1
|
He R, Song Z, Bai Y, He S, Huang J, Wang Y, Zhou F, Huang W, Guo J, Wang Z, Tu ZC, Ren X, Zhang Z, Xu J, Ding K. Discovery of AXL Degraders with Improved Potencies in Triple-Negative Breast Cancer (TNBC) Cells. J Med Chem 2023; 66:1873-1891. [PMID: 36695404 DOI: 10.1021/acs.jmedchem.2c01682] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
AXL kinase is heavily involved in tumorigenesis, metastasis, and drug resistance of many cancers, and several AXL inhibitors are in clinical investigations. Recent studies demonstrated that the N-terminal distal region of AXL plays more important roles in cell invasiveness than its C-terminal kinase domain. Therefore, degradation of AXL may present a novel superior therapeutic approach than the kinase inhibitor therapy. Herein, we report the discovery of a series of new AXL PROTAC degraders. One representative compound 6n potently depletes AXL with a DC50 value of 5 nM in MDA-MB-231 TNBC cells. It also demonstrates significantly improved potencies against the AXL signaling activation, cell proliferation, migration and invasion of TNBC cells comparing with the corresponding kinase inhibitor. Moreover, the compound exhibits promising therapeutic potential both in patient-derived organoids and a xenograft mouse model of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Rui He
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Zhiqiang Song
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Yu Bai
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Sheng He
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Jing Huang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Yongxing Wang
- Livzon Research Institute, Livzon Pharmaceutical Group Inc., 38 Chuangye North Road, Jinwan District, Zhuhai 519000, China
| | - Fengtao Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Weixue Huang
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Jing Guo
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Zhen Wang
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Zheng-Chao Tu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Xiaomei Ren
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China
| | - Jian Xu
- Livzon Research Institute, Livzon Pharmaceutical Group Inc., 38 Chuangye North Road, Jinwan District, Zhuhai 519000, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 855 Xingye Avenue, Panyu District, Guangzhou 510632, China.,State Key Laboratory of Bioorganic & Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China.,The First Affiliated Hospital (Huaqiao Hospital), Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
2
|
Malvankar C, Kumar D. AXL kinase inhibitors- A prospective model for medicinal chemistry strategies in anticancer drug discovery. Biochim Biophys Acta Rev Cancer 2022; 1877:188786. [PMID: 36058379 DOI: 10.1016/j.bbcan.2022.188786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/19/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022]
Abstract
Deviant expressions of the tyrosine kinase AXL receptor are strongly correlated with a plethora of malignancies. Henceforth, the topic of targeting AXL is beginning to gain prominence due to mounting evidence of the protein's substantial connection to poor prognosis and treatment resistance. This year marked a milestone in clinical testing for AXL as an anti-carcinogenic target, with the start of the first AXL-branded inhibitor study. It is critical to emphasize that AXL is a primary and secondary target in various kinase inhibitors that have been approved or are on the verge of being approved while interpreting the present benefits and future potential effects of AXL suppression in the clinical setting. Several research arenas across the globe resolutely affirm the crucial significance of AXL receptors in the case study of several pathophysiologies including AML, prostate cancer, and breast cancer. This review endeavors to delve deeply into the biological, chemical, and structural features of AXL kinase; primary AXL inhibitors that target the enzyme (either purposefully or unintentionally); and the prospects and barriers for turning AXL inhibitors into a feasible treatment alternative. Furthermore, we analyse the co-crystal structure of AXL, which remains extensively unexplored, as well as the mutations of AXL that may be valuable in the development of novel inhibitors in the upcoming future and take a comprehensive look at the medicinal chemistry of AXL inhibitors of recent years.
Collapse
Affiliation(s)
- Chinmay Malvankar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra 411038, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra 411038, India; Department of Entomology, University of California, Davis, One Shields Ave, Davis, CA 95616, USA; UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
3
|
Bhojwani HR, Joshi UJ. Homology Modelling, Docking-based Virtual Screening, ADME Properties, and Molecular Dynamics Simulation for Identification of Probable Type II Inhibitors of AXL Kinase. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666211004102043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
AXL kinase is an important member of the TAM family for kinases which is
involved in most cancers. Considering its role in different cancers due to its pro-tumorigenic effects and its
involvement in the resistance, it has gained importance recently. Majority of research carried out is on Type I
inhibitors and limited studies have been carried out for Type II inhibitors. Taking this into consideration, we
have attempted to build Homology models to identify the Type II inhibitors for the AXL kinase.
