIR) fluorescent imaging probe and test its potency as a photosensitizing agent for photodynamic therapy (PDT) against triple-negative breast cancer cells. The active component in the nanocomposite is a small molecule, pyropheophorbide a-phosphatidylethanolamine-QSY21 (Pyro-PtdEtn-QSY), which is imbedded into lipid nanoparticles for transport in the body. The probe targets abnormal choline metabolism in cancer cells; specifically, the overexpression of phosphatidylcholine-specific phospholipase C (PC-PLC) in breast, prostate, and ovarian cancers. Pyro-PtdEtn-QSY consists of a NIR fluorophore and a quencher, attached to a PtdEtn moiety. It is selectively activated by PC-PLC resulting in enhanced fluorescence in cancer cells compared to normal cells. In our in vitro investigation, four breast cancer cell lines showed higher probe activation levels than noncancerous control cells, immortalized human mammary gland cells, and normal human T cells. Moreover, the ability of this nanocomposite to function as a sensitizer in PDT experiments on MDA-MB-231 cells suggests that the probe is promising as a theranostic agent.
Collapse
Affiliation(s)
- Natalia I Rubtsova
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| | - Michael C Hart
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| | - Alejandro D Arroyo
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| | - Sofya A Osharovich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| | - Benjamin K Liebov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| | - Joann Miller
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg 421, Philadelphia, Pennsylvania 19104, United States
| | - Min Yuan
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg 421, Philadelphia, Pennsylvania 19104, United States
| | - Jeffrey M Cochran
- Department of Physics and Astronomy, University of Pennsylvania, 3231 Walnut Street, Philadelphia, Pennsylvania 19104, United States
| | - Sanghoon Chong
- Department of Physics and Astronomy, University of Pennsylvania, 3231 Walnut Street, Philadelphia, Pennsylvania 19104, United States
| | - Arjun G Yodh
- Department of Physics and Astronomy, University of Pennsylvania, 3231 Walnut Street, Philadelphia, Pennsylvania 19104, United States
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Bldg 421, Philadelphia, Pennsylvania 19104, United States
| | - E James Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| | - Nadia Anikeeva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Anatoliy V Popov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
12
|
Simultaneous Recording of the Uptake and Conversion of Glucose and Choline in Tumors by Deuterium Metabolic Imaging. Cancers (Basel) 2021; 13:cancers13164034. [PMID: 34439188 PMCID: PMC8394025 DOI: 10.3390/cancers13164034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Tumors increase their glucose and choline uptake to support growth. These properties are employed to detect and identify tumors in the body by imaging the uptake of radio-isotope analogs of these compounds. In this study we show that deuterium metabolic imaging (DMI) (a new MRI method to image metabolites using non-radioactive labeling with deuterium) can image choline uptake in tumors. Furthermore, we demonstrate that DMI can image the tumor uptake of choline and glucose (and additionally its metabolic conversion) simultaneously, in contrast to radio-isotope imaging, which only assesses the uptake of one radio-isotope labeled compound at a time. For these reasons (and also because DMI is relatively simple and can be combined with other MR methods), it is a promising modality for a more specific tumor characterization than by separate imaging of the uptake of radio-isotope labeled glucose or choline. Abstract Increased glucose and choline uptake are hallmarks of cancer. We investigated whether the uptake and conversion of [2H9]choline alone and together with that of [6,6′-2H2]glucose can be assessed in tumors via deuterium metabolic imaging (DMI) after administering these compounds. Therefore, tumors with human renal carcinoma cells were grown subcutaneously in mice. Isoflurane anesthetized mice were IV infused in the MR magnet for ~20 s with ~0.2 mL solutions containing either [2H9]choline (0.05 g/kg) alone or together with [6,6′-2H2]glucose (1.3 g/kg). 2H MR was performed on a 11.7T MR system with a home-built 2H/1H coil using a 90° excitation pulse and 400 ms repetition time. 3D DMI was recorded at high resolution (2 × 2 × 2 mm) in 37 min or at low resolution (3.7 × 3.7 × 3.7 mm) in 2:24 min. Absolute tissue concentrations were calculated assuming natural deuterated water [HOD] = 13.7 mM. Within 5 min after [2H9]choline infusion, its signal appeared in tumor spectra representing a concentration increase to 0.3–1.2 mM, which then slowly decreased or remained constant over 100 min. In plasma, [2H9]choline disappeared within 15 min post-infusion, implying that its signal arises from tumor tissue and not from blood. After infusing a mixture of [2H9]choline and [6,6′-2H2]glucose, their signals were observed separately in tumor 2H spectra. Over time, the [2H9]choline signal broadened, possibly due to conversion to other choline compounds, [[6,6′-2H2]glucose] declined, [HOD] increased and a lactate signal appeared, reflecting glycolysis. Metabolic maps of 2H compounds, reconstructed from high resolution DMIs, showed their spatial tumor accumulation. As choline infusion and glucose DMI is feasible in patients, their simultaneous detection has clinical potential for tumor characterization.
