Exploring the Advantages of Quantum Generative Adversarial Networks in Generative ChemistryClick to copy article linkArticle link copied!
- Po-Yu Kao
- Ya-Chu Yang
- Wei-Yin Chiang
- Jen-Yueh Hsiao
- Yudong Cao
- Alex Aliper
- Feng Ren
- Alán Aspuru-GuzikAlán Aspuru-GuzikDepartment of Chemistry, University of Toronto, Toronto, ON M5S 3H6, CanadaDepartment of Computer Science, University of Toronto, Toronto, ON M5S 2E4, CanadaVector Institute for Artificial Intelligence, Toronto, ON M5S 1M1, CanadaLebovic Fellow, Canadian Institute for Advanced Research, Toronto, ON M5S 1M1, CanadaMore by Alán Aspuru-Guzik
- Alex Zhavoronkov*Alex Zhavoronkov*E-mail: [email protected]Insilico Medicine Hong Kong Ltd., Hong Kong SAR, 999077, ChinaMore by Alex Zhavoronkov
- Min-Hsiu Hsieh*Min-Hsiu Hsieh*E-mail: [email protected]Hon Hai (Foxconn) Research Institute, Taipei 114699, TaiwanMore by Min-Hsiu Hsieh
- Yen-Chu Lin*Yen-Chu Lin*E-mail: [email protected]Insilico Medicine Taiwan Ltd., Taipei 110208, TaiwanDepartment of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, TaiwanMore by Yen-Chu Lin
Abstract
De novo drug design with desired biological activities is crucial for developing novel therapeutics for patients. The drug development process is time- and resource-consuming, and it has a low probability of success. Recent advances in machine learning and deep learning technology have reduced the time and cost of the discovery process and therefore, improved pharmaceutical research and development. In this paper, we explore the combination of two rapidly developing fields with lead candidate discovery in the drug development process. First, artificial intelligence has already been demonstrated to successfully accelerate conventional drug design approaches. Second, quantum computing has demonstrated promising potential in different applications, such as quantum chemistry, combinatorial optimizations, and machine learning. This article explores hybrid quantum-classical generative adversarial networks (GAN) for small molecule discovery. We substituted each element of GAN with a variational quantum circuit (VQC) and demonstrated the quantum advantages in the small drug discovery. Utilizing a VQC in the noise generator of a GAN to generate small molecules achieves better physicochemical properties and performance in the goal-directed benchmark than the classical counterpart. Moreover, we demonstrate the potential of a VQC with only tens of learnable parameters in the generator of GAN to generate small molecules. We also demonstrate the quantum advantage of a VQC in the discriminator of GAN. In this hybrid model, the number of learnable parameters is significantly less than the classical ones, and it can still generate valid molecules. The hybrid model with only tens of training parameters in the quantum discriminator outperforms the MLP-based one in terms of both generated molecule properties and the achieved KL divergence. However, the hybrid quantum-classical GANs still face challenges in generating unique and valid molecules compared to their classical counterparts.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Computational Results and Discussion
Figure 1
Figure 1. Overall pipeline. (a) The overall pipeline of MolGAN with different combinations of classical/quantum components. The reward neural network branch is enabled in the goal-directed benchmark. The classical noise generator samples from the Gaussian distribution, and the quantum one uses the variational quantum circuit (VQC). The classical generator is built by neural networks, and the quantum one uses the patch-based VQC to generate the molecular graph. The molecular graph is represented by a bond matrix and atom vector. The classical discriminator is built by a graph-based neural network or multilayer perceptron (MLP), and the VQC is used in the quantum one. (b) The example of VQC in the noise generator. (c) The patch method uses multiple VQCs as subgenerators. Each subgenerator takes noise as input and outputs a partial part of the final molecular graph. The final molecular graph is constructed by concatenating all the partial patches together. (d) The example of VQC in the quantum generator. (e) The VQC of the quantum discriminator consists of the amplitude embedding circuit (Sx), the strong entanglement layers (Uθ), and the measurement. (f) The VQC of strongly entanglement layers contains multiple CNOT gates and parametrized rotational gates (R). (g) MLP-based discriminator architecture in MolGAN-CC.
Quantum Noise Generator
z_dim = 2 | z_dim = 3 | z_dim = 4 | |||||||
---|---|---|---|---|---|---|---|---|---|
QuMolGAN | MolGAN | p-value | QuMolGAN | MolGAN | p-value | QuMolGAN | MolGAN | p-value | |
number of moleculesb | 363 | 657 | – | 414 | 2163 | – | 511 | 3085 | – |
QED ↑ | 0.489 | 0.475 | <0.01 | 0.489 | 0.465 | <0.01 | 0.473 | 0.465 | <0.05 |
solubility ↑ | 0.343 | 0.324 | <0.05 | 0.370 | 0.305 | <0.01 | 0.317 | 0.298 | <0.01 |
SA ↑ | 0.367 | 0.336 | <0.05 | 0.310 | 0.307 | <0.05 | 0.308 | 0.296 | 0.246 |
KL score (S)c ↑ | 0.653 | 0.824 | – | 0.797 | 0.913 | – | 0.846 | 0.957 | – |
Bold numbers highlight the better scores in QuMolGAN, compared to the corresponding MolGAN. Note that the QED, Solute, and SA scores in this table are calculated from the valid and unique molecules.
Number of valid and unique molecules from 5000 samples.
From eq 2.
Figure 2
Figure 2. Property distributions of molecules. (a) Drug properties distributions (left to right: QED, SA, and Solute) from valid and unique MolGAN-generated (in blue) and QuMolGAN-generated (in orange) molecules. (b) KL-divergence distributions (left to right: MolLogP, BertzCT, and MolWt) of valid and unique MolGAN-generated (blue), QuMolGAN-generated (orange), and QM9 (gray) molecules. (c) KL-divergence distributions (from left to right: MolLogP, BertzCT, and MolWt) of MolGAN-CC-ER-generated (orange), MolGAN-CQ-generated (yellow), MolGAN-generated (blue), and QM9 (gray) molecules.
Figure 3
Figure 3. Example molecules. (a) Example molecules of MolGAN with z_dim = 2. (b) Example molecules of QuMolGAN with z_dim = 2. (c) Example molecules of MolGAN-CQ.
Goal-Directed Benchmark
MolGAN | QuMolGAN | |||||
---|---|---|---|---|---|---|
α = 1.0 | α = 0.5 | α = 0.01 | α = 1.0 | α = 0.5 | α = 0.01 | |
number of moleculesb | 2890 | 2700 | 696 | 534 | 309 | 116 |
validity ↑ | 80.40 | 78.48 | 68.76 | 70.02 | 70.32 | 42.94 |
uniqueness ↑ | 71.89 | 68.81 | 20.24 | 15.25 | 8.78 | 5.40 |
QED ↑ | 0.47 | 0.48 | 0.52 | 0.47 | 0.49 | 0.57 |
Solute ↑ | 0.31 | 0.31 | 0.45 | 0.32 | 0.30 | 0.44 |
SA ↑ | 0.30 | 0.31 | 0.65 | 0.29 | 0.28 | 0.76 |
KL Score (S)c ↑ | 0.95 | 0.94 | 0.58 | 0.92 | 0.82 | 0.31 |
Note that the QED, Solute, and SA scores in this table are calculated from the valid molecules. Bold numbers indicate better scores among the same type of models with different RL weights α, and the underlined numbers indicate the best scores across different types of models.
Number of valid and unique molecules from 5000 samples.
From eq 2.
Quantum Generator
# epoch | number of moleculesb | validity ↑ | uniqueness ↑ | novelty ↑ | diversity ↑ | QED ↑ | solubility ↑ | SA ↑ | KL Score (S)c ↑ |
---|---|---|---|---|---|---|---|---|---|
1 | 73 | 79.39 | 4.49 | 100 | 1.00 | 0.43 | 0.75 | 0.24 | 0.24 |
2 | 54 | 76.37 | 3.45 | 100 | 1.00 | 0.47 | 0.75 | 0.24 | 0.25 |
3 | 43 | 78.47 | 2.68 | 100 | 1.00 | 0.48 | 0.75 | 0.11 | 0.28 |
4 | 29 | 78.61 | 1.80 | 100 | 1.00 | 0.48 | 0.75 | 0.13 | 0.29 |
5 | 30 | 77.93 | 1.88 | 100 | 1.00 | 0.48 | 0.75 | 0.14 | 0.30 |
6 | 40 | 78.37 | 2.49 | 100 | 1.00 | 0.48 | 0.75 | 0.09 | 0.21 |
7 | 29 | 80.27 | 1.76 | 100 | 1.00 | 0.47 | 0.75 | 0.06 | 0.28 |
8 | 39 | 78.91 | 2.41 | 100 | 1.00 | 0.48 | 0.75 | 0.16 | 0.28 |
9 | 41 | 74.66 | 2.68 | 100 | 1.00 | 0.48 | 0.75 | 0.22 | 0.25 |
10 | 29 | 79.74 | 1.78 | 100 | 1.00 | 0.48 | 0.75 | 0.08 | 0.27 |
The QED, Solute, and SA scores in this table are calculated from the valid molecules.
Number of valid and unique molecules from 2048 samples from Gaussian distribution.
From eq 2.
Quantum Discriminator
MolGAN-CQ Architecture and Training Details
Comparison with Classical MolGAN
MolGAN | MolGAN-CQ | |
---|---|---|
# of moleculesb | 2693 | 730 |
Validity ↑ | 76 | 31.34 |
Uniqueness ↑ | 70.87 | 46.59 |
QED ↑ | 0.47 | 0.48 |
Solute ↑ | 0.31 | 0.44 |
SA ↑ | 0.31 | 0.66 |
KL Score (S)c↑ | 0.94 | 0.75 |
The models are only trained for 30 epochs. Bold numbers indicate better scores. The QED, Solute, and SA scores in this table are calculated from the valid molecules.
Number of valid and unique molecules from 5000 samples.
From eq 2
Comparison with MolGAN-CC
MolGAN-CQ | MolGAN-CC-ER | MolGAN-CC-HR | MolGAN-CC-NR | |
---|---|---|---|---|
number of parameters | 50 | 22K | 45K | 82K |
number of moleculesb | 730 | 104 | 1919 | 2284 |
Validity ↑ | 31.34 | 99.78 | 44.1 | 54.7 |
Uniqueness ↑ | 46.59 | 2.08 | 87.03 | 83.51 |
QED ↑ | 0.48 | 0.51 | 0.49 | 0.5 |
Solute ↑ | 0.44 | 0.63 | 0.35 | 0.38 |
SA ↑ | 0.66 | 0.97 | 0.48 | 0.50 |
KL Score (S)c↑ | 0.75 | 0.28 | 0.84 | 0.81 |
Bold numbers indicate better performance across different types of models. The QED, Solute, and SA scores in this table are calculated from the valid molecules.
Number of valid and unique molecules from 5000 samples.
From eq 2
Conclusions
Materials and Methods
Evaluation Method and Metrics
Dataset
Methodology
Variational Quantum Circuits (VQCs)
VQC of Noise Generator
VQC of Quantum Generator
VQC of Quantum Discriminator
Data Availability
The data acquisition code and source codes associated with this study are publicly available at https://github.com/pykao/QuantumMolGAN-PyTorch. The discriminator branch contains a classical/quantum noise generator, a classical generator, and a classical/quantum discriminator. This branch can be used for the quantum noise generator and the quantum discriminator. The generator branch contains a classical/quantum noise generator, a classical/quantum generator, and a classical discriminator. This branch can be used for the quantum generator.
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.3c00562.
Different complexities of generators in MolGAN; different input noise dimensions of the generator in MolGAN-HR; different parametrized layers of QuMolGAN-HR; all the combinations of the classical/quantum noise/generator/discriminator and their corresponding model name; different numbers of qubits in the quantum circuit of QuMolGAN-HR; the details of MolGAN-CC models with the varied size of discriminators; example molecules from the quantum generator; integration of proposed hybrid generative models with Insilico Medicine Chemistry42 (62) platform; example molecules of QM9 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 78 other publications.