Methods:
Homology Models for DFG-out C-helix-in/out state were developed using SWISS Model,
PRIMO, and Prime. These models were validated by different methods and further evaluated for stability
by molecular dynamics simulation using Desmond software. Selected models PED1-EB and PEDI1-EB
were used for the docking-based virtual screening of four compound libraries using Glide software. The
hits identified were subjected to interaction analysis and shortlisted compounds were subjected to Prime
MM-GBSA studies for energy calculation. These compounds were also docked in the DFG-in state to
check for binding and elimination of any compounds that may not be Type II inhibitors. The Prime energies
were calculated for these complexes as well and some compounds were eliminated. ADMET studies
were carried out using Qikprop. Some selected compounds were subjected to molecular dynamics simulation
using Desmond for evaluating the stability of the complexes.
Results:
Out of 78 models inclusive of both DFG-out C-helix-in and DFG-out C-helix-out, 5 models were
identified after different types of evaluation as well as validation studies. 1 model representing each type
(PED1-EB and PEDI1-EB) was selected for the screening studies. The screening studies resulted in the
identification of 29 compounds from the screen on PED1-EB and 10 compounds from the screen on
PEDI1-EB. Hydrogen bonding interactions with Pro621, Met623, and Asp690 were observed for these
compounds primarily. In some compounds, hydrogen bonding with Leu542, Glu544, Lys567, and
Asn677 as well as pi-pi stacking interactions with either Phe622 or Phe691 were also seen. 4 compounds
identified from PED1-EB screen were subjected to molecular dynamics simulation and their interactions
were found to be consistent during the simulation. 2 compounds identified from PEDI1-EB screen were
also subjected to the simulation studies, however, their interactions with Asp690 were not observed for a
significant time and in both cases differed from the docked pose.
Conclusion:
Multiple models of DFG-out conformations of AXL kinase were built, validated and used
for virtual screening. Different compounds were identified in the virtual screening, which may possibly
act as Type II inhibitors for AXL kinase. Some more experimental studies can be done to validate these
findings in future. This study will play a guiding role in the further development of the newer Type II
inhibitors of the AXL kinase for the probable treatment of cancer.
Collapse
Affiliation(s)
- Heena R. Bhojwani
- Department of Pharmaceutical Chemistry, Principal K.M. Kundnani College of Pharmacy, Colaba, Cuffe Parade,
Mumbai 400005, India
| | - Urmila J. Joshi
- Department of Pharmaceutical Chemistry, Principal K.M. Kundnani College of Pharmacy, Colaba, Cuffe Parade,
Mumbai 400005, India
| |
Collapse
|
4
|
Zaman A, Bivona TG. Targeting AXL in NSCLC. LUNG CANCER (AUCKLAND, N.Z.) 2021; 12:67-79. [PMID: 34408519 PMCID: PMC8364399 DOI: 10.2147/lctt.s305484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
State-of-the-art cancer precision medicine approaches involve targeted inactivation of chemically and immunologically addressable vulnerabilities that often yield impressive initial anti-tumor responses in patients. Nonetheless, these responses are overshadowed by therapy resistance that follows. AXL, a receptor tyrosine kinase with bona fide oncogenic capacity, has been associated with the emergence of resistance in an array of cancers with varying pathophysiology and cellular origins, including in non-small-cell lung cancers (NSCLCs). Here in this review, we summarize AXL biology during normal homeostasis, oncogenic development and therapy resistance with a focus on NSCLC. In the context of NSCLC therapy resistance, we delineate AXL's role in mediating resistance to tyrosine kinase inhibitors (TKIs) deployed against epidermal growth factor receptor (EGFR) as well as other notable oncogenes and to chemotherapeutics. We also discuss the current understanding of AXL's role in mediating cell-biological variables that function as important modifiers of therapy resistance such as epithelial to mesenchymal transition (EMT), the tumor microenvironment and tumor heterogeneity. We also catalog and discuss a set of effective pharmacologic tools that are emerging to strategically perturb AXL mediated resistance programs in NSCLC. Finally, we enumerate ongoing and future exciting precision medicine approaches targeting AXL as well as challenges in this regard. We highlight that a holistic understanding of AXL biology in NSCLC may allow us to predict and improve targeted therapeutic strategies, such as through polytherapy approaches, potentially against a broad spectrum of NSCLC sub-types to forestall tumor evolution and drug resistance.