Collapse
|
13
|
Lesbats C, Katoch N, Minhas AS, Taylor A, Kim HJ, Woo EJ, Poptani H. High-frequency electrical properties tomography at 9.4T as a novel contrast mechanism for brain tumors. Magn Reson Med 2021; 86:382-392. [PMID: 33533114 PMCID: PMC8603929 DOI: 10.1002/mrm.28685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/03/2020] [Accepted: 12/24/2020] [Indexed: 11/11/2022]
Abstract
PURPOSE To establish high-frequency magnetic resonance electrical properties tomography (MREPT) as a novel contrast mechanism for the assessment of glioblastomas using a rat brain tumor model. METHODS Six F98 intracranial tumor bearing rats were imaged longitudinally 8, 11 and 14 days after tumor cell inoculation. Conductivity and mean diffusivity maps were generated using MREPT and Diffusion Tensor Imaging. These maps were co-registered with T2 -weighted images and volumes of interests (VOIs) were segmented from the normal brain, ventricles, edema, viable tumor, tumor rim, and tumor core regions. Longitudinal changes in conductivity and mean diffusivity (MD) values were compared in these regions. A correlation analysis was also performed between conductivity and mean diffusivity values. RESULTS The conductivity of ventricles, edematous area and tumor regions (tumor rim, viable tumor, tumor core) was significantly higher (P < .01) compared to the contralateral cortex. The conductivity of the tumor increased over time while MD from the tumor did not change. A marginal positive correlation was noted between conductivity and MD values for tumor rim and viable tumor, whereas this correlation was negative for the tumor core. CONCLUSION We demonstrate a novel contrast mechanism based on ionic concentration and mobility, which may aid in providing complementary information to water diffusion in probing the microenvironment of brain tumors.
Collapse
Affiliation(s)
- Clémentine Lesbats
- Centre for Preclinical ImagingDepartment of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Nitish Katoch
- Department of Biomedical EngineeringKyung Hee UniversitySeoulSouth Korea
| | - Atul Singh Minhas
- Centre for Preclinical ImagingDepartment of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
- School of EngineeringMacquarie UniversitySydneyNSWAustralia
| | - Arthur Taylor
- Centre for Preclinical ImagingDepartment of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Hyung Joong Kim
- Department of Biomedical EngineeringKyung Hee UniversitySeoulSouth Korea
| | - Eung Je Woo
- Department of Biomedical EngineeringKyung Hee UniversitySeoulSouth Korea
| | - Harish Poptani
- Centre for Preclinical ImagingDepartment of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
14
|
Lacal JC, Zimmerman T, Campos JM. Choline Kinase: An Unexpected Journey for a Precision Medicine Strategy in Human Diseases. Pharmaceutics 2021; 13:788. [PMID: 34070409 PMCID: PMC8226952 DOI: 10.3390/pharmaceutics13060788] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
Choline kinase (ChoK) is a cytosolic enzyme that catalyzes the phosphorylation of choline to form phosphorylcholine (PCho) in the presence of ATP and magnesium. ChoK is required for the synthesis of key membrane phospholipids and is involved in malignant transformation in a large variety of human tumours. Active compounds against ChoK have been identified and proposed as antitumor agents. The ChoK inhibitory and antiproliferative activities of symmetrical bispyridinium and bisquinolinium compounds have been defined using quantitative structure-activity relationships (QSARs) and structural parameters. The design strategy followed in the development of the most active molecules is presented. The selective anticancer activity of these structures is also described. One promising anticancer compound has even entered clinical trials. Recently, ChoKα inhibitors have also been proposed as a novel therapeutic approach against parasites, rheumatoid arthritis, inflammatory processes, and pathogenic bacteria. The evidence for ChoKα as a novel drug target for approaches in precision medicine is discussed.
