1
|
Ryzhkov FV, Ryzhkova YE, Elinson MN. Machine learning: Python tools for studying biomolecules and drug design. Mol Divers 2025:10.1007/s11030-025-11199-2. [PMID: 40301135 DOI: 10.1007/s11030-025-11199-2] [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: 03/06/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
The increasing adoption of computational methods and artificial intelligence in scientific research has led to a growing interest in versatile tools like Python. In the fields of medical chemistry, biochemistry, and bioinformatics, Python has emerged as a key language for tackling complex challenges. It is used to solve various tasks, such as drug discovery, high-throughput and virtual screening, protein and genome analysis, and predicting drug efficacy. This review presents a list of tools for these tasks, including scripts, libraries, and ready-made programs, and serves as a starting point for scientists wishing to apply automation or optimization to routine tasks in medical chemistry and bioinformatics.
Collapse
Affiliation(s)
- Fedor V Ryzhkov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia.
| | - Yuliya E Ryzhkova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia
| | - Michail N Elinson
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia
| |
Collapse
|
2
|
Ramos MC, Collison CJ, White AD. A review of large language models and autonomous agents in chemistry. Chem Sci 2025; 16:2514-2572. [PMID: 39829984 PMCID: PMC11739813 DOI: 10.1039/d4sc03921a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Large language models (LLMs) have emerged as powerful tools in chemistry, significantly impacting molecule design, property prediction, and synthesis optimization. This review highlights LLM capabilities in these domains and their potential to accelerate scientific discovery through automation. We also review LLM-based autonomous agents: LLMs with a broader set of tools to interact with their surrounding environment. These agents perform diverse tasks such as paper scraping, interfacing with automated laboratories, and synthesis planning. As agents are an emerging topic, we extend the scope of our review of agents beyond chemistry and discuss across any scientific domains. This review covers the recent history, current capabilities, and design of LLMs and autonomous agents, addressing specific challenges, opportunities, and future directions in chemistry. Key challenges include data quality and integration, model interpretability, and the need for standard benchmarks, while future directions point towards more sophisticated multi-modal agents and enhanced collaboration between agents and experimental methods. Due to the quick pace of this field, a repository has been built to keep track of the latest studies: https://github.com/ur-whitelab/LLMs-in-science.
Collapse
Affiliation(s)
- Mayk Caldas Ramos
- FutureHouse Inc. San Francisco CA USA
- Department of Chemical Engineering, University of Rochester Rochester NY USA
| | - Christopher J Collison
- School of Chemistry and Materials Science, Rochester Institute of Technology Rochester NY USA
| | - Andrew D White
- FutureHouse Inc. San Francisco CA USA
- Department of Chemical Engineering, University of Rochester Rochester NY USA
| |
Collapse
|
3
|
Singh A, Tanwar M, Singh TP, Sharma S, Sharma P. An escape from ESKAPE pathogens: A comprehensive review on current and emerging therapeutics against antibiotic resistance. Int J Biol Macromol 2024; 279:135253. [PMID: 39244118 DOI: 10.1016/j.ijbiomac.2024.135253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
The rise of antimicrobial resistance has positioned ESKAPE pathogens as a serious global health threat, primarily due to the limitations and frequent failures of current treatment options. This growing risk has spurred the scientific community to seek innovative antibiotic therapies and improved oversight strategies. This review aims to provide a comprehensive overview of the origins and resistance mechanisms of ESKAPE pathogens, while also exploring next-generation treatment strategies for these infections. In addition, it will address both traditional and novel approaches to combating antibiotic resistance, offering insights into potential new therapeutic avenues. Emerging research underscores the urgency of developing new antimicrobial agents and strategies to overcome resistance, highlighting the need for novel drug classes and combination therapies. Advances in genomic technologies and a deeper understanding of microbial pathogenesis are crucial in identifying effective treatments. Integrating precision medicine and personalized approaches could enhance therapeutic efficacy. The review also emphasizes the importance of global collaboration in surveillance and stewardship, as well as policy reforms, enhanced diagnostic tools, and public awareness initiatives, to address resistance on a worldwide scale.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - T P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
4
|
Shen T, Guo J, Han Z, Zhang G, Liu Q, Si X, Wang D, Wu S, Xia J. AutoMolDesigner for Antibiotic Discovery: An AI-Based Open-Source Software for Automated Design of Small-Molecule Antibiotics. J Chem Inf Model 2024; 64:575-583. [PMID: 38265916 DOI: 10.1021/acs.jcim.3c01562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Discovery of small-molecule antibiotics with novel chemotypes serves as one of the essential strategies to address antibiotic resistance. Although a considerable number of computational tools committed to molecular design have been reported, there is a deficit in holistic and efficient tools specifically developed for small-molecule antibiotic discovery. To address this issue, we report AutoMolDesigner, a computational modeling software dedicated to small-molecule antibiotic design. It is a generalized framework comprising two functional modules, i.e., generative-deep-learning-enabled molecular generation and automated machine-learning-based antibacterial activity/property prediction, wherein individually trained models and curated datasets are out-of-the-box for whole-cell-based antibiotic screening and design. It is open-source, thus allowing for the incorporation of new features for flexible use. Unlike most software programs based on Linux and command lines, this application equipped with a Qt-based graphical user interface can be run on personal computers with multiple operating systems, making it much easier to use for experimental scientists. The software and related materials are freely available at GitHub (https://github.com/taoshen99/AutoMolDesigner) and Zenodo (https://zenodo.org/record/10097899).