Collapse
Affiliation(s)
- Aubhishek Zaman
- Department of Medicine, University of California, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
5
|
Mansi I, Al-Sha'er MA, Mhaidat N, Taha M. Ligand Based Pharmacophore Modeling Followed by Biological Screening Lead to Discovery of Novel PDK1 Inhibitors as Anticancer Agents. Anticancer Agents Med Chem 2020; 20:476-485. [PMID: 31889497 DOI: 10.2174/1871520620666191224110600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/21/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Phosphoinositide-Dependent Kinase-1 (PDK1) is a serine/threonine kinase, which belongs to AGC kinase family required by cancer cells. METHODS Pharmacophoric space of 86 PDK1 inhibitors using six diverse sets of inhibitors was explored to identify high-quality pharmacophores. The best combination of pharmacophoric models and physicochemical descriptors was selected by genetic algorithm-based QSAR analysis that can elucidate the variation of bioactivity within the training inhibitors. Two successful orthogonal pharmacophores emerged in the optimum QSAR equation (r2 69 = 0.90, r2 LOO= 0.86, F= 51.92, r2 PRESS against 17 test inhibitors = 0.79). Receiver Operating Characteristic (ROC) curve analyses were used to estimate the QSAR-selected pharmacophores. RESULTS 5 out of 11 compounds tested had shown potential intracellular PDK1 inhibition with the highest inhibition percent for compounds 92 and 93 as follows; 90 and 92% PDK1 inhibition, respectively. CONCLUSION PDK1 inhibitors are potential anticancer agents that can be discovered by combination method of ligand based design with QSAR and ROC analysis.
Collapse
Affiliation(s)
- Iman Mansi
- Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. Box 330127 Zarqa, 13133, Jordan
| | | | - Nizar Mhaidat
- Clinical Pharmacy Department, Faculty of Pharmacy, Jordan University of Science & Technology, Irbid, Jordan
| | - Mutasem Taha
- Drug Design Center, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| |
Collapse
|
6
|
Ghrifi F, Allam L, Wiame L, Ibrahimi A. Curcumin-Synthetic Analogs Library Screening by Docking and Quantitative Structure-Activity Relationship Studies for AXL Tyrosine Kinase Inhibition in Cancers. J Comput Biol 2019; 26:1156-1167. [PMID: 31009237 PMCID: PMC6786334 DOI: 10.1089/cmb.2019.0052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AXL is an important drug target for cancers. Two-dimensional quantitative structure-activity relationship (2D-QSAR) tests were performed to elucidate a relationship between molecular structures and the activity of a series of 400 curcumin derivatives subjected to AXL kinase by ATP competition in the catalytic site. The partial least square regression method implanted in molecular operating environment software was applied to develop QSAR models, which were further validated for statistical significance by internal and external validation. The best model has proven to be statistically robust with a good predictive correlation of
R
2
= 0.996 and a significant cross-validation correlation coefficient of
q
2
= 0.707. Docking analysis reveled that three curcumin derivatives have the best affinity for AXL and formed a hydrogen bond with the important amino acid residues in the binding pocket. As treated in this article, the docking studies and 2D-QSAR approach will pave the way for the development of new drugs while highlighting curcumin and its derivatives.