- 1Hingorani, A. D.; Kuan, V.; Finan, C.; Kruger, F. A.; Gaulton, A.; Chopade, S.; Sofat, R.; MacAllister, R. J.; Overington, J. P.; Hemingway, H.; Denaxas, S.; Prieto, D.; Casas, J. P. Improving the odds of drug development success through human genomics: modelling study. Sci. Rep. 2019, 9, 1– 25, DOI: 10.1038/s41598-019-54849-wGoogle ScholarThere is no corresponding record for this reference.
- 2Abreu, J. L. Ivermectin for the Prevention of COVID-19 So...WHO is Telling the Truth. Revista Daena (Int. J. Good Conscience) 2020, 15 (2), 1– 30Google ScholarThere is no corresponding record for this reference.
- 3Wouters, O. J.; McKee, M.; Luyten, J. Estimated research and development investment needed to bring a new medicine to market, 2009–2018. Jama 2020, 323, 844– 853, DOI: 10.1001/jama.2020.1166Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB387oslyhsA%253D%253D&md5=ff63c2f03e59ef0223585a71fb6d5df0Estimated Research and Development Investment Needed to Bring a New Medicine to Market, 2009-2018Wouters Olivier J; McKee Martin; Luyten JeroenJAMA (2020), 323 (9), 844-853 ISSN:.IMPORTANCE: The mean cost of developing a new drug has been the subject of debate, with recent estimates ranging from $314 million to $2.8 billion. OBJECTIVE: To estimate the research and development investment required to bring a new therapeutic agent to market, using publicly available data. DESIGN AND SETTING: Data were analyzed on new therapeutic agents approved by the US Food and Drug Administration (FDA) between 2009 and 2018 to estimate the research and development expenditure required to bring a new medicine to market. Data were accessed from the US Securities and Exchange Commission, Drugs@FDA database, and ClinicalTrials.gov, alongside published data on clinical trial success rates. EXPOSURES: Conduct of preclinical and clinical studies of new therapeutic agents. MAIN OUTCOMES AND MEASURES: Median and mean research and development spending on new therapeutic agents approved by the FDA, capitalized at a real cost of capital rate (the required rate of return for an investor) of 10.5% per year, with bootstrapped CIs. All amounts were reported in 2018 US dollars. RESULTS: The FDA approved 355 new drugs and biologics over the study period. Research and development expenditures were available for 63 (18%) products, developed by 47 different companies. After accounting for the costs of failed trials, the median capitalized research and development investment to bring a new drug to market was estimated at $985.3 million (95% CI, $683.6 million-$1228.9 million), and the mean investment was estimated at $1335.9 million (95% CI, $1042.5 million-$1637.5 million) in the base case analysis. Median estimates by therapeutic area (for areas with ≥5 drugs) ranged from $765.9 million (95% CI, $323.0 million-$1473.5 million) for nervous system agents to $2771.6 million (95% CI, $2051.8 million-$5366.2 million) for antineoplastic and immunomodulating agents. Data were mainly accessible for smaller firms, orphan drugs, products in certain therapeutic areas, first-in-class drugs, therapeutic agents that received accelerated approval, and products approved between 2014 and 2018. Results varied in sensitivity analyses using different estimates of clinical trial success rates, preclinical expenditures, and cost of capital. CONCLUSIONS AND RELEVANCE: This study provides an estimate of research and development costs for new therapeutic agents based on publicly available data. Differences from previous studies may reflect the spectrum of products analyzed, the restricted availability of data in the public domain, and differences in underlying assumptions in the cost calculations.
- 4Aliper, A.; Plis, S.; Artemov, A.; Ulloa, A.; Mamoshina, P.; Zhavoronkov, A. Deep learning applications for predicting pharmacological properties of drugs and drug repurposing using transcriptomic data. Mol. Pharmaceutics 2016, 13, 2524– 2530, DOI: 10.1021/acs.molpharmaceut.6b00248Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xot1ers7w%253D&md5=853efb9b636e4c3c8a15bd4c53ecac99Deep Learning Applications for Predicting Pharmacological Properties of Drugs and Drug Repurposing Using Transcriptomic DataAliper, Alexander; Plis, Sergey; Artemov, Artem; Ulloa, Alvaro; Mamoshina, Polina; Zhavoronkov, AlexMolecular Pharmaceutics (2016), 13 (7), 2524-2530CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Deep learning is rapidly advancing many areas of science and technol. with multiple success stories in image, text, voice and video recognition, robotics, and autonomous driving. In this paper we demonstrate how deep neural networks (DNN) trained on large transcriptional response data sets can classify various drugs to therapeutic categories solely based on their transcriptional profiles. We used the perturbation samples of 678 drugs across A549, MCF-7, and PC-3 cell lines from the LINCS Project and linked those to 12 therapeutic use categories derived from MeSH. To train the DNN, we utilized both gene level transcriptomic data and transcriptomic data processed using a pathway activation scoring algorithm, for a pooled data set of samples perturbed with different concns. of the drug for 6 and 24 h. In both pathway and gene level classification, DNN achieved high classification accuracy and convincingly outperformed the support vector machine (SVM) model on every multiclass classification problem, however, models based on pathway level data performed significantly better. For the first time we demonstrate a deep learning neural net trained on transcriptomic data to recognize pharmacol. properties of multiple drugs across different biol. systems and conditions. We also propose using deep neural net confusion matrixes for drug repositioning. This work is a proof of principle for applying deep learning to drug discovery and development.
- 5Kadurin, A.; Aliper, A.; Kazennov, A.; Mamoshina, P.; Vanhaelen, Q.; Khrabrov, K.; Zhavoronkov, A. The cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncology. Oncotarget 2017, 8, 10883, DOI: 10.18632/oncotarget.14073Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1c%252FpvFKruw%253D%253D&md5=677ef0264494eb8a7ef8c6584c1202abThe cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncologyKadurin Artur; Khrabrov Kuzma; Kadurin Artur; Aliper Alexander; Kazennov Andrey; Mamoshina Polina; Vanhaelen Quentin; Zhavoronkov Alex; Kadurin Artur; Kadurin Artur; Kazennov Andrey; Zhavoronkov Alex; Mamoshina Polina; Zhavoronkov AlexOncotarget (2017), 8 (7), 10883-10890 ISSN:.Recent advances in deep learning and specifically in generative adversarial networks have demonstrated surprising results in generating new images and videos upon request even using natural language as input. In this paper we present the first application of generative adversarial autoencoders (AAE) for generating novel molecular fingerprints with a defined set of parameters. We developed a 7-layer AAE architecture with the latent middle layer serving as a discriminator. As an input and output the AAE uses a vector of binary fingerprints and concentration of the molecule. In the latent layer we also introduced a neuron responsible for growth inhibition percentage, which when negative indicates the reduction in the number of tumor cells after the treatment. To train the AAE we used the NCI-60 cell line assay data for 6252 compounds profiled on MCF-7 cell line. The output of the AAE was used to screen 72 million compounds in PubChem and select candidate molecules with potential anti-cancer properties. This approach is a proof of concept of an artificially-intelligent drug discovery engine, where AAEs are used to generate new molecular fingerprints with the desired molecular properties.
- 6Zhavoronkov, A. Artificial intelligence for drug discovery, biomarker development, and generation of novel chemistry. Mol. Pharmaceutics 2018, 15, 4311, DOI: 10.1021/acs.molpharmaceut.8b00930Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslOgtrjE&md5=2c51295d4e682eeaf8069eddc09ed3e8Artificial Intelligence for Drug Discovery, Biomarker Development, and Generation of Novel ChemistryZhavoronkov, AlexMolecular Pharmaceutics (2018), 15 (10), 4311-4313CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The potential of artificial intelligence in drug discovery is discussed and reviewed.
- 7Carracedo-Reboredo, P.; Liñares-Blanco, J.; Rodríguez-Fernández, N.; Cedrón, F.; Novoa, F. J.; Carballal, A.; Maojo, V.; Pazos, A.; Fernandez-Lozano, C. A review on machine learning approaches and trends in drug discovery. Comput. Struct. Biotechnol. J. 2021, 19, 4538– 4558, DOI: 10.1016/j.csbj.2021.08.011Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisF2gs7zM&md5=eccac145867b8821f0719463ec1be641A review on machine learning approaches and trends in drug discoveryCarracedo-Reboredo, Paula; Linares-Blanco, Jose; Rodriguez-Fernandez, Nereida; Cedron, Francisco; Novoa, Francisco J.; Carballal, Adrian; Maojo, Victor; Pazos, Alejandro; Fernandez-Lozano, CarlosComputational and Structural Biotechnology Journal (2021), 19 (), 4538-4558CODEN: CSBJAC; ISSN:2001-0370. (Elsevier B.V.)A review. Drug discovery aims at finding new compds. with specific chem. properties for the treatment of diseases. In the last years, the approach used in this search presents an important component in computer science with the skyrocketing of machine learning techniques due to its democratization. With the objectives set by the Precision Medicine initiative and the new challenges generated, it is necessary to establish robust, std. and reproducible computational methodologies to achieve the objectives set. Currently, predictive models based on Machine Learning have gained great importance in the step prior to preclin. studies. This stage manages to drastically reduce costs and research times in the discovery of new drugs. This review article focuses on how these new methodologies are being used in recent years of research. Analyzing the state of the art in this field will give us an idea of where cheminformatics will be developed in the short term, the limitations it presents and the pos. results it has achieved. This review will focus mainly on the methods used to model the mol. data, as well as the biol. problems addressed and the Machine Learning algorithms used for drug discovery in recent years.
- 8Kao, P.-Y.; Kao, S.-M.; Huang, N.-L.; Lin, Y.-C. Toward Drug-Target Interaction Prediction via Ensemble Modeling and Transfer Learning. In 2021 IEEE International Conference on Bioinformatics and Biomedicine (BIBM) , 2021; pp 2384– 2391.Google ScholarThere is no corresponding record for this reference.
- 9Kolluri, S.; Lin, J.; Liu, R.; Zhang, Y.; Zhang, W. Machine Learning and Artificial Intelligence in Pharmaceutical Research and Development: a Review. AAPS J. 2022, 24, 1– 10, DOI: 10.1208/s12248-021-00644-3Google ScholarThere is no corresponding record for this reference.
- 10Zhavoronkov, A.; Ivanenkov, Y. A.; Aliper, A.; Veselov, M. S.; Aladinskiy, V. A.; Aladinskaya, A. V.; Terentiev, V. A.; Polykovskiy, D. A.; Kuznetsov, M. D.; Asadulaev, A.; Volkov, Y.; Zholus, A.; Shayakhmetov, R. R.; Zhebrak, A.; Minaeva, L. I.; Zagribelnyy, B. A.; Lee, L. H.; Soll, R.; Madge, D.; Xing, L.; Guo, T.; Aspuru-Guzik, A. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat. Biotechnol. 2019, 37, 1038– 1040, DOI: 10.1038/s41587-019-0224-xGoogle Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12gurnM&md5=b15262b61b9172ab2bc37e534a70f010Deep learning enables rapid identification of potent DDR1 kinase inhibitorsZhavoronkov, Alex; Ivanenkov, Yan A.; Aliper, Alex; Veselov, Mark S.; Aladinskiy, Vladimir A.; Aladinskaya, Anastasiya V.; Terentiev, Victor A.; Polykovskiy, Daniil A.; Kuznetsov, Maksim D.; Asadulaev, Arip; Volkov, Yury; Zholus, Artem; Shayakhmetov, Rim R.; Zhebrak, Alexander; Minaeva, Lidiya I.; Zagribelnyy, Bogdan A.; Lee, Lennart H.; Soll, Richard; Madge, David; Xing, Li; Guo, Tao; Aspuru-Guzik, AlanNature Biotechnology (2019), 37 (9), 1038-1040CODEN: NABIF9; ISSN:1087-0156. (Nature Research)We have developed a deep generative model, generative tensorial reinforcement learning (GENTRL), for de novo small-mol. design. GENTRL optimizes synthetic feasibility, novelty, and biol. activity. We used GENTRL to discover potent inhibitors of discoidin domain receptor 1 (DDR1), a kinase target implicated in fibrosis and other diseases, in 21 days. Four compds. were active in biochem. assays, and two were validated in cell-based assays. One lead candidate was tested and demonstrated favorable pharmacokinetics in mice.