Collapse
Affiliation(s)
- Juan Carlos Lacal
- Instituto de Investigaciones Biomédicas, CSIC, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz, IDIPAZ, 28046 Madrid, Spain
| | - Tahl Zimmerman
- Food Microbiology and Biotechnology Laboratory, Department of Family and Consumer Sciences, College of Agriculture and Environmental Sciences, North Carolina University, 1601 East Market Street, Greensboro, NC 27411, USA;
| | - Joaquín M. Campos
- Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, c/Campus de Cartuja, s/n, Universidad de Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs. GRANADA), SAS-Universidad de Granada, 18071 Granada, Spain
| |
Collapse
|
15
|
Rubio-Ruiz B, Serrán-Aguilera L, Hurtado-Guerrero R, Conejo-García A. Recent advances in the design of choline kinase α inhibitors and the molecular basis of their inhibition. Med Res Rev 2020; 41:902-927. [PMID: 33103259 DOI: 10.1002/med.21746] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/25/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022]
Abstract
Upregulated choline metabolism, characterized by an increase in phosphocholine (PCho), is a hallmark of oncogenesis and tumor progression. Choline kinase (ChoK), the enzyme responsible for PCho synthesis, has consequently become a promising drug target for cancer therapy and as such a significant number of ChoK inhibitors have been developed over the last few decades. More recently, due to the role of this enzyme in other pathologies, ChoK inhibitors have also been used in new therapeutic approaches against malaria and rheumatoid arthritis. Here, we review research results in the field of ChoKα inhibitors from their synthesis to the molecular basis of their binding mode. Strategies for the development of inhibitors and their selectivity on ChoKα over ChoKβ, the plasticity of the choline-binding site, the discovery of new exploitable binding sites, and the allosteric properties of this enzyme are highlighted. The outcomes summarized in this review will be a useful guide to develop new multifunctional potent drugs for the treatment of various human diseases.
Collapse
Affiliation(s)
- Belén Rubio-Ruiz
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain.,Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, Spain
| | - Lucía Serrán-Aguilera
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Ramón Hurtado-Guerrero
- Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Zaragoza, Spain.,Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.,Laboratorio de Microscopías Avanzada, University of Zaragoza, Zaragoza, Spain.,ARAID Foundation, Zaragoza, Spain
| | - Ana Conejo-García
- Department of Medicinal and Organic Chemistry, Faculty of Pharmacy, University of Granada, Granada, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, Spain
| |
Collapse
|
16
|
Arlauckas SP, Browning EA, Poptani H, Delikatny EJ. Imaging of cancer lipid metabolism in response to therapy. NMR IN BIOMEDICINE 2019; 32:e4070. [PMID: 31107583 DOI: 10.1002/nbm.4070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 06/09/2023]
Abstract
Lipids represent a diverse array of molecules essential to the cell's structure, defense, energy, and communication. Lipid metabolism can often become dysregulated during tumor development. During cancer therapy, targeted inhibition of cell proliferation can likewise cause widespread and drastic changes in lipid composition. Molecular imaging techniques have been developed to monitor altered lipid profiles as a biomarker for cancer diagnosis and treatment response. For decades, MRS has been the dominant non-invasive technique for studying lipid metabolite levels. Recent insights into the oncogenic transformations driving changes in lipid metabolism have revealed new mechanisms and signaling molecules that can be exploited using optical imaging, mass spectrometry imaging, and positron emission tomography. These novel imaging modalities have provided researchers with a diverse toolbox to examine changes in lipids in response to a wide array of anticancer strategies including chemotherapy, radiation therapy, signal transduction inhibitors, gene therapy, immunotherapy, or a combination of these strategies. The understanding of lipid metabolism in response to cancer therapy continues to evolve as each therapeutic method emerges, and this review seeks to summarize the current field and areas of unmet needs.