Collapse
Affiliation(s)
- Fatima Ghrifi
- The Biotechnology Lab (MedBiotech), BioInova Research center, Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco.,Address correspondence to: Fatima Ghrifi, PhD Student, The Biotechnology Lab (MedBiotech), BioInova Research center, Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Avenue Mr belarbi Alaoui, Suissi-Rabat, BP6203 Rabat Institutes, Rabat 10000, Morocco
| | - Loubna Allam
- The Biotechnology Lab (MedBiotech), BioInova Research center, Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco
| | - Lakhlili Wiame
- The Biotechnology Lab (MedBiotech), BioInova Research center, Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco
| | - Azeddine Ibrahimi
- The Biotechnology Lab (MedBiotech), BioInova Research center, Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
7
|
Shelby T, Banerjee T, Zegar I, Santra S. Highly Sensitive, Engineered Magnetic Nanosensors to Investigate the Ambiguous Activity of Zika Virus and Binding Receptors. Sci Rep 2017; 7:7377. [PMID: 28785095 PMCID: PMC5547150 DOI: 10.1038/s41598-017-07620-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of this research is twofold: 1) to shed light on zika's binding and entry mechanism while 2) demonstrating the effectiveness of our magnetic relaxation platform to achieve this goal. Magnetic relaxation-sensitive nanoparticles (MRNPs) are used in a novel fashion to analyze binding interactions between the zika envelope protein (ZENV) and proposed host cell receptors: AXL, HSP70, and TIM-1. Computational analysis is also utilized to examine these binding interactions for the first time. In addition, the role of crizotinib as a potential binding inhibitor is demonstrated and the possibility of ligand-independent phosphatidylserine-mediated binding is explored. Our findings suggest that while the extracellular domain of AXL has the highest affinity for ZENV; HSP70, TIM-1, and phosphatidylserine might also play active roles in zika tropism, which offers a potential explanation for the variety of zika-associated symptoms. This is, to our knowledge, the first time that MRNPs have been used to examine and quantify host-zika interactions. Our magnetic relaxation platform allows for timely and sensitive analysis of these intricate binding relationships, and it is easily customizable for further examination of additional host-pathogen interactions.
Collapse
Affiliation(s)
- Tyler Shelby
- Department of Chemistry, Pittsburg State University, 1701 S. Broadway Street, Pittsburg, KS, 66762, USA
| | - Tuhina Banerjee
- Department of Chemistry, Pittsburg State University, 1701 S. Broadway Street, Pittsburg, KS, 66762, USA
| | - Irene Zegar
- Department of Chemistry, Pittsburg State University, 1701 S. Broadway Street, Pittsburg, KS, 66762, USA
| | - Santimukul Santra
- Department of Chemistry, Pittsburg State University, 1701 S. Broadway Street, Pittsburg, KS, 66762, USA.
| |
Collapse
|
8
|
Myers SH, Brunton VG, Unciti-Broceta A. AXL Inhibitors in Cancer: A Medicinal Chemistry Perspective. J Med Chem 2015; 59:3593-608. [DOI: 10.1021/acs.jmedchem.5b01273] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Samuel H. Myers
- Edinburgh Cancer Research
UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K
| | - Valerie G. Brunton
- Edinburgh Cancer Research
UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research
UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, U.K
| |
Collapse
|
9
|
Baladi T, Abet V, Piguel S. State-of-the-art of small molecule inhibitors of the TAM family: the point of view of the chemist. Eur J Med Chem 2015; 105:220-37. [PMID: 26498569 DOI: 10.1016/j.ejmech.2015.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/01/2015] [Accepted: 10/03/2015] [Indexed: 01/04/2023]
Abstract
The TAM family of tyrosine kinases receptors (Tyro3, Axl and Mer) is implicated in cancer development, autoimmune reactions and viral infection and is therefore emerging as an effective and attractive therapeutic target. To date, only a few small molecules have been intentionally designed to block the TAM kinases, while most of the inhibitors were developed for blocking different protein kinases and then identified through selectivity profile studies. This minireview will examine in terms of chemical structure the different compounds able to act on either one, two or three TAM kinases with details about structure-activity relationships, drug-metabolism and pharmacokinetics properties where they exist.
Collapse
Affiliation(s)
- Tom Baladi
- Institut Curie/UMR9187-U1196, 91405 Orsay cedex, France; Univ Paris Sud, 91405 Orsay cedex, France
| | | | - Sandrine Piguel
- Institut Curie/UMR9187-U1196, 91405 Orsay cedex, France; Univ Paris Sud, 91405 Orsay cedex, France.
| |
Collapse
|