- 11Chan, H. S.; Shan, H.; Dahoun, T.; Vogel, H.; Yuan, S. Advancing drug discovery via artificial intelligence. Trends Pharmacol. Sci. 2019, 40, 592– 604, DOI: 10.1016/j.tips.2019.06.004Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlalsbbL&md5=03ebb3bc60278061eb29feb531b92d02Advancing Drug Discovery via Artificial IntelligenceChan, H. C. Stephen; Shan, Hanbin; Dahoun, Thamani; Vogel, Horst; Yuan, ShuguangTrends in Pharmacological Sciences (2019), 40 (8), 592-604CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)Drug discovery and development are among the most important translational science activities that contribute to human health and wellbeing. However, the development of a new drug is a very complex, expensive, and long process which typically costs 2.6 billion USD and takes 12 years on av. How to decrease the costs and speed up new drug discovery has become a challenging and urgent question in industry. Artificial intelligence (AI) combined with new exptl. technologies is expected to make the hunt for new pharmaceuticals quicker, cheaper, and more effective. We discuss here emerging applications of AI to improve the drug discovery process.
- 12Schneider, G.; Fechner, U. Computer-based de novo design of drug-like molecules. Nat. Rev. Drug Discovery 2005, 4, 649– 663, DOI: 10.1038/nrd1799Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmvVOqtro%253D&md5=a30dbc58ed81e0b7fe3f7d41a668e9acComputer-based de novo design of drug-like moleculesSchneider, Gisbert; Fechner, UliNature Reviews Drug Discovery (2005), 4 (8), 649-663CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review with refs. Ever since the first automated de novo design techniques were conceived only 15 years ago, the computer-based design of hit and lead structure candidates has emerged as a complementary approach to high-throughput screening. Although many challenges remain, de novo design supports drug discovery projects by generating novel pharmaceutically active agents with desired properties in a cost- and time-efficient manner. In this review, we outline the various design concepts and highlight current developments in computer-based de novo design.
- 13Fischer, T.; Gazzola, S.; Riedl, R. Approaching target selectivity by de novo drug design. Expert Opin. Drug Discovery 2019, 14, 791– 803, DOI: 10.1080/17460441.2019.1615435Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFeqtrbJ&md5=fd845eb91cde2a8042ec28fc2b97dd32Approaching Target Selectivity by De Novo Drug DesignFischer, Thomas; Gazzola, Silvia; Riedl, RainerExpert Opinion on Drug Discovery (2019), 14 (8), 791-803CODEN: EODDBX; ISSN:1746-0441. (Taylor & Francis Ltd.)A review. The development of drug candidates with a defined selectivity profile and a unique mol. structure is of fundamental interest for drug discovery. In contrast to the costly screening of large substance libraries, the targeted de novo design of a drug by using structural information of either the biol. target and/or structure-activity relationship data of active modulators offers an efficient and intellectually appealing alternative.: This review provides an overview on the different techniques of de novo drug design (ligand-based drug design, structure-based drug design, and fragment-based drug design) and highlights successful examples of this targeted approach toward selective modulators of therapeutically relevant targets.: De novo drug design has established itself as a very efficient method for the development of potent and selective modulators for a variety of different biol. target classes. The ever-growing wealth of structural data on therapeutic targets will certainly further enhance the importance of de novo design for the drug discovery process in the future. However, a consistent use of the terminol. of de novo drug design in the scientific literature should be sought.
- 14Mouchlis, V. D.; Afantitis, A.; Serra, A.; Fratello, M.; Papadiamantis, A. G.; Aidinis, V.; Lynch, I.; Greco, D.; Melagraki, G. Advances in de novo drug design: from conventional to machine learning methods. Int. J. Mol. Sci. 2021, 22, 1676, DOI: 10.3390/ijms22041676Google ScholarThere is no corresponding record for this reference.
- 15Speck-Planche, A. Recent advances in fragment-based computational drug design: tackling simultaneous targets/biological effects. Future Med. Chem. 2018, 10, 2021– 2024, DOI: 10.4155/fmc-2018-0213Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsF2gsLzJ&md5=a6a28a9e6dde2e290064bdf0f646b7e1Recent advances in fragment-based computational drug design: tackling simultaneous targets/biological effectsSpeck-Planche, AlejandroFuture Medicinal Chemistry (2018), 10 (17), 2021-2024CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)There is no expanded citation for this reference.
- 16Mamoshina, P.; Ojomoko, L.; Yanovich, Y.; Ostrovski, A.; Botezatu, A.; Prikhodko, P.; Izumchenko, E.; Aliper, A.; Romantsov, K.; Zhebrak, A.; Ogu, I. O.; Zhavoronkov, A. Converging blockchain and next-generation artificial intelligence technologies to decentralize and accelerate biomedical research and healthcare. Oncotarget 2018, 9, 5665, DOI: 10.18632/oncotarget.22345Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mrjs1Ontw%253D%253D&md5=d2ee7d11f2b610408b9ad2007d0d022bConverging blockchain and next-generation artificial intelligence technologies to decentralize and accelerate biomedical research and healthcareMamoshina Polina; Ojomoko Lucy; Aliper Alexander; Romantsov Konstantin; Zhebrak Alexander; Zhavoronkov Alex; Mamoshina Polina; Yanovich Yury; Ostrovski Alex; Botezatu Alex; Prikhodko Pavel; Izumchenko Eugene; Ogu Iraneus Obioma; Zhavoronkov AlexOncotarget (2018), 9 (5), 5665-5690 ISSN:.The increased availability of data and recent advancements in artificial intelligence present the unprecedented opportunities in healthcare and major challenges for the patients, developers, providers and regulators. The novel deep learning and transfer learning techniques are turning any data about the person into medical data transforming simple facial pictures and videos into powerful sources of data for predictive analytics. Presently, the patients do not have control over the access privileges to their medical records and remain unaware of the true value of the data they have. In this paper, we provide an overview of the next-generation artificial intelligence and blockchain technologies and present innovative solutions that may be used to accelerate the biomedical research and enable patients with new tools to control and profit from their personal data as well with the incentives to undergo constant health monitoring. We introduce new concepts to appraise and evaluate personal records, including the combination-, time- and relationship-value of the data. We also present a roadmap for a blockchain-enabled decentralized personal health data ecosystem to enable novel approaches for drug discovery, biomarker development, and preventative healthcare. A secure and transparent distributed personal data marketplace utilizing blockchain and deep learning technologies may be able to resolve the challenges faced by the regulators and return the control over personal data including medical records back to the individuals.
- 17Zhavoronkov, A.; Zagribelnyy, B.; Zhebrak, A.; Aladinskiy, V.; Terentiev, V.; Vanhaelen, Q.; Bezrukov, D. S.; Polykovskiy, D.; Shayakhmetov, R.; Filimonov, A.; Filimonov, A.; Bishop, M.; McCloskey, S.; Lejia, E.; Bright, D.; Funakawa, K.; Lin, Y.-C.; Huang, S.-H.; Liao, H.-J.; Aliper, A.; Ivanenkov, Y. Potential non-covalent SARS-CoV-2 3C-like protease inhibitors designed using generative deep learning approaches and reviewed by human medicinal chemist in virtual reality. ChemRxiv , 2020.Google ScholarThere is no corresponding record for this reference.
- 18Paul, D.; Sanap, G.; Shenoy, S.; Kalyane, D.; Kalia, K.; Tekade, R. K. Artificial intelligence in drug discovery and development. Drug Discovery Today 2021, 26, 80, DOI: 10.1016/j.drudis.2020.10.010Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlentrbI&md5=092fc22fb6e29075f3ccd8867f0c65f1Artificial intelligence in drug discovery and developmentPaul, Debleena; Sanap, Gaurav; Shenoy, Snehal; Kalyane, Dnyaneshwar; Kalia, Kiran; Tekade, Rakesh K.Drug Discovery Today (2021), 26 (1), 80-93CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. Artificial Intelligence (AI) has recently started to gear-up its application in various sectors of the society with the pharmaceutical industry as a front-runner beneficiary. This review highlights the impactful use of AI in diverse areas of the pharmaceutical sectors viz., drug discovery and development, drug repurposing, improving pharmaceutical productivity, clin. trials, etc. to name a few, thus reducing the human workload as well as achieving targets in a short period. Crosstalk on the tools and techniques utilized in enforcing AI, ongoing challenges, and ways to overcome them, along with the future of AI in the pharmaceutical industry, is also discussed.
- 19Martinelli, D. Generative machine learning for de novo drug discovery: A systematic review. Comput. Biol. Med. 2022, 145, 105403, DOI: 10.1016/j.compbiomed.2022.105403Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2MzptFantQ%253D%253D&md5=01320d8d3a5987a1634dc2142343cd39Generative machine learning for de novo drug discovery: A systematic reviewMartinelli Dominic DComputers in biology and medicine (2022), 145 (), 105403 ISSN:.Recent research on artificial intelligence indicates that machine learning algorithms can auto-generate novel drug-like molecules. Generative models have revolutionized de novo drug discovery, rendering the explorative process more efficient. Several model frameworks and input formats have been proposed to enhance the performance of intelligent algorithms in generative molecular design. In this systematic literature review of experimental articles and reviews over the last five years, machine learning models, challenges associated with computational molecule design along with proposed solutions, and molecular encoding methods are discussed. A query-based search of the PubMed, ScienceDirect, Springer, Wiley Online Library, arXiv, MDPI, bioRxiv, and IEEE Xplore databases yielded 87 studies. Twelve additional studies were identified via citation searching. Of the articles in which machine learning was implemented, six prominent algorithms were identified: long short-term memory recurrent neural networks (LSTM-RNNs), variational autoencoders (VAEs), generative adversarial networks (GANs), adversarial autoencoders (AAEs), evolutionary algorithms, and gated recurrent unit (GRU-RNNs). Furthermore, eight central challenges were designated: homogeneity of generated molecular libraries, deficient synthesizability, limited assay data, model interpretability, incapacity for multi-property optimization, incomparability, restricted molecule size, and uncertainty in model evaluation. Molecules were encoded either as strings, which were occasionally augmented using randomization, as 2D graphs, or as 3D graphs. Statistical analysis and visualization are performed to illustrate how approaches to machine learning in de novo drug design have evolved over the past five years. Finally, future opportunities and reservations are discussed.
- 20Gao, K.; Nguyen, D. D.; Tu, M.; Wei, G.-W. Generative Network Complex for the Automated Generation of Drug-like Molecules. J. Chem. Inf. Model. 2020, 60, 5682– 5698, DOI: 10.1021/acs.jcim.0c00599Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVSms7jL&md5=d19c56b0b0752814fc8b8c5d2e50144cGenerative Network Complex for the Automated Generation of Drug-like MoleculesGao, Kaifu; Nguyen, Duc Duy; Tu, Meihua; Wei, Guo-WeiJournal of Chemical Information and Modeling (2020), 60 (12), 5682-5698CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Current drug discovery is expensive and time-consuming. It remains a challenging task to create a wide variety of novel compds. that not only have desirable pharmacol. properties but also are cheaply available to low-income people. In this work, we develop a generative network complex (GNC) to generate new drug-like mols. based on the multiproperty optimization via the gradient descent in the latent space of an autoencoder. In our GNC, both multiple chem. properties and similarity scores are optimized to generate drug-like mols. with desired chem. properties. To further validate the reliability of the predictions, these mols. are reevaluated and screened by independent 2D fingerprint-based predictors to come up with a few hundreds of new drug candidates. As a demonstration, we apply our GNC to generate a large no. of new BACE1 inhibitors, as well as thousands of novel alternative drug candidates for eight existing market drugs, including Ceritinib, Ribociclib, Acalabrutinib, Idelalisib, Dabrafenib, Macimorelin, Enzalutamide, and Panobinostat.
- 21Goodfellow, I.; Pouget-Abadie, J.; Mirza, M.; Xu, B.; Warde-Farley, D.; Ozair, S.; Courville, A.; Bengio, Y. Generative adversarial nets. In Advances in Neural Information Processing Systems 27 (NIPS 2014); Ghahramani, Z.; Welling, M.; Cortes, C.; Lawrence, N.; Weinberger, K. Q., Eds.; 2014.Google ScholarThere is no corresponding record for this reference.