Collapse
Affiliation(s)
- Sean Philip Arlauckas
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Systems Biology, Mass General Hospital, Boston, MA, USA
| | - Elizabeth Anne Browning
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, Institute of Regenerative Medicine, University of Liverpool, Liverpool, UK
| | - Edward James Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Pillai SR, Damaghi M, Marunaka Y, Spugnini EP, Fais S, Gillies RJ. Causes, consequences, and therapy of tumors acidosis. Cancer Metastasis Rev 2019; 38:205-222. [PMID: 30911978 PMCID: PMC6625890 DOI: 10.1007/s10555-019-09792-7] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
While cancer is commonly described as "a disease of the genes," it is also associated with massive metabolic reprogramming that is now accepted as a disease "Hallmark." This programming is complex and often involves metabolic cooperativity between cancer cells and their surrounding stroma. Indeed, there is emerging clinical evidence that interrupting a cancer's metabolic program can improve patients' outcomes. The most commonly observed and well-studied metabolic adaptation in cancers is the fermentation of glucose to lactic acid, even in the presence of oxygen, also known as "aerobic glycolysis" or the "Warburg Effect." Much has been written about the mechanisms of the Warburg effect, and this remains a topic of great debate. However, herein, we will focus on an important sequela of this metabolic program: the acidification of the tumor microenvironment. Rather than being an epiphenomenon, it is now appreciated that this acidosis is a key player in cancer somatic evolution and progression to malignancy. Adaptation to acidosis induces and selects for malignant behaviors, such as increased invasion and metastasis, chemoresistance, and inhibition of immune surveillance. However, the metabolic reprogramming that occurs during adaptation to acidosis also introduces therapeutic vulnerabilities. Thus, tumor acidosis is a relevant therapeutic target, and we describe herein four approaches to accomplish this: (1) neutralizing acid directly with buffers, (2) targeting metabolic vulnerabilities revealed by acidosis, (3) developing acid-activatable drugs and nanomedicines, and (4) inhibiting metabolic processes responsible for generating acids in the first place.
Collapse
Affiliation(s)
- Smitha R Pillai
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33602, USA
| | - Mehdi Damaghi
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33602, USA
| | - Yoshinori Marunaka
- Research Institute for Clinical Physiology, Kyoto, 604-8472, Japan
- Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, 525-8577, Japan
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | | | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr., Tampa, FL, 33602, USA.
| |
Collapse
|
18
|
Lead optimization-hit expansion of new asymmetrical pyridinium/quinolinium compounds as choline kinase α1 inhibitors. Future Med Chem 2018; 10:1769-1786. [PMID: 30043647 DOI: 10.4155/fmc-2018-0059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Choline kinase α inhibitors represent one of the newest classes of cytotoxic drugs for cancer treatment, since aberrant choline metabolism is a characteristic shared by many human cancers. RESULTS Here, we present a new class of asymmetrical pyridinium/quinolinium derivatives developed and designed based on drug optimization. CONCLUSION Among all compounds described here, compound 8, bearing a 7-chloro-4N-methyl-p-chloroaniline quinolinium moiety, exhibited the greatest inhibitory activity at the enzyme (IC50 = 0.29 μM) and antiproliferative activity in cellular assays (GI50 = 0.29-0.92 μM). Specifically, compound 8 strongly induces a cell-cycle arrest in G1 phase, but it does not significantly induce apoptosis while causing senescence in the MDA-MB-231 cell line.
Collapse
|
19
|
Liebov B, Arroyo AD, Rubtsova NI, Osharovich SA, Delikatny EJ, Popov AV. Nonprotecting Group Synthesis of a Phospholipase C Activatable Probe with an Azo-Free Quencher. ACS OMEGA 2018; 3:6867-6873. [PMID: 29978148 PMCID: PMC6026834 DOI: 10.1021/acsomega.8b00635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/18/2018] [Indexed: 06/08/2023]
Abstract
The near-infrared fluorescent activatable smart probe Pyro-phosphatidylethanolamine (PtdEtn)-QSY was synthesized and observed to selectively fluoresce in the presence of phosphatidylcholine-specific phospholipase C (PC-PLC). PC-PLC is an important biological target as it is known to be upregulated in a variety of cancers, including triple negative breast cancer. Pyro-PtdEtn-QSY features a QSY21 quenching moiety instead of the Black Hole Quencher-3 (BHQ-3) used previously because the latter contains an azo bond, which could lead to biological instability.