Collapse
Affiliation(s)
- Tao Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiale Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zunsheng Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Gao Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qingxin Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu 222005, China
| | - Xinxin Si
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu 222005, China
| | - Dongmei Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
5
|
Ansari SB, Kamboj S, Ramalingam K, Meena R, Lal J, Kant R, Shukla SK, Goyal N, Reddy DN. Design and synthesis of N-acyl and dimeric N-Arylpiperazine derivatives as potential antileishmanial agents. Bioorg Chem 2023; 137:106593. [PMID: 37186964 DOI: 10.1016/j.bioorg.2023.106593] [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: 03/15/2023] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
The current regime for leishmaniasis is associated with several adverse effects, expensive, parenteral treatment for longer periods and the emergence of drug resistance. To develop affordable and potent antileishmanial agents, a series of N-acyl and homodimeric aryl piperazines were synthesized with high purity, predicted druggable properties by in silico methods and investigated their antileishmanial activity. The in vitro biological activity of synthesized compounds against clinically validated intracellular amastigote and extracellular promastigote form of Leishmania donovani parasite showed eight compounds inhibited 50% amastigotes growth below 25 µM. The half maximal inhibitory concentration (IC50) and cytotoxicity assessment of eight active compounds, 4a, 4d and 4e demonstrated activity with an IC50 2.0 - 9.1 µM and selectivity index 10 - 42. Compound 4d (IC50 2.0 µM, SI = 42) found to be the best among them with four-folds more potent and eight-folds less toxic than the control drug miltefosine. Overall, results demonstrated that compound 4d is a promising lead candidate for further development as antileishmanial drug.
Collapse
Affiliation(s)
- Shabina B Ansari
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Sakshi Kamboj
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Karthik Ramalingam
- Division Of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Rachana Meena
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Jhajan Lal
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Ruchir Kant
- Division Of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Sanjeev K Shukla
- Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Neena Goyal
- Division Of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Damodara N Reddy
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad 201002, India.
| |
Collapse
|
6
|
Talat A, Khan AU. Artificial intelligence as a smart approach to develop antimicrobial drug molecules: A paradigm to combat drug-resistant infections. Drug Discov Today 2023; 28:103491. [PMID: 36646245 DOI: 10.1016/j.drudis.2023.103491] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Antimicrobial resistance (AMR) is a silent pandemic with the third highest global mortality. The antibiotic development pipeline is scarce even though AMR has escalated uncontrollably. Artificial intelligence (AI) is a revolutionary approach, accelerating drug discovery because of its fast pace, cost efficiency, lower labor requirements, and fewer chances of failure. AI has been used to discover several beta-lactamase inhibitors and antibiotic alternatives from antimicrobial peptides (AMPs), nonribosomal peptides, bacteriocins, and marine natural products. The significant recent increase in the use of AI platforms by pharmaceutical companies could result in the discovery of efficient antibiotic alternatives with lower chances of resistance generation.
Collapse
Affiliation(s)
- Absar Talat
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Asad U Khan
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India.