- 22Karras, T.; Aila, T.; Laine, S.; Lehtinen, J. Progressive growing of gans for improved quality, stability, and variation. arXiv preprint , 2017, arXiv:1710.10196.Google ScholarThere is no corresponding record for this reference.
- 23Isola, P.; Zhu, J.-Y.; Zhou, T.; Efros, A. A. Image-to-image translation with conditional adversarial networks. In Proceedings of the IEEE Conference on Computer Vision and Pattern Recognition , 2017; pp 1125– 1134.Google ScholarThere is no corresponding record for this reference.
- 24Zhu, J.-Y.; Park, T.; Isola, P.; Efros, A. A. Unpaired image-to-image translation using cycle-consistent adversarial networks. In Proceedings of the IEEE International Conference on Computer Vision , 2017; pp 2223– 2232.Google ScholarThere is no corresponding record for this reference.
- 25Karras, T.; Laine, S.; Aila, T. A style-based generator architecture for generative adversarial networks. Proceedings of the IEEE/CVF Conference on Computer Vision and Pattern Recognition , 2019; pp 4401– 4410.Google ScholarThere is no corresponding record for this reference.
- 26Jangid, D. K.; Brodnik, N. R.; Khan, A.; Goebel, M. G.; Echlin, M. P.; Pollock, T. M.; Daly, S. H.; Manjunath, B. 3D Grain Shape Generation in Polycrystals Using Generative Adversarial Networks. Integr. Mater. Manuf. Innov. 2022, 11, 71– 84, DOI: 10.1007/s40192-021-00244-1Google ScholarThere is no corresponding record for this reference.
- 27Kadurin, A.; Nikolenko, S.; Khrabrov, K.; Aliper, A.; Zhavoronkov, A. druGAN: an advanced generative adversarial autoencoder model for de novo generation of new molecules with desired molecular properties in silico. Mol. Pharmaceutics 2017, 14, 3098– 3104, DOI: 10.1021/acs.molpharmaceut.7b00346Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFGitbbF&md5=6fc20afd4c6a8188e830aa90a7875dd6druGAN: An Advanced Generative Adversarial Autoencoder Model for de Novo Generation of New Molecules with Desired Molecular Properties in SilicoKadurin, Artur; Nikolenko, Sergey; Khrabrov, Kuzma; Aliper, Alex; Zhavoronkov, AlexMolecular Pharmaceutics (2017), 14 (9), 3098-3104CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Deep generative adversarial networks (GANs) are the emerging technol. in drug discovery and biomarker development. In our recent work, we demonstrated a proof-of-concept of implementing deep generative adversarial autoencoder (AAE) to identify new mol. fingerprints with predefined anticancer properties. Another popular generative model is the variational autoencoder (VAE), which is based on deep neural architectures. In this work, we developed an advanced AAE model for mol. feature extn. problems, and demonstrated its advantages compared to VAE in terms of (a) adjustability in generating mol. fingerprints; (b) capacity of processing very large mol. data sets; and (c) efficiency in unsupervised pretraining for regression model. Our results suggest that the proposed AAE model significantly enhances the capacity and efficiency of development of the new mols. with specific anticancer properties using the deep generative models.
- 28Vanhaelen, Q.; Lin, Y.-C.; Zhavoronkov, A. The advent of generative chemistry. ACS Med. Chem. Lett. 2020, 11, 1496– 1505, DOI: 10.1021/acsmedchemlett.0c00088Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtl2jtbfF&md5=847e493bb1da64aeae859a40b031d9c2The Advent of Generative ChemistryVanhaelen, Quentin; Lin, Yen-Chu; Zhavoronkov, AlexACS Medicinal Chemistry Letters (2020), 11 (8), 1496-1505CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)A review. Generative adversarial networks (GANs), first published in 2014, are among the most important concepts in modern artificial intelligence (AI). Bridging deep learning and game theory, GANs are used to generate or "imagine" new objects with desired properties. Since 2016, multiple GANs with reinforcement learning (RL) have been successfully applied in pharmacol. for de novo mol. design. Those techniques aim at a more efficient use of the data and a better exploration of the chem. space. We review recent advances for the generation of novel mols. with desired properties with a focus on the applications of GANs, RL, and related techniques. We also discuss the current limitations and challenges in the new growing field of generative chem.
- 29Xu, M.; Cheng, J.; Liu, Y.; Huang, W. DeepGAN: Generating Molecule for Drug Discovery Based on Generative Adversarial Network. In 2021 IEEE Symposium on Computers and Communications (ISCC) , 2021; pp 1– 6.Google ScholarThere is no corresponding record for this reference.
- 30Guimaraes, G. L.; Sanchez-Lengeling, B.; Outeiral, C.; Farias, P. L. C.; Aspuru-Guzik, A. Objective-reinforced generative adversarial networks (ORGAN) for sequence generation models. arXiv preprint , arXiv:1705.10843, 2017.Google ScholarThere is no corresponding record for this reference.
- 31Yu, L.; Zhang, W.; Wang, J.; Yu, Y. SeqGAN: sequence generative adversarial nets with policy gradient. In Proceedings of the Thirty-First AAAI Conference on Artificial Intelligence , 2017; pp 2852– 2858.Google ScholarThere is no corresponding record for this reference.
- 32Prykhodko, O.; Johansson, S. V.; Kotsias, P.-C.; Arús-Pous, J.; Bjerrum, E. J.; Engkvist, O.; Chen, H. A de novo molecular generation method using latent vector based generative adversarial network. J. Cheminf. 2019, 11, 1– 13, DOI: 10.1186/s13321-019-0397-9Google ScholarThere is no corresponding record for this reference.
- 33Kotsias, P.-C.; Arús-Pous, J.; Chen, H.; Engkvist, O.; Tyrchan, C.; Bjerrum, E. J. Direct steering of de novo molecular generation with descriptor conditional recurrent neural networks. Nat. Mach. Intell. 2020, 2, 254– 265, DOI: 10.1038/s42256-020-0174-5Google ScholarThere is no corresponding record for this reference.
- 34De Cao, N.; Kipf, T. MolGAN: An implicit generative model for small molecular graphs. arXiv preprint arXiv:1805.11973 2018,.Google ScholarThere is no corresponding record for this reference.
- 35Neukart, F.; Compostella, G.; Seidel, C.; Von Dollen, D.; Yarkoni, S.; Parney, B. Traffic flow optimization using a quantum annealer. Front. ICT 2017, 4, 29, DOI: 10.3389/fict.2017.00029Google ScholarThere is no corresponding record for this reference.
- 36Harwood, S.; Gambella, C.; Trenev, D.; Simonetto, A.; Bernal Neira, D.; Greenberg, D. Formulating and solving routing problems on quantum computers. IEEE Trans. Quantum Eng. 2021, 2, 1– 17, DOI: 10.1109/TQE.2021.3049230Google ScholarThere is no corresponding record for this reference.
- 37Orus, R.; Mugel, S.; Lizaso, E. Quantum computing for finance: Overview and prospects. Rev. Phys. 2019, 4, 100028, DOI: 10.1016/j.revip.2019.100028Google ScholarThere is no corresponding record for this reference.
- 38Liu, G.; Ma, W. A quantum artificial neural network for stock closing price prediction. Inf. Sci. (N.Y.) 2022, 598, 75– 85, DOI: 10.1016/j.ins.2022.03.064Google ScholarThere is no corresponding record for this reference.
- 39Du, Y.; Yang, Y.; Tao, D.; Hsieh, M.-H. Demystify Problem-Dependent Power of Quantum Neural Networks on Multi-Class Classification. arXiv preprint , arXiv:2301.01597, 2022.Google ScholarThere is no corresponding record for this reference.
- 40Yin, X.-F.; Du, Y.; Fei, Y.-Y.; Zhang, R.; Liu, L.-Z.; Mao, Y.; Liu, T.; Hsieh, M.-H.; Li, L.; Liu, N.-L.; Tao, D.; Chen, Y.-A.; Pan, J.-W. Efficient Bipartite Entanglement Detection Scheme with a Quantum Adversarial Solver. Phys. Rev. Lett. 2022, 128, 110501, DOI: 10.1103/PhysRevLett.128.110501Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XpsFKgu74%253D&md5=d13b175a37f194b174270568ff59afc4Efficient Bipartite Entanglement Detection Scheme with a Quantum Adversarial SolverYin, Xu-Fei; Du, Yuxuan; Fei, Yue-Yang; Zhang, Rui; Liu, Li-Zheng; Mao, Yingqiu; Liu, Tongliang; Hsieh, Min-Hsiu; Li, Li; Liu, Nai-Le; Tao, Dacheng; Chen, Yu-Ao; Pan, Jian-WeiPhysical Review Letters (2022), 128 (11), 110501CODEN: PRLTAO; ISSN:1079-7114. (American Physical Society)The recognition of entanglement states is a notoriously difficult problem when no prior information is available. Here, we propose an efficient quantum adversarial bipartite entanglement detection scheme to address this issue. Our proposal reformulates the bipartite entanglement detection as a two-player zero-sum game completed by parameterized quantum circuits, where a two-outcome measurement can be used to query a classical binary result about whether the input state is bipartite entangled or not. In principle, for an N-qubit quantum state, the runtime complexity of our proposal is O(poly(N)T) with T being the no. of iterations. We exptl. implement our protocol on a linear optical network and exhibit its effectiveness to accomplish the bipartite entanglement detection for 5-qubit quantum pure states and 2-qubit quantum mixed states. Our work paves the way for using near-term quantum machines to tackle entanglement detection on multipartite entangled quantum systems.
- 41Rudolph, M. S.; Toussaint, N. B.; Katabarwa, A.; Johri, S.; Peropadre, B.; Perdomo-Ortiz, A. Generation of high-resolution handwritten digits with an ion-trap quantum computer. Phys. Rev. X 2022, 12, 031010, DOI: 10.1103/PhysRevX.12.031010Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1ymurzM&md5=2c6059f94ef9b16a6ea891ad14559e4aGeneration of High-Resolution Handwritten Digits with an Ion-Trap Quantum ComputerRudolph, Manuel S.; Toussaint, Ntwali Bashige; Katabarwa, Amara; Johri, Sonika; Peropadre, Borja; Perdomo-Ortiz, AlejandroPhysical Review X (2022), 12 (3), 031010CODEN: PRXHAE; ISSN:2160-3308. (American Physical Society)Generating high-quality data (e.g., images or video) is one of the most exciting and challenging frontiers in unsupervised machine learning. Utilizing quantum computers in such tasks to potentially enhance conventional machine-learning algorithms has emerged as a promising application but poses big challenges due to the limited no. of qubits and the level of gate noise in available devices. In this work, we provide the first practical and exptl. implementation of a quantum-classical generative algorithm capable of generating high-resoln. images of handwritten digits with state-of-the-art gate-based quantum computers. In our quantum-assisted machine-learning framework, we implement a quantum-circuit-based generative model to learn and sample the prior distribution of a generative adversarial network. We introduce a multibasis technique which leverages the unique possibility of measuring quantum states in different bases, hence enhancing the expressivity of the prior distribution. We train this hybrid algorithm on an ion-trap device based on Yb+171 ion qubits to generate high-quality images and quant. outperform comparable classical generative adversarial networks trained on the popular MNIST dataset for handwritten digits.
- 42Aspuru-Guzik, A.; Dutoi, A. D.; Love, P. J.; Head-Gordon, M. Simulated quantum computation of molecular energies. Science 2005, 309, 1704– 1707, DOI: 10.1126/science.1113479Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpvFCisrg%253D&md5=c11f20a70de762c9a4b933e4a35d0af3Simulated Quantum Computation of Molecular EnergiesAspuru-Guzik, Alan; Dutoi, Anthony D.; Love, Peter J.; Head-Gordon, MartinScience (Washington, DC, United States) (2005), 309 (5741), 1704-1707CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The calcn. time for the energy of atoms and mols. scales exponentially with system size on a classical computer but polynomially using quantum algorithms. We demonstrate that such algorithms can be applied to problems of chem. interest using modest nos. of quantum bits. Calcns. of the water and lithium hydride mol. ground-state energies have been carried out on a quantum computer simulator using a recursive phase-estn. algorithm. The recursive algorithm reduces the no. of quantum bits required for the readout register from about 20 to 4. Mappings of the mol. wave function to the quantum bits are described. An adiabatic method for the prepn. of a good approx. ground-state wave function is described and demonstrated for a stretched hydrogen mol. The no. of quantum bits required scales linearly with the no. of basis functions, and the no. of gates required grows polynomially with the no. of quantum bits.