Collapse
|
20
|
Hu L, Wang RY, Cai J, Feng D, Yang GZ, Xu QG, Zhai YX, Zhang Y, Zhou WP, Cai QP. Overexpression of CHKA contributes to tumor progression and metastasis and predicts poor prognosis in colorectal carcinoma. Oncotarget 2018; 7:66660-66678. [PMID: 27556502 PMCID: PMC5341828 DOI: 10.18632/oncotarget.11433] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/13/2016] [Indexed: 12/13/2022] Open
Abstract
Aberrant expression of choline kinase alpha (CHKA) has been reported in a variety of human malignancies including colorectal carcinoma (CRC). However, the role of CHKA in the progression and prognosis of CRC remains unknown. In this study, we found that CHKA was frequently upregulated in CRC clinical samples and CRC-derived cell lines and was significantly correlated with lymph node metastasis (p = 0.028), TNM stage (p = 0.009), disease recurrence (p = 0.004) and death (p < 0.001). Survival analyses indicated that patients with higher CHKA expression had a significantly shorter disease-free survival (DFS) and disease-specific survival (DSS) than those with lower CHKA expression. Multivariate analyses confirmed that increased CHKA expression was an independent unfavorable prognostic factor for CRC patients. In addition, combination of CHKA with TNM stage was a more powerful predictor of poor prognosis than either parameter alone. Functional study demonstrated that knockdown of CHKA expression profoundly suppressed the growth and metastasis of CRC cells both in vitro and in vivo. Mechanistic investigation revealed that EGFR/PI3K/AKT pathway was essential for mediating CHKA function. In conclusion, our results provide the first evidence that CHKA contributes to tumor progression and metastasis and may serve as a novel prognostic biomarker and potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Liang Hu
- Anal-Colorectal Surgery Institute, 150th Hospital of PLA, Luoyang, China.,Department of Gastrointestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ruo-Yu Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jian Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan Feng
- Department of Oncology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guang-Zhen Yang
- Department of Clinical Laboratory, 150th Hospital of PLA, Luoyang, China
| | - Qing-Guo Xu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yan-Xia Zhai
- Anal-Colorectal Surgery Institute, 150th Hospital of PLA, Luoyang, China
| | - Yu Zhang
- Anal-Colorectal Surgery Institute, 150th Hospital of PLA, Luoyang, China
| | - Wei-Ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Qing-Ping Cai
- Department of Gastrointestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
21
|
Kall SL, Delikatny EJ, Lavie A. Identification of a Unique Inhibitor-Binding Site on Choline Kinase α. Biochemistry 2018; 57:1316-1325. [PMID: 29389115 DOI: 10.1021/acs.biochem.7b01257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Choline kinase α (ChoKα) is an enzyme that is upregulated in many types of cancer and has been shown to be tumorigenic. As such, it makes a promising target for inhibiting tumor growth. Though there have been several inhibitors synthesized for ChoKα, not all of them demonstrate the same efficacy in vivo, though the reasons behind this difference in potency are not clear. One particular inhibitor, designated TCD-717, has recently completed phase I clinical trials. Cell culture and in vitro studies support the powerful inhibitory effect TCD-717 has on ChoKα, but an examination of the inhibitor's interaction with the ChoKα enzyme has been missing prior to this work. Here we detail the 2.35 Å structure of ChoKα in complex with TCD-717. Examination of this structure in conjunction with kinetic assays reveals that TCD-717 does not bind directly in the choline pocket as do previously characterized ChoKα inhibitors, but rather in a proximal but novel location near the surface of the enzyme. The unique binding site identified for TCD-717 lends insight for the future design of more potent in vivo inhibitors for ChoKα.