| |
Collapse
|
7
|
Nag S, Baidya ATK, Mandal A, Mathew AT, Das B, Devi B, Kumar R. Deep learning tools for advancing drug discovery and development. 3 Biotech 2022; 12:110. [PMID: 35433167 PMCID: PMC8994527 DOI: 10.1007/s13205-022-03165-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/18/2022] [Indexed: 12/26/2022] Open
Abstract
A few decades ago, drug discovery and development were limited to a bunch of medicinal chemists working in a lab with enormous amount of testing, validations, and synthetic procedures, all contributing to considerable investments in time and wealth to get one drug out into the clinics. The advancements in computational techniques combined with a boom in multi-omics data led to the development of various bioinformatics/pharmacoinformatics/cheminformatics tools that have helped speed up the drug development process. But with the advent of artificial intelligence (AI), machine learning (ML) and deep learning (DL), the conventional drug discovery process has been further rationalized. Extensive biological data in the form of big data present in various databases across the globe acts as the raw materials for the ML/DL-based approaches and helps in accurate identifications of patterns and models which can be used to identify therapeutically active molecules with much fewer investments on time, workforce and wealth. In this review, we have begun by introducing the general concepts in the drug discovery pipeline, followed by an outline of the fields in the drug discovery process where ML/DL can be utilized. We have also introduced ML and DL along with their applications, various learning methods, and training models used to develop the ML/DL-based algorithms. Furthermore, we have summarized various DL-based tools existing in the public domain with their application in the drug discovery paradigm which includes DL tools for identification of drug targets and drug-target interaction such as DeepCPI, DeepDTA, WideDTA, PADME DeepAffinity, and DeepPocket. Additionally, we have discussed various DL-based models used in protein structure prediction, de novo design of new chemical scaffolds, virtual screening of chemical libraries for hit identification, absorption, distribution, metabolism, excretion, and toxicity (ADMET) prediction, metabolite prediction, clinical trial design, and oral bioavailability prediction. In the end, we have tried to shed light on some of the successful ML/DL-based models used in the drug discovery and development pipeline while also discussing the current challenges and prospects of the application of DL tools in drug discovery and development. We believe that this review will be useful for medicinal and computational chemists searching for DL tools for use in their drug discovery projects.
Collapse
Affiliation(s)
- Sagorika Nag
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| | - Anurag T. K. Baidya
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| | - Abhimanyu Mandal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| | - Alen T. Mathew
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| | - Bhanuranjan Das
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| | - Bharti Devi
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U.), Varanasi, UP 221005 India
| |
Collapse
|
8
|
Zhang J, Zhang J, Liu Q, Fan XX, Leung ELH, Yao XJ, Liu L. Resistance looms for KRAS G12C inhibitors and rational tackling strategies. Pharmacol Ther 2021; 229:108050. [PMID: 34864132 DOI: 10.1016/j.pharmthera.2021.108050] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022]
Abstract
KRAS mutations are one of the most frequent activating alterations in carcinoma. Recent efforts have witnessed a revolutionary strategy for KRAS G12C inhibitors with exhibiting conspicuous clinical responses across multiple tumor types, providing new impetus for renewed drug development and culminating in sotorasib with approximately 6-month median progression-free survival in KRAS G12C-driven lung cancer. However, diverse genomic and histological mechanisms conferring resistance to KRAS G12C inhibitors may limit their clinical efficacy. Herein, we first briefly discuss the recent resistance looms for KRAS G12C inhibitors, focusing on their clinical trials. We then comprehensively interrogate and underscore our current understanding of resistance mechanisms and the necessity of incorporating genomic analyses into the clinical investigation to further decipher resistance mechanisms. Finally, we highlight the future role of novel treatment strategies especially rational identification of targeted combinatorial approaches in tackling drug resistance, and propose our views on including the application of robust biomarkers to precisely guide combination medication regimens.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China; School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Juanhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China; School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China; College of Life Science, Northwest Normal University, Lanzhou 730070, China
| | - Qing Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China.
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China.
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China.
| |
Collapse
|
9
|
Sharma S, Arya A, Cruz R, Cleaves II HJ. Automated Exploration of Prebiotic Chemical Reaction Space: Progress and Perspectives. Life (Basel) 2021; 11:1140. [PMID: 34833016 PMCID: PMC8624352 DOI: 10.3390/life11111140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Prebiotic chemistry often involves the study of complex systems of chemical reactions that form large networks with a large number of diverse species. Such complex systems may have given rise to emergent phenomena that ultimately led to the origin of life on Earth. The environmental conditions and processes involved in this emergence may not be fully recapitulable, making it difficult for experimentalists to study prebiotic systems in laboratory simulations. Computational chemistry offers efficient ways to study such chemical systems and identify the ones most likely to display complex properties associated with life. Here, we review tools and techniques for modelling prebiotic chemical reaction networks and outline possible ways to identify self-replicating features that are central to many origin-of-life models.
Collapse
Affiliation(s)
- Siddhant Sharma
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA; (S.S.); (A.A.); (R.C.)
- Department of Biochemistry, Deshbandhu College, University of Delhi, New Delhi 110019, India
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Aayush Arya
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA; (S.S.); (A.A.); (R.C.)
- Department of Physics, Lovely Professional University, Jalandhar-Delhi GT Road, Phagwara 144001, India
| | - Romulo Cruz
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA; (S.S.); (A.A.); (R.C.)
- Big Data Laboratory, Information and Communications Technology Center (CTIC), National University of Engineering, Amaru 210, Lima 15333, Peru
| | - Henderson James Cleaves II
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA; (S.S.); (A.A.); (R.C.)
- Earth-Life Science Institute, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| |
Collapse
|