- 43Lanyon, B. P.; Whitfield, J. D.; Gillett, G. G.; Goggin, M. E.; Almeida, M. P.; Kassal, I.; Biamonte, J. D.; Mohseni, M.; Powell, B. J.; Barbieri, M.; Aspuru-Guzik, A.; White, A. G. Towards quantum chemistry on a quantum computer. Nat. Chem. 2010, 2, 106– 111, DOI: 10.1038/nchem.483Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXos1Crug%253D%253D&md5=3434a0ba7b2b832ac19cd0e792e7af2fTowards quantum chemistry on a quantum computerLanyon, B. P.; Whitfield, J. D.; Gillett, G. G.; Goggin, M. E.; Almeida, M. P.; Kassal, I.; Biamonte, J. D.; Mohseni, M.; Powell, B. J.; Barbieri, M.; Aspuru-Guzik, A.; White, A. G.Nature Chemistry (2010), 2 (2), 106-111CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Exact first-principles calcns. of mol. properties are currently intractable because their computational cost grows exponentially with both the no. of atoms and basis set size. A soln. is to move to a radically different model of computing by building a quantum computer, which is a device that uses quantum systems themselves to store and process data. Here we report the application of the latest photonic quantum computer technol. to calc. properties of the smallest mol. system: the mol. in a minimal basis. We calc. the complete energy spectrum to 20 bits of precision and discuss how the technique can be expanded to solve large-scale chem. problems that lie beyond the reach of modern supercomputers. These results represent an early practical step toward a powerful tool with a broad range of quantum-chem. applications.
- 44Peruzzo, A.; McClean, J.; Shadbolt, P.; Yung, M.-H.; Zhou, X.-Q.; Love, P. J.; Aspuru-Guzik, A.; O’brien, J. L. A variational eigenvalue solver on a photonic quantum processor. Nat. Commun. 2014, 5, 1– 7, DOI: 10.1038/ncomms5213Google ScholarThere is no corresponding record for this reference.
- 45O’Malley, P. J. J.; Babbush, R.; Kivlichan, I. D.; Romero, J.; McClean, J. R.; Barends, R.; Kelly, J.; Roushan, P.; Tranter, A.; Ding, N.; Campbell, B.; Chen, Y.; Chen, Z.; Chiaro, B.; Dunsworth, A.; Fowler, A. G.; Jeffrey, E.; Lucero, E.; Megrant, A.; Mutus, J. Y.; Neeley, M.; Neill, C.; Quintana, C.; Sank, D.; Vainsencher, A.; Wenner, J.; White, T. C.; Coveney, P. V.; Love, P. J.; Neven, H.; Aspuru-Guzik, A.; Martinis, J. M. Scalable quantum simulation of molecular energies. Phys. Rev. X 2016, 6, 031007, DOI: 10.1103/PhysRevX.6.031007Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslyrtbvF&md5=750ce9fcb52cba1233a0b592d09142e3Scalable quantum simulation of molecular energiesO'Malley, P. J. J.; Babbush, R.; Kivlichan, I. D.; Romero, J.; McClean, J. R.; Barends, R.; Kelly, J.; Roushan, P.; Tranter, A.; Ding, N.; Campbell, B.; Chen, Y.; Chen, Z.; Chiaro, B.; Dunsworth, A.; Fowler, A. G.; Jeffrey, E.; Lucero, E.; Megrant, A.; Mutus, J. Y.; Neeley, M.; Neill, C.; Quintana, C.; Sank, D.; Vainsencher, A.; Wenner, J.; White, T. C.; Coveney, P. V.; Love, P. J.; Neven, H.; Aspuru-Guzik, A.; Martinis, J. M.Physical Review X (2016), 6 (3), 031007/1-031007/13CODEN: PRXHAE; ISSN:2160-3308. (American Physical Society)We report the first electronic structure calcn. performed on a quantum computer without exponentially costly precompilation. We use a programmable array of superconducting qubits to compute the energy surface of mol. hydrogen using two distinct quantum algorithms. First, we exptl. execute the unitary coupled cluster method using the variational quantum eigensolver. Our efficient implementation predicts the correct dissocn. energy to within chem. accuracy of the numerically exact result. Second, we exptl. demonstrate the canonical quantum algorithm for chem., which consists of Trotterization and quantum phase estn. We compare the exptl. performance of these approaches to show clear evidence that the variational quantum eigensolver is robust to certain errors. This error tolerance inspires hope that variational quantum simulations of classically intractable mols. may be viable in the near future.
- 46Kandala, A.; Mezzacapo, A.; Temme, K.; Takita, M.; Brink, M.; Chow, J. M.; Gambetta, J. M. Hardware-efficient variational quantum eigensolver for small molecules and quantum magnets. Nature 2017, 549, 242– 246, DOI: 10.1038/nature23879Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsV2isLjP&md5=4315a0cb0cda7fb07584cacd895b576fHardware-efficient variational quantum eigensolver for small molecules and quantum magnetsKandala, Abhinav; Mezzacapo, Antonio; Temme, Kristan; Takita, Maika; Brink, Markus; Chow, Jerry M.; Gambetta, Jay M.Nature (London, United Kingdom) (2017), 549 (7671), 242-246CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Quantum computers can be used to address electronic-structure problems and problems in materials science and condensed matter physics that can be formulated as interacting fermionic problems, problems which stretch the limits of existing high-performance computers. Finding exact solns. to such problems numerically has a computational cost that scales exponentially with the size of the system, and Monte Carlo methods are unsuitable owing to the fermionic sign problem. These limitations of classical computational methods have made solving even few-atom electronic-structure problems interesting for implementation using medium-sized quantum computers. Yet exptl. implementations have so far been restricted to mols. involving only hydrogen and helium. Here we demonstrate the exptl. optimization of Hamiltonian problems with up to six qubits and more than one hundred Pauli terms, detg. the ground-state energy for mols. of increasing size, up to BeH2. We achieve this result by using a variational quantum eigenvalue solver (eigensolver) with efficiently prepd. trial states that are tailored specifically to the interactions that are available in our quantum processor, combined with a compact encoding of fermionic Hamiltonians and a robust stochastic optimization routine. We demonstrate the flexibility of our approach by applying it to a problem of quantum magnetism, an antiferromagnetic Heisenberg model in an external magnetic field. In all cases, we find agreement between our expts. and numerical simulations using a model of the device with noise. Our results help to elucidate the requirements for scaling the method to larger systems and for bridging the gap between key problems in high-performance computing and their implementation on quantum hardware.
- 47Cao, Y.; Romero, J.; Olson, J. P.; Degroote, M.; Johnson, P. D.; Kieferová, M.; Kivlichan, I. D.; Menke, T.; Peropadre, B.; Sawaya, N. P. D.; Sim, S.; Veis, L.; Aspuru-Guzik, A. Quantum Chemistry in the Age of Quantum Computing. Chem. Rev. 2019, 119, 10856– 10915, DOI: 10.1021/acs.chemrev.8b00803Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1Krtb7K&md5=42620699778f2ef25d3f6958b8d4e776Quantum Chemistry in the Age of Quantum ComputingCao, Yudong; Romero, Jonathan; Olson, Jonathan P.; Degroote, Matthias; Johnson, Peter D.; Kieferova, Maria; Kivlichan, Ian D.; Menke, Tim; Peropadre, Borja; Sawaya, Nicolas P. D.; Sim, Sukin; Veis, Libor; Aspuru-Guzik, AlanChemical Reviews (Washington, DC, United States) (2019), 119 (19), 10856-10915CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Practical challenges in simulating quantum systems on classical computers have been widely recognized in the quantum physics and quantum chem. communities over the past century. Although many approxn. methods have been introduced, the complexity of quantum mechanics remains hard to appease. The advent of quantum computation brings new pathways to navigate this challenging complexity landscape. By manipulating quantum states of matter and taking advantage of their unique features such as superposition and entanglement, quantum computers promise to efficiently deliver accurate results for many important problems in quantum chem. such as the electronic structure of mols. In the past two decades significant advances have been made in developing algorithms and phys. hardware for quantum computing, heralding a revolution in simulation of quantum systems. This article is an overview of the algorithms and results that are relevant for quantum chem. The intended audience is both quantum chemists who seek to learn more about quantum computing, and quantum computing researchers who would like to explore applications in quantum chem.
- 48Quantum, G. A.; Collaborators*†; Arute, F.; Arya, K.; Babbush, R.; Bacon, D.; Bardin, J. C.; Barends, R.; Boixo, S.; Broughton, M. Hartree-Fock on a superconducting qubit quantum computer. Science 2020, 369, 1084– 1089, DOI: 10.1126/science.abb9811Google ScholarThere is no corresponding record for this reference.
- 49Gao, Q.; Nakamura, H.; Gujarati, T. P.; Jones, G. O.; Rice, J. E.; Wood, S. P.; Pistoia, M.; Garcia, J. M.; Yamamoto, N. Computational investigations of the lithium superoxide dimer rearrangement on noisy quantum devices. J. Phys. Chem. A 2021, 125, 1827– 1836, DOI: 10.1021/acs.jpca.0c09530Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXlt1ShsbY%253D&md5=233ec03e41ecf3978d94c885afcf1e1cComputational Investigations of the Lithium Superoxide Dimer Rearrangement on Noisy Quantum DevicesGao, Qi; Nakamura, Hajime; Gujarati, Tanvi P.; Jones, Gavin O.; Rice, Julia E.; Wood, Stephen P.; Pistoia, Marco; Garcia, Jeannette M.; Yamamoto, NaokiJournal of Physical Chemistry A (2021), 125 (9), 1827-1836CODEN: JPCAFH; ISSN:1089-5639. (American Chemical Society)Quantum chem. studies of biradical systems are challenging due to the required multiconfigurational nature of the wavefunction. In this work, Variational Quantum Eigensolver (VQE) is used to compute the energy profile for the lithium superoxide dimer rearrangement, involving biradical species, on quantum simulators and devices. Considering that current quantum devices can only handle limited no. of qubits, we present guidelines for selecting an appropriate active space to perform computations on chem. systems that require many qubits. We show that with VQE performed with a quantum simulator reproduces results obtained with full-CI (Full CI) for the chosen active space. However, results deviate from exact values by about 39 mHa for calcns. on a quantum device. This deviation can be improved to about 4 mHa using the readout mitigation approach and can be further improved to 2 mHa, approaching chem. accuracy, using the state tomog. technique to purify the calcd. quantum state.
- 50Shee, Y.; Yeh, T.-L.; Hsiao, J.-Y.; Yang, A.; Lin, Y.-C.; Hsieh, M.-H. Quantum Simulation of Preferred Tautomeric State Prediction. arXiv preprint , arXiv:2210.02977, 2022.Google ScholarThere is no corresponding record for this reference.
- 51Cao, Y.; Romero, J.; Aspuru-Guzik, A. Potential of quantum computing for drug discovery. IBM J. Res. Dev. 2018, 62, 1– 20, DOI: 10.1147/JRD.2018.2888987Google ScholarThere is no corresponding record for this reference.
- 52Li, J.; Topaloglu, R. O.; Ghosh, S. Quantum generative models for small molecule drug discovery. IEEE Trans. Quantum Eng. 2021, 2, 1– 8, DOI: 10.1109/TQE.2021.3104804Google ScholarThere is no corresponding record for this reference.