Collapse
Affiliation(s)
- Stefanie L Kall
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, Illinois 60607, United States
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania 19083, United States
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, Illinois 60607, United States.,The Jesse Brown VA Medical Center , Chicago, Illinois 60612, United States
| |
Collapse
|
22
|
Arlauckas SP, Kumar M, Popov AV, Poptani H, Delikatny EJ. Near infrared fluorescent imaging of choline kinase alpha expression and inhibition in breast tumors. Oncotarget 2017; 8:16518-16530. [PMID: 28157707 PMCID: PMC5369982 DOI: 10.18632/oncotarget.14965] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Choline kinase alpha (ChoKα) overexpression is associated with an aggressive tumor phenotype. ChoKα inhibitors induce apoptosis in tumors, however validation of their specificity is difficult in vivo. We report the use of optical imaging to assess ChoKα status in cells and in vivo using JAS239, a carbocyanine-based ChoKα inhibitor with inherent near infrared fluorescence. JAS239 attenuated choline phosphorylation and viability in a panel of human breast cancer cell lines. Antibody blockade prevented cellular retention of JAS239 indicating direct interaction with ChoKα independent of the choline transporters and catabolic choline pathways. In mice bearing orthotopic MCF7 breast xenografts, optical imaging with JAS239 distinguished tumors overexpressing ChoKα from their empty vector counterparts and delineated tumor margins. Pharmacological inhibition of ChoK by the established inhibitor MN58b led to a growth inhibition in 4175-Luc+ tumors that was accompanied by concomitant reduction in JAS239 uptake and decreased total choline metabolite levels as measured using magnetic resonance spectroscopy. At higher therapeutic doses, JAS239 was as effective as MN58b at arresting tumor growth and inducing apoptosis in MDA-MB-231 tumors, significantly reducing tumor choline below baseline levels without observable systemic toxicity. These data introduce a new method to monitor therapeutically effective inhibitors of choline metabolism in breast cancer using a small molecule companion diagnostic.
Collapse
Affiliation(s)
- Sean P Arlauckas
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Manoj Kumar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anatoliy V Popov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Harish Poptani
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cellular and Molecular Physiology, Institute of Regenerative Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
23
|
Podo F, Paris L, Cecchetti S, Spadaro F, Abalsamo L, Ramoni C, Ricci A, Pisanu ME, Sardanelli F, Canese R, Iorio E. Activation of Phosphatidylcholine-Specific Phospholipase C in Breast and Ovarian Cancer: Impact on MRS-Detected Choline Metabolic Profile and Perspectives for Targeted Therapy. Front Oncol 2016; 6:171. [PMID: 27532027 PMCID: PMC4969288 DOI: 10.3389/fonc.2016.00171] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/05/2016] [Indexed: 12/12/2022] Open
Abstract
Elucidation of molecular mechanisms underlying the aberrant phosphatidylcholine cycle in cancer cells plays in favor of the use of metabolic imaging in oncology and opens the way for designing new targeted therapies. The anomalous choline metabolic profile detected in cancer by magnetic resonance spectroscopy and spectroscopic imaging provides molecular signatures of tumor progression and response to therapy. The increased level of intracellular phosphocholine (PCho) typically detected in cancer cells is mainly attributed to upregulation of choline kinase, responsible for choline phosphorylation in the biosynthetic Kennedy pathway, but can also be partly produced by activation of phosphatidylcholine-specific phospholipase C (PC-PLC). This hydrolytic enzyme, known for implications in bacterial infection and in plant survival to hostile environmental conditions, is reported to be activated in mitogen- and oncogene-induced phosphatidylcholine cycles in mammalian cells, with effects on cell signaling, cell cycle regulation, and cell proliferation. Recent investigations showed that PC-PLC activation could account for 20–50% of the intracellular PCho production in ovarian and breast cancer cells of different subtypes. Enzyme activation was associated with PC-PLC protein overexpression and subcellular redistribution in these cancer cells compared with non-tumoral counterparts. Moreover, PC-PLC coimmunoprecipitated with the human epidermal growth factor receptor-2 (HER2) and EGFR in HER2-overexpressing breast and ovarian cancer cells, while pharmacological PC-PLC inhibition resulted into long-lasting HER2 downregulation, retarded receptor re-expression on plasma membrane and antiproliferative effects. This body of evidence points to PC-PLC as a potential target for newly designed therapies, whose effects can be preclinically and clinically monitored by metabolic imaging methods.