- 53Bharti, K.; Cervera-Lierta, A.; Kyaw, T. H.; Haug, T.; Alperin-Lea, S.; Anand, A.; Degroote, M.; Heimonen, H.; Kottmann, J. S.; Menke, T.; Mok, W.-K.; Sim, S.; Kwek, L.-C.; Aspuru-Guzik, A. Noisy intermediate-scale quantum algorithms. Rev. Mod. Phys. 2022, 94, 015004, DOI: 10.1103/RevModPhys.94.015004Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtVOnsLrE&md5=1d31a740165d58c2a52b3baa15f9f9a1Noisy intermediate-scale quantum algorithmsBharti, Kishor; Cervera-Lierta, Alba; Kyaw, Thi Ha; Haug, Tobias; Alperin-Lea, Sumner; Anand, Abhinav; Degroote, Matthias; Heimonen, Hermanni; Kottmann, Jakob S.; Menke, Tim; Mok, Wai-Keong; Sim, Sukin; Kwek, Leong-Chuan; Aspuru-Guzik, AlanReviews of Modern Physics (2022), 94 (1), 015004CODEN: RMPHAT; ISSN:1539-0756. (American Physical Society)A review. A universal fault-tolerant quantum computer that can efficiently solve problems such as integer factorization and unstructured database search requires millions of qubits with low error rates and long coherence times. While the exptl. advancement toward realizing such devices will potentially take decades of research, noisy intermediate-scale quantum (NISQ) computers already exist. These computers are composed of hundreds of noisy qubits, i.e., qubits that are not error cor., and therefore perform imperfect operations within a limited coherence time. In the search for achieving quantum advantage with these devices, algorithms have been proposed for applications in various disciplines spanning physics, machine learning, quantum chem., and combinatorial optimization. The overarching goal of such algorithms is to leverage the limited available resources to perform classically challenging tasks. In this review, a thorough summary of NISQ computational paradigms and algorithms is provided. The key structure of these algorithms and their limitations and advantages are discussed. A comprehensive overview of various benchmarking and software tools useful for programming and testing NISQ devices is addnl. provided.
- 54Du, Y.; Hsieh, M.-H.; Liu, T.; Tao, D. Expressive power of parametrized quantum circuits. Phys. Rev. Res. 2020, 2, 033125, DOI: 10.1103/PhysRevResearch.2.033125Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitV2gs7nE&md5=288c5ca7b2455ce8389ffe97a441b449Expressive power of parametrized quantum circuitsDu, Yuxuan; Hsieh, Min-Hsiu; Liu, Tongliang; Tao, DachengPhysical Review Research (2020), 2 (3), 033125CODEN: PRRHAI; ISSN:2643-1564. (American Physical Society)Parametrized quantum circuits (PQCs) have been broadly used as a hybrid quantum-classical machine learning scheme to accomplish generative tasks. However, whether PQCs have better expressive power than classical generative neural networks, such as restricted or deep Boltzmann machines, remains an open issue. In this paper, we prove that PQCs with a simple structure already outperform any classical neural network for generative tasks, unless the polynomial hierarchy collapses. Our proof builds on known results from tensor networks and quantum circuits (in particular, instantaneous quantum polynomial circuits). In addn., PQCs equipped with ancillary qubits for postselection may possess expressive power stronger than that of those without postselection. We employ them as an application for Bayesian learning, since it is possible to learn prior probabilities rather than assuming they are known. We expect that it will find many more applications in semisupervised learning where prior distributions are normally assumed to be unknown. Lastly, we conduct several numerical expts. using the Rigetti Forest platform to demonstrate the performance of the proposed Bayesian quantum circuit.
- 55Du, Y.; Hsieh, M.-H.; Liu, T.; You, S.; Tao, D. Learnability of quantum neural networks. PRX Quantum 2021, 2, 040337, DOI: 10.1103/PRXQuantum.2.040337Google ScholarThere is no corresponding record for this reference.
- 56Du, Y.; Hsieh, M.-H.; Liu, T.; Tao, D.; Liu, N. Quantum noise protects quantum classifiers against adversaries. Phys. Rev. Res. 2021, 3, 023153, DOI: 10.1103/PhysRevResearch.3.023153Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFSgtbvI&md5=56ed26f0c7b485611e974308cddcbee8Quantum noise protects quantum classifiers against adversariesDu, Yuxuan; Hsieh, Min-Hsiu; Liu, Tongliang; Tao, Dacheng; Liu, NanaPhysical Review Research (2021), 3 (2), 023153CODEN: PRRHAI; ISSN:2643-1564. (American Physical Society)Noise in quantum information processing is often viewed as a disruptive and difficult-to-avoid feature, esp. in near-term quantum technologies. However, noise has often played beneficial roles, from enhancing weak signals in stochastic resonance to protecting the privacy of data in differential privacy. It is then natural to ask: Can we harness the power of quantum noise that is beneficial to quantum computing. An important current direction for quantum computing is its application to machine learning, such as classification problems. One outstanding problem in machine learning for classification is its sensitivity to adversarial examples. These are small, undetectable perturbations from the original data where the perturbed data is completely misclassified in otherwise extremely accurate classifiers. They can also be considered as worst-case perturbations by unknown noise sources. We show that by taking advantage of depolarization noise in quantum circuits for classification, a robustness bound against adversaries can be derived where the robustness improves with increasing noise. This robustness property is intimately connected with an important security concept called differential privacy, which can be extended to quantum differential privacy. For the protection of quantum data, this quantum protocol can be used against the most general adversaries. Furthermore, we show how the robustness in the classical case can be sensitive to the details of the classification model, but in the quantum case the details of the classification model are absent, thus also providing a potential quantum advantage for classical data. This opens the opportunity to explore other ways in which quantum noise can be used in our favor, as well as identifying other ways quantum algorithms can be helpful in a way which is distinct from quantum speedups.
- 57Lloyd, S.; Weedbrook, C. Quantum generative adversarial learning. Phys. Rev. Lett. 2018, 121, 040502, DOI: 10.1103/PhysRevLett.121.040502Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXltFSjs7g%253D&md5=c7b5965318a3af921f7e97e9e243cbcaQuantum Generative Adversarial LearningLloyd, Seth; Weedbrook, ChristianPhysical Review Letters (2018), 121 (4), 040502CODEN: PRLTAO; ISSN:1079-7114. (American Physical Society)Generative adversarial networks represent a powerful tool for classical machine learning: a generator tries to create statistics for data that mimics those of a true data set, while a discriminator tries to discriminate between the true and fake data. The learning process for generator and discriminator can be thought of as an adversarial game, and under reasonable assumptions, the game converges to the point where the generator generates the same statistics as the true data and the discriminator is unable to discriminate between the true and the generated data. This Letter introduces the notion of quantum generative adversarial networks, where the data consist either of quantum states or of classical data, and the generator and discriminator are equipped with quantum information processors. We show that the unique fixed point of the quantum adversarial game also occurs when the generator produces the same statistics as the data. Neither the generator nor the discriminator perform quantum tomog.; linear programing drives them to the optimal. Since quantum systems are intrinsically probabilistic, the proof of the quantum case is different from-and simpler than-the classical case. We show that, when the data consist of samples of measurements made on high-dimensional spaces, quantum adversarial networks may exhibit an exponential advantage over classical adversarial networks.
- 58Huang, H.-L.; Du, Y.; Gong, M.; Zhao, Y.; Wu, Y.; Wang, C.; Li, S.; Liang, F.; Lin, J.; Xu, Y.; Yang, R.; Liu, T.; Hsieh, M.-H.; Deng, H.; Rong, H.; Peng, C.-Z.; Lu, C.-Y.; Chen, Y.-A.; Tao, D.; Zhu, X.; Pan, J.-W. Experimental quantum generative adversarial networks for image generation. Phys. Rev. Appl. 2021, 16, 024051, DOI: 10.1103/PhysRevApplied.16.024051Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVekurvF&md5=f8aa2cb8ed6846dc955f570486b24cdaExperimental Quantum Generative Adversarial Networks for Image GenerationHuang, He-Liang; Du, Yuxuan; Gong, Ming; Zhao, Youwei; Wu, Yulin; Wang, Chaoyue; Li, Shaowei; Liang, Futian; Lin, Jin; Xu, Yu; Yang, Rui; Liu, Tongliang; Hsieh, Min-Hsiu; Deng, Hui; Rong, Hao; Peng, Cheng-Zhi; Lu, Chao-Yang; Chen, Yu-Ao; Tao, Dacheng; Zhu, Xiaobo; Pan, Jian-WeiPhysical Review Applied (2021), 16 (2), 024051CODEN: PRAHB2; ISSN:2331-7019. (American Physical Society)Quantum machine learning is expected to be one of the first practical applications of near-term quantum devices. Pioneer theor. works suggest that quantum generative adversarial networks (GANs) may exhibit a potential exponential advantage over classical GANs, thus attracting widespread attention. However, it remains elusive whether quantum GANs implemented on near-term quantum devices can actually solve real-world learning tasks. Here, we devise a flexible quantum GAN scheme to narrow this knowledge gap. In principle, this scheme has the ability to complete image generation with high-dimensional features and could harness quantum superposition to train multiple examples in parallel. We exptl. achieve the learning and generating of real-world handwritten digit images on a superconducting quantum processor. Moreover, we utilize a gray-scale bar dataset to exhibit competitive performance between quantum GANs and the classical GANs based on multilayer perceptron and convolutional neural network architectures, resp., benchmarked by the Fr´echet distance score. Our work provides guidance for developing advanced quantum generative models on near-term quantum devices and opens up an avenue for exploring quantum advantages in various GAN-related learning tasks.
- 59Dallaire-Demers, P.-L.; Killoran, N. Quantum generative adversarial networks. Phys. Rev. A 2018, 98, 012324, DOI: 10.1103/PhysRevA.98.012324Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXlsV2ht7Y%253D&md5=9c61c6048cbf7251849f2fb0d689c3bbQuantum generative adversarial networksDallaire-Demers, Pierre-Luc; Killoran, NathanPhysical Review A (2018), 98 (1), 012324CODEN: PRAHC3; ISSN:2469-9934. (American Physical Society)Quantum machine learning is expected to be one of the first potential general-purpose applications of near-term quantum devices. A major recent breakthrough in classical machine learning is the notion of generative adversarial training, where the gradients of a discriminator model are used to train a sep. generative model. In this work and a companion paper, we extend adversarial training to the quantum domain and show how to construct generative adversarial networks using quantum circuits. Furthermore, we also show how to compute gradients-a key element in generative adversarial network training-using another quantum circuit. We give an example of a simple practical circuit ansatz to parametrize quantum machine learning models and perform a simple numerical expt. to demonstrate that quantum generative adversarial networks can be trained successfully.
- 60Romero, J.; Aspuru-Guzik, A. Variational quantum generators: Generative adversarial quantum machine learning for continuous distributions. Adv. Quantum Technol. 2021, 4, 2000003, DOI: 10.1002/qute.202000003Google ScholarThere is no corresponding record for this reference.
- 61Li, J. Quantum GAN with Hybrid Generator. https://github.com/jundeli/quantum-gan, accessed Aug. 2, 2021.Google ScholarThere is no corresponding record for this reference.
- 62Ivanenkov, Y. A.; Polykovskiy, D.; Bezrukov, D.; Zagribelnyy, B.; Aladinskiy, V.; Kamya, P.; Aliper, A.; Ren, F.; Zhavoronkov, A. Chemistry42: An AI-Driven Platform for Molecular Design and Optimization. J. Chem. Inf. Model. 2023, 63, 695– 701, DOI: 10.1021/acs.jcim.2c01191Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXit1yjsrc%253D&md5=04066dad851fb49f198d9c7e4007fc02Chemistry42: An AI-Driven Platform for Molecular Design and OptimizationIvanenkov, Yan A.; Polykovskiy, Daniil; Bezrukov, Dmitry; Zagribelnyy, Bogdan; Aladinskiy, Vladimir; Kamya, Petrina; Aliper, Alex; Ren, Feng; Zhavoronkov, AlexJournal of Chemical Information and Modeling (2023), 63 (3), 695-701CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Chem.42 is a software platform for de novo small mol. design and optimization that integrates Artificial Intelligence (AI) techniques with computational and medicinal chem. methodologies. Chem.42 efficiently generates novel mol. structures with optimized properties validated in both in vitro and in vivo studies and is available through licensing or collaboration. Chem.42 is the core component of Insilico Medicine's Pharma.ai drug discovery suite. Pharma.ai also includes PandaOmics for target discovery and multiomics data anal., and inClinico-a data-driven multimodal forecast of a clin. trial's probability of success (PoS). In this paper, we demonstrate how the platform can be used to efficiently find novel mol. structures against DDR1 and CDK20.
- 63Bergholm, V.; Izaac, J.; Schuld, M.; Gogolin, C.; Ahmed, S.; Ajith, V.; Alam, M. S.; Alonso-Linaje, G.; AkashNarayanan, B.; Asadi, A.; Arrazola, J. M.; Azad, U.; Banning, S.; Blank, C.; Bromley, T. R.; Cordier, B. A.; Ceroni, J.; Delgado, A.; Di Matteo, O.; Dusko, A.; Garg, T.; Guala, D.; Hayes, A.; Hill, R.; Ijaz, A.; Isacsson, T.; Ittah, D.; Jahangiri, S.; Jain, P.; Jiang, E.; Khandelwal, A.; Kottmann, K.; Lang, R. A.; Lee, C.; Loke, T.; Lowe, A.; McKiernan, K.; Meyer, J. J.; Montañez-Barrera, J. A.; Moyard, R.; Niu, Z.; O’Riordan, L. J.; Oud, S.; Panigrahi, A.; Park, C.-Y.; Polatajko, D.; Quesada, N.; Roberts, C.; Sá, N.; Schoch, I.; Shi, B.; Shu, S.; Sim, S.; Singh, A.; Strandberg, I.; Soni, J.; Száva, A.; Thabet, S.; Vargas-Hernández, R. A.; Vincent, T.; Vitucci, N.; Weber, M.; Wierichs, D.; Wiersema, R.; Willmann, M.; Wong, V.; Zhang, S.; Killoran, N. Pennylane: Automatic differentiation of hybrid quantum-classical computations. arXiv preprint , arXiv:1811.04968, 2018.Google ScholarThere is no corresponding record for this reference.
- 64Paszke, A.; Gross, S.; Chintala, S.; Chanan, G.; Yang, E.; DeVito, Z.; Lin, Z.; Desmaison, A.; Antiga, L.; Lerer, A. Automatic differentiation in pytorch. Presented at the NIPS 2017 Autodiff Workshop , 2017.Google ScholarThere is no corresponding record for this reference.
- 65Arjovsky, M.; Chintala, S.; Bottou, L. Wasserstein generative adversarial networks. In International Conference on Machine Learning , 2017; pp 214 223.Google ScholarThere is no corresponding record for this reference.
- 66Brown, N.; Fiscato, M.; Segler, M. H.; Vaucher, A. C. GuacaMol: benchmarking models for de novo molecular design. J. Chem. Inf. Model. 2019, 59, 1096– 1108, DOI: 10.1021/acs.jcim.8b00839Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXltVWrsbY%253D&md5=d3fb616b81a4b146cf77950a1c92e4d1GuacaMol: Benchmarking Models for de Novo Molecular DesignBrown, Nathan; Fiscato, Marco; Segler, Marwin H. S.; Vaucher, Alain C.Journal of Chemical Information and Modeling (2019), 59 (3), 1096-1108CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)De novo design seeks to generate mols. with required property profiles by virtual design-make-test cycles. With the emergence of deep learning and neural generative models in many application areas, models for mol. design based on neural networks appeared recently and show promising results. However, the new models have not been profiled on consistent tasks, and comparative studies to well-established algorithms have only seldom been performed. To standardize the assessment of both classical and neural models for de novo mol. design, we propose an evaluation framework, GuacaMol, based on a suite of standardized benchmarks. The benchmark tasks encompass measuring the fidelity of the models to reproduce the property distribution of the training sets, the ability to generate novel mols., the exploration and exploitation of chem. space, and a variety of single and multiobjective optimization tasks. The benchmarking open-source Python code and a leaderboard can be found on https://benevolent.ai/guacamol.
- 67Kaelbling, L. P.; Littman, M. L.; Moore, A. W. Reinforcement learning: A survey. J. Artif. Intell. Res. 1996, 4, 237– 285, DOI: 10.1613/jair.301Google ScholarThere is no corresponding record for this reference.
- 68Samanta, B.; De, A.; Ganguly, N.; Gomez-Rodriguez, M. Designing random graph models using variational autoencoders with applications to chemical design. arXiv preprint , arXiv:1802.05283, 2018.Google ScholarThere is no corresponding record for this reference.
- 69Polykovskiy, D.; Zhebrak, A.; Sanchez-Lengeling, B.; Golovanov, S.; Tatanov, O.; Belyaev, S.; Kurbanov, R.; Artamonov, A.; Aladinskiy, V.; Veselov, M.; Kadurin, A.; Johansson, S.; Chen, H.; Nikolenko, S.; Aspuru-Guzik, A.; Zhavoronkov, A. Molecular sets (MOSES): a benchmarking platform for molecular generation models. Front. Pharmacol. 2020, 11, 565644, DOI: 10.3389/fphar.2020.565644Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsl2isr4%253D&md5=bf849713f33d3c7e4b931ca98c5d0b6bMolecular sets (MOSES): a benchmarking platform for molecular generation modelsPolykovskiy, Daniil; Zhebrak, Alexander; Sanchez-Lengeling, Benjamin; Golovanov, Sergey; Tatanov, Oktai; Belyaev, Stanislav; Kurbanov, Rauf; Artamonov, Aleksey; Aladinskiy, Vladimir; Veselov, Mark; Kadurin, Artur; Johansson, Simon; Chen, Hongming; Nikolenko, Sergey; Aspuru-Guzik, Alan; Zhavoronkov, AlexFrontiers in Pharmacology (2020), 11 (), 565644CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)Generative models are becoming a tool of choice for exploring the mol. space. These models learn on a large training dataset and produce novel mol. structures with similar properties. Generated structures can be utilized for virtual screening or training semi-supervized predictive models in the downstream tasks. While there are plenty of generative models, it is unclear how to compare and rank them. In this work, we introduce a benchmarking platform called Mol. Sets (MOSES) to standardize training and comparison of mol. generative models. MOSES provides training and testing datasets, and a set of metrics to evaluate the quality and diversity of generated structures. We have implemented and compared several mol. generation models and suggest to use our results as ref. points for further advancements in generative chem. research. The platform and source code are available at https://github.com/molecularsets/moses.
- 70Bickerton, G. R.; Paolini, G. V.; Besnard, J.; Muresan, S.; Hopkins, A. L. Quantifying the chemical beauty of drugs. Nat. Chem. 2012, 4, 90– 98, DOI: 10.1038/nchem.1243Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1aktLk%253D&md5=7ab125d4ab381924f965d072695f7432Quantifying the chemical beauty of drugsBickerton, G. Richard; Paolini, Gaia V.; Besnard, Jeremy; Muresan, Sorel; Hopkins, Andrew L.Nature Chemistry (2012), 4 (2), 90-98CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Drug-likeness is a key consideration when selecting compds. during the early stages of drug discovery. However, evaluation of drug-likeness in abs. terms does not reflect adequately the whole spectrum of compd. quality. More worryingly, widely used rules may inadvertently foster undesirable mol. property inflation as they permit the encroachment of rule-compliant compds. towards their boundaries. We propose a measure of drug-likeness based on the concept of desirability called the quant. est. of drug-likeness (QED). The empirical rationale of QED reflects the underlying distribution of mol. properties. QED is intuitive, transparent, straightforward to implement in many practical settings and allows compds. to be ranked by their relative merit. We extended the utility of QED by applying it to the problem of mol. target druggability assessment by prioritizing a large set of published bioactive compds. The measure may also capture the abstr. notion of aesthetics in medicinal chem.
- 71Wildman, S. A.; Crippen, G. M. Prediction of physicochemical parameters by atomic contributions. J. Chem. Inf. Model. 1999, 39, 868– 873, DOI: 10.1021/ci990307lGoogle Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXlt1WjtbY%253D&md5=5eb46da66f7861906be7078f0b7e1b95Prediction of Physicochemical Parameters by Atomic ContributionsWildman, Scott A.; Crippen, Gordon M.Journal of Chemical Information and Computer Sciences (1999), 39 (5), 868-873CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)We present a new atom type classification system for use in atom-based calcn. of partition coeff. (log P) and molar refractivity (MR) designed in part to address published concerns of previous at. methods. The 68 at. contributions to log P have been detd. by fitting an extensive training set of 9920 mols., with r2 = 0.918 and σ = 0.677. A sep. set of 3412 mols. was used for the detn. of contributions to MR with r2 = 0.997 and σ = 1.43. Both calcns. are shown to have high predictive ability.
- 72Ertl, P.; Schuffenhauer, A. Estimation of synthetic accessibility score of drug-like molecules based on molecular complexity and fragment contributions. J. Cheminf. 2009, 1, 1– 11, DOI: 10.1186/1758-2946-1-8Google ScholarThere is no corresponding record for this reference.
- 73Landrum, G.; Tosco, P.; Kelley, B.; Sriniker, R.; Gedeck; ; Vianello, R.; Schneider, N.; Kawashima, E.; Dalke, A.; N, D.; Cosgrove, D.; Cole, B.; Swain, M.; Turk, S.; Savelyev, A.; Jones, G.; Vaucher, A.; Wójcikowski, M.; Take, I.; Probst, D.; Ujihara, K.; Scalfani, V. F.; godin, G.; Pahl, A.; Berenger, F.; Varjo, J. L.; Strets, J. P.; Doliath Gavid rdkit/rdkit: 2022_03_1 (Q1 2022) Release. 2022, DOI: 10.5281/zenodo.6388425 .Google ScholarThere is no corresponding record for this reference.
- 74Kullback, S.; Leibler, R. A. On information and sufficiency. Ann. Math. Stat. 1951, 22, 79– 86, DOI: 10.1214/aoms/1177729694Google ScholarThere is no corresponding record for this reference.
- 75Ramakrishnan, R.; Dral, P. O.; Rupp, M.; Von Lilienfeld, O. A. Quantum chemistry structures and properties of 134 kilo molecules. Sci. Data 2014, 1, 1– 7, DOI: 10.1038/sdata.2014.22Google ScholarThere is no corresponding record for this reference.
- 76Ruddigkeit, L.; Van Deursen, R.; Blum, L. C.; Reymond, J.-L. Enumeration of 166 billion organic small molecules in the chemical universe database GDB-17. J. Chem. Inf. Model. 2012, 52, 2864– 2875, DOI: 10.1021/ci300415dGoogle Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFClsL3J&md5=d0bf9a29f3e9ae1e57bb1c953a562cedEnumeration of 166 Billion Organic Small Molecules in the Chemical Universe Database GDB-17Ruddigkeit, Lars; van Deursen, Ruud; Blum, Lorenz C.; Reymond, Jean-LouisJournal of Chemical Information and Modeling (2012), 52 (11), 2864-2875CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Drug mols. consist of a few tens of atoms connected by covalent bonds. How many such mols. are possible in total and what is their structure. This question is of pressing interest in medicinal chem. to help solve the problems of drug potency, selectivity, and toxicity and reduce attrition rates by pointing to new mol. series. To better define the unknown chem. space, we have enumerated 166.4 billion mols. of up to 17 atoms of C, N, O, S, and halogens forming the chem. universe database GDB-17, covering a size range contg. many drugs and typical for lead compds. GDB-17 contains millions of isomers of known drugs, including analogs with high shape similarity to the parent drug. Compared to known mols. in PubChem, GDB-17 mols. are much richer in nonarom. heterocycles, quaternary centers, and stereoisomers, densely populate the third dimension in shape space, and represent many more scaffold types.
- 77Schuld, M.; Petruccione, F. Supervised Learning with Quantum Computers, Vol. 17; Springer, 2018.Google ScholarThere is no corresponding record for this reference.
- 78Schuld, M.; Bocharov, A.; Svore, K. M.; Wiebe, N. Circuit-centric quantum classifiers. Phys. Rev. A 2020, 101, 032308, DOI: 10.1103/PhysRevA.101.032308Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXptVyjsLo%253D&md5=727a05768b62c15944502aaa96467a03Circuit-centric quantum classifiersSchuld, Maria; Bocharov, Alex; Svore, Krysta M.; Wiebe, NathanPhysical Review A (2020), 101 (3), 032308CODEN: PRAHC3; ISSN:2469-9934. (American Physical Society)Variational quantum circuits are becoming tools of choice in quantum optimization and machine learning. In this paper we investigate a class of variational circuits for the purposes of supervised machine learning. We propose a circuit architecture suitable for predicting class labels of quantumly encoded data via measurements of certain observables. We observe that the required depth of a trainable classification circuit is related to the no. of representative principal components of the data distribution. Quantum circuit architectures used in our design are validated by numerical simulation, which shows significant model size redn. compared to classical predictive models. Circuit-based models demonstrate good resilience to noise, which makes then robust and error tolerant.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 27 publications.