Collapse
Affiliation(s)
- Franca Podo
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Luisa Paris
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Serena Cecchetti
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Francesca Spadaro
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Laura Abalsamo
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Carlo Ramoni
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Alessandro Ricci
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Maria Elena Pisanu
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Research Hospital Policlinico San Donato , Milan , Italy
| | - Rossella Canese
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Egidio Iorio
- Molecular and Cellular Imaging Unit, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
24
|
Feng Y, Clayton EH, Okamoto RJ, Engelbach J, Bayly PV, Garbow JR. A longitudinal magnetic resonance elastography study of murine brain tumors following radiation therapy. Phys Med Biol 2016; 61:6121-31. [PMID: 27461395 DOI: 10.1088/0031-9155/61/16/6121] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
An accurate and noninvasive method for assessing treatment response following radiotherapy is needed for both treatment monitoring and planning. Measurement of solid tumor volume alone is not sufficient for reliable early detection of therapeutic response, since changes in physiological and/or biomechanical properties can precede tumor volume change following therapy. In this study, we use magnetic resonance elastography to evaluate the treatment effect after radiotherapy in a murine brain tumor model. Shear modulus was calculated and compared between the delineated tumor region of interest (ROI) and its contralateral, mirrored counterpart. We also compared the shear modulus from both the irradiated and non-irradiated tumor and mirror ROIs longitudinally, sampling four time points spanning 9-19 d post tumor implant. Results showed that the tumor ROI had a lower shear modulus than that of the mirror ROI, independent of radiation. The shear modulus of the tumor ROI decreased over time for both the treated and untreated groups. By contrast, the shear modulus of the mirror ROI appeared to be relatively constant for the treated group, while an increasing trend was observed for the untreated group. The results provide insights into the tumor properties after radiation treatment and demonstrate the potential of using the mechanical properties of the tumor as a biomarker. In future studies, more closely spaced time points will be employed for detailed analysis of the radiation effect.
Collapse
Affiliation(s)
- Y Feng
- School of Mechanical and Electronic Engineering, Soochow University, Suzhou, Jiangsu, People's Republic of China. Robotics and Microsystems Center, Soochow University, Suzhou, Jiangsu, People's Republic of China. School of Computer Science and Engineering, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | | | | | | | | | | |
Collapse
|
25
|
Arlauckas SP, Popov AV, Delikatny EJ. Choline kinase alpha-Putting the ChoK-hold on tumor metabolism. Prog Lipid Res 2016; 63:28-40. [PMID: 27073147 PMCID: PMC5360181 DOI: 10.1016/j.plipres.2016.03.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/14/2016] [Accepted: 03/26/2016] [Indexed: 12/24/2022]
Abstract
It is well established that lipid metabolism is drastically altered during tumor development and response to therapy. Choline kinase alpha (ChoKα) is a key mediator of these changes, as it represents the first committed step in the Kennedy pathway of phosphatidylcholine biosynthesis and ChoKα expression is upregulated in many human cancers. ChoKα activity is associated with drug resistant, metastatic, and malignant phenotypes, and represents a robust biomarker and therapeutic target in cancer. Effective ChoKα inhibitors have been developed and have recently entered clinical trials. ChoKα's clinical relevance was, until recently, attributed solely to its production of second messenger intermediates of phospholipid synthesis. The recent discovery of a non-catalytic scaffolding function of ChoKα may link growth receptor signaling to lipid biogenesis and requires a reinterpretation of the design and validation of ChoKα inhibitors. Advances in positron emission tomography, magnetic resonance spectroscopy, and optical imaging methods now allow for a comprehensive understanding of ChoKα expression and activity in vivo. We will review the current understanding of ChoKα metabolism, its role in tumor biology and the development and validation of targeted therapies and companion diagnostics for this important regulatory enzyme. This comes at a critical time as ChoKα-targeting programs receive more clinical interest.
Collapse
Affiliation(s)
- Sean P Arlauckas
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anatoliy V Popov
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E James Delikatny
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Bagnoli M, Granata A, Nicoletti R, Krishnamachary B, Bhujwalla ZM, Canese R, Podo F, Canevari S, Iorio E, Mezzanzanica D. Choline Metabolism Alteration: A Focus on Ovarian Cancer. Front Oncol 2016; 6:153. [PMID: 27446799 PMCID: PMC4916225 DOI: 10.3389/fonc.2016.00153] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/07/2016] [Indexed: 12/31/2022] Open
Abstract
Compared with normal differentiated cells, cancer cells require a metabolic reprograming to support their high proliferation rates and survival. Aberrant choline metabolism is a fairly new metabolic hallmark reflecting the complex reciprocal interactions between oncogenic signaling and cellular metabolism. Alterations of the involved metabolic network may be sustained by changes in activity of several choline transporters as well as of enzymes such as choline kinase-alpha (ChoK-α) and phosphatidylcholine-specific phospholipases C and D. Of note, the net outcome of these enzymatic alterations is an increase of phosphocholine and total choline-containing compounds, a "cholinic phenotype" that can be monitored in cancer by magnetic resonance spectroscopy. This review will highlight the molecular basis for targeting this pathway in epithelial ovarian cancer (EOC), a highly heterogeneous and lethal malignancy characterized by late diagnosis, frequent relapse, and development of chemoresistance. Modulation of ChoK-α expression impairs only EOC but not normal ovarian cells, thus supporting the hypothesis that "cholinic phenotype" is a peculiar feature of transformed cells and indicating ChoK-α targeting as a novel approach to improve efficacy of standard EOC chemotherapeutic treatments.
Collapse
Affiliation(s)
- Marina Bagnoli
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | - Anna Granata
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | - Roberta Nicoletti
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| | - Balaji Krishnamachary
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, In Vivo Cellular and Molecular Imaging Center, The Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Zaver M Bhujwalla
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, In Vivo Cellular and Molecular Imaging Center, The Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Rossella Canese
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Franca Podo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Silvana Canevari
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Functional Genomics and Informatics, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Egidio Iorio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità , Rome , Italy
| | - Delia Mezzanzanica
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori , Milan , Italy
| |
Collapse
|
27
|
Mignion L, Danhier P, Magat J, Porporato PE, Masquelier J, Gregoire V, Muccioli GG, Sonveaux P, Gallez B, Jordan BF. Non-invasive in vivo imaging of early metabolic tumor response to therapies targeting choline metabolism. Int J Cancer 2015; 138:2043-9. [PMID: 26595604 DOI: 10.1002/ijc.29932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 01/17/2023]
Abstract
The cholinic phenotype, characterized by elevated phosphocholine and a high production of total-choline (tCho)-containing metabolites, is a metabolic hallmark of cancer. It can be exploited for targeted therapy. Non-invasive imaging biomarkers are required to evaluate an individual's response to targeted anticancer agents that usually do not rapidly cause tumor shrinkage. Because metabolic changes can manifest at earlier stages of therapy than changes in tumor size, the aim of the current study was to evaluate (1)H-MRS and diffusion-weighted MRI (DW-MRI) as markers of tumor response to the modulation of the choline pathway in mammary tumor xenografts. Inhibition of choline kinase activity was achieved with the direct pharmacological inhibitor H-89, indirect inhibitor sorafenib and down-regulation of choline-kinase α (ChKA) expression using specific short-hairpin RNA (shRNA). While all three strategies significantly decreased tCho tumor content in vivo, only sorafenib and anti-ChKA shRNA significantly repressed tumor growth. The increase of apparent-diffusion-coefficient of water (ADCw) measured by DW-MRI, was predictive of the induced necrosis and inhibition of the tumor growth in sorafenib treated mice, while the absence of change in ADC values in H89 treated mice predicted the absence of effect in terms of tumor necrosis and tumor growth. In conclusion, (1)H-choline spectroscopy can be useful as a pharmacodynamic biomarker for choline targeted agents, while DW-MRI can be used as an early marker of effective tumor response to choline targeted therapies. DW-MRI combined to choline spectroscopy may provide a useful non-invasive marker for the early clinical assessment of tumor response to therapies targeting choline signaling.
Collapse
Affiliation(s)
- Lionel Mignion
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Pierre Danhier
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Julie Magat
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Paolo E Porporato
- Pole of Pharmacology, Institut De Recherche Expérimentale Et Clinique (IREC), Université Catholique De Louvain (UCL), Avenue Mounier, 52 Box B1.53.09, Brussels, Belgium
| | - Julien Masquelier
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 72 Box B1.72.01, Brussels, Belgium
| | - Vincent Gregoire
- Center for Molecular Imaging, Radiotherapy and Oncology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Hippocrate, 55 Box B1.55.02, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 72 Box B1.72.01, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology, Institut De Recherche Expérimentale Et Clinique (IREC), Université Catholique De Louvain (UCL), Avenue Mounier, 52 Box B1.53.09, Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique De Louvain (UCL), Avenue Mounier, 73 Box B1.73.08, Brussels, Belgium
| |
Collapse
|