- Hongni Jin, Kenneth M. Merz, Jr. Integrating Machine Learning and Quantum Circuits for Proton Affinity Predictions. Journal of Chemical Theory and Computation 2025, 21
(5)
, 2235-2243. https://doi.org/10.1021/acs.jctc.4c01609
- Hongfu Lu, Deheng Sun, Zhen Wang, Hui Cui, Lihua Min, Haoyu Zhang, Yihong Zhang, Jianping Wu, Xin Cai, Xiao Ding, Man Zhang, Alex Aliper, Feng Ren, Alex Zhavoronkov. Design, Synthesis, and Biological Evaluation of Novel Orally Available Covalent CDK12/13 Dual Inhibitors for the Treatment of Tumors. Journal of Medicinal Chemistry 2025, 68
(4)
, 4148-4167. https://doi.org/10.1021/acs.jmedchem.4c01616
- Meng Zhang, Xiaoyu Ding, Zhongying Cao, Yilin Yang, Xiao Ding, Xin Cai, Man Zhang, Alex Aliper, Feng Ren, Hongfu Lu, Alex Zhavoronkov. Discovery of Potent, Highly Selective, and Orally Bioavailable MTA Cooperative PRMT5 Inhibitors with Robust In Vivo Antitumor Activity. Journal of Medicinal Chemistry 2025, 68
(2)
, 1940-1955. https://doi.org/10.1021/acs.jmedchem.4c02732
- Hongfu Lu, Yihong Zhang, Jinxin Liu, Tao Jiang, Xiang Yu, Haoyu Zhang, Tao Liang, Jingjing Peng, Xin Cai, Xiaoling Lan, Jinmin Ren, Mei Ge, Jingyang Zhang, Jingjin Shang, Jiaojiao Yu, Hongcan Ren, Qiang Liu, Jinting Gao, Lili Tang, Xiao Ding, Man Zhang, Alex Aliper, Qiang Lu, Fusheng Zhou, Jiong Lan, Feng Ren, Alex Zhavoronkov. Discovery of a Novel Macrocyclic Noncovalent CDK7 Inhibitor for Cancer Therapy. Journal of Medicinal Chemistry 2024, 67
(22)
, 20580-20594. https://doi.org/10.1021/acs.jmedchem.4c02098
- Matthew A. Dorsey, Kelvin Dsouza, Dhruv Ranganath, Joshua S. Harris, Thomas R. Lane, Fabio Urbina, Sean Ekins. Near-Term Quantum Classification Algorithms Applied to Antimalarial Drug Discovery. Journal of Chemical Information and Modeling 2024, 64
(15)
, 5922-5930. https://doi.org/10.1021/acs.jcim.4c00953
- Anthony M. Smaldone, Victor S. Batista. Quantum-to-Classical Neural Network Transfer Learning Applied to Drug Toxicity Prediction. Journal of Chemical Theory and Computation 2024, 20
(11)
, 4901-4908. https://doi.org/10.1021/acs.jctc.4c00432
- Yazhou Wang, Chao Wang, Tingting Liu, Hongyun Qi, Shan Chen, Xin Cai, Man Zhang, Alex Aliper, Feng Ren, Xiao Ding, Alex Zhavoronkov. Discovery of Tetrahydropyrazolopyrazine Derivatives as Potent and Selective MYT1 Inhibitors for the Treatment of Cancer. Journal of Medicinal Chemistry 2024, 67
(1)
, 420-432. https://doi.org/10.1021/acs.jmedchem.3c01476
- Amandeep Singh Bhatia, Mandeep Kaur Saggi, Sabre Kais. Quantum Machine Learning Predicting ADME-Tox Properties in Drug Discovery. Journal of Chemical Information and Modeling 2023, 63
(21)
, 6476-6486. https://doi.org/10.1021/acs.jcim.3c01079
- Alexander Hagg, Karl N. Kirschner. Open-Source Machine Learning in Computational Chemistry. Journal of Chemical Information and Modeling 2023, 63
(15)
, 4505-4532. https://doi.org/10.1021/acs.jcim.3c00643
- Nikolay Shilov, Andrew Ponomarev, Dmitry Ryumin, Alexey Karpov. Generative Adversarial Framework with Composite Discriminator for Organization and Process Modelling—Smart City Cases. Smart Cities 2025, 8
(2)
, 38. https://doi.org/10.3390/smartcities8020038
- Orkid Coskuner-Weber, Pier Luigi Gentili, Vladimir N. Uversky. Integrating chemical artificial intelligence and cognitive computing for predictive analysis of biological pathways: a case for intrinsically disordered proteins. Biophysical Reviews 2025, 122 https://doi.org/10.1007/s12551-025-01286-x
- Tigran Sedrakyan, Alexia Salavrakos. Photonic quantum generative adversarial networks for classical data. Optica Quantum 2024, 2
(6)
, 458. https://doi.org/10.1364/OPTICAQ.530346
- Zhijiang Yang, Tengxin Huang, Li Pan, Jingjing Wang, Liangliang Wang, Junjie Ding, Junhua Xiao. QuanDB: a quantum chemical property database towards enhancing 3D molecular representation learning. Journal of Cheminformatics 2024, 16
(1)
https://doi.org/10.1186/s13321-024-00843-y
- Jay Shree Mathivanan, Victor Violet Dhayabaran, Mary Rajathei David, Muthugobal Bagayalakshmi Karuna Nidhi, Karuppasamy Muthuvel Prasath, Suvaiyarasan Suvaithenamudhan. Application of Deep Learning Neural Networks in Computer-Aided Drug Discovery: A Review. Current Bioinformatics 2024, 19
(9)
, 851-858. https://doi.org/10.2174/0115748936276510231123121404
- Edward Naveen V, Jenefa A, Thiyagu T.M, Lincy A, Antony Taurshia. DeepGAN: Utilizing generative adversarial networks for improved deep learning. International Journal of Knowledge-Based and Intelligent Engineering Systems 2024, 28
(4)
, 732-748. https://doi.org/10.3233/KES-230326
- Haoran Ma, Liao Ye, Xiaoqing Guo, Fanjie Ruan, Zichao Zhao, Maohui Li, Yuehai Wang, Jianyi Yang. Quantum Generative Adversarial Networks in a Silicon Photonic Chip with Maximum Expressibility. Advanced Quantum Technologies 2024, 12 https://doi.org/10.1002/qute.202400171
- Yan Guo, Yongqiang Gao, Jiawei Song. MolCFL: A personalized and privacy-preserving drug discovery framework based on generative clustered federated learning. Journal of Biomedical Informatics 2024, 157 , 104712. https://doi.org/10.1016/j.jbi.2024.104712
- Amit Gangwal, Azim Ansari, Iqrar Ahmad, Abul Kalam Azad, Wan Mohd Azizi Wan Sulaiman. Current strategies to address data scarcity in artificial intelligence-based drug discovery: A comprehensive review. Computers in Biology and Medicine 2024, 179 , 108734. https://doi.org/10.1016/j.compbiomed.2024.108734
- Zamara Mariam, Sarfaraz K. Niazi, Matthias Magoola. Unlocking the Future of Drug Development: Generative AI, Digital Twins, and Beyond. BioMedInformatics 2024, 4
(2)
, 1441-1456. https://doi.org/10.3390/biomedinformatics4020079
- Mengxian Yu, Yin-Ning Zhou, Qiang Wang, Fangyou Yan. Extrapolation validation (EV): a universal validation method for mitigating machine learning extrapolation risk. Digital Discovery 2024, 3
(5)
, 1058-1067. https://doi.org/10.1039/D3DD00256J
- Evan Xie, Karin Hasegawa, Georgios Kementzidis, Evangelos Papadopoulos, Bertal Huseyin Aktas, Yuefan Deng. An AI-Driven Framework for Discovery of BACE1 Inhibitors for Alzheimer’s Disease. 2024https://doi.org/10.1101/2024.05.15.594361
- Wenfeng Fan, Yue He, Fei Zhu. RM-GPT: Enhance the comprehensive generative ability of molecular GPT model via LocalRNN and RealFormer. Artificial Intelligence in Medicine 2024, 150 , 102827. https://doi.org/10.1016/j.artmed.2024.102827
- Alexey Pyrkov, Alex Aliper, Dmitry Bezrukov, Dmitriy Podolskiy, Feng Ren, Alex Zhavoronkov. Complexity of life sciences in quantum and
AI
era. WIREs Computational Molecular Science 2024, 14
(1)
https://doi.org/10.1002/wcms.1701
- Kit-Kay Mak, Yi-Hang Wong, Mallikarjuna Rao Pichika. Artificial Intelligence in Drug Discovery and Development. 2024, 1461-1498. https://doi.org/10.1007/978-3-031-35529-5_92
- Matvei Anoshin, Asel Sagingalieva, Christopher Mansell, Dmitry Zhiganov, Vishal Shete, Markus Pflitsch, Alexey Melnikov. Hybrid Quantum Cycle Generative Adversarial Network for Small Molecule Generation. IEEE Transactions on Quantum Engineering 2024, 5 , 1-14. https://doi.org/10.1109/TQE.2024.3414264
- Fangyou Yan, Mengxian Yu, Yin-Ning Zhou, Qiang Wang. A Universal Validation Method for Mitigating Machine Learning Extrapolation Risk. 2023https://doi.org/10.21203/rs.3.rs-3758965/v1
- Minjae J. Kim, Cole A. Martin, Jinhwa Kim, Monica M. Jablonski. Computational methods in glaucoma research: Current status and future outlook. Molecular Aspects of Medicine 2023, 94 , 101222. https://doi.org/10.1016/j.mam.2023.101222
- Alexey Pyrkov, Alex Aliper, Dmitry Bezrukov, Yen-Chu Lin, Daniil Polykovskiy, Petrina Kamya, Feng Ren, Alex Zhavoronkov. Quantum computing for near-term applications in generative chemistry and drug discovery. Drug Discovery Today 2023, 28
(8)
, 103675. https://doi.org/10.1016/j.drudis.2023.103675
- Kit-Kay Mak, Yi-Hang Wong, Mallikarjuna Rao Pichika. Artificial Intelligence in Drug Discovery and Development. 2023, 1-38. https://doi.org/10.1007/978-3-030-73317-9_92-1
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Overall pipeline. (a) The overall pipeline of MolGAN with different combinations of classical/quantum components. The reward neural network branch is enabled in the goal-directed benchmark. The classical noise generator samples from the Gaussian distribution, and the quantum one uses the variational quantum circuit (VQC). The classical generator is built by neural networks, and the quantum one uses the patch-based VQC to generate the molecular graph. The molecular graph is represented by a bond matrix and atom vector. The classical discriminator is built by a graph-based neural network or multilayer perceptron (MLP), and the VQC is used in the quantum one. (b) The example of VQC in the noise generator. (c) The patch method uses multiple VQCs as subgenerators. Each subgenerator takes noise as input and outputs a partial part of the final molecular graph. The final molecular graph is constructed by concatenating all the partial patches together. (d) The example of VQC in the quantum generator. (e) The VQC of the quantum discriminator consists of the amplitude embedding circuit (Sx), the strong entanglement layers (Uθ), and the measurement. (f) The VQC of strongly entanglement layers contains multiple CNOT gates and parametrized rotational gates (R). (g) MLP-based discriminator architecture in MolGAN-CC.
Figure 2
Figure 2. Property distributions of molecules. (a) Drug properties distributions (left to right: QED, SA, and Solute) from valid and unique MolGAN-generated (in blue) and QuMolGAN-generated (in orange) molecules. (b) KL-divergence distributions (left to right: MolLogP, BertzCT, and MolWt) of valid and unique MolGAN-generated (blue), QuMolGAN-generated (orange), and QM9 (gray) molecules. (c) KL-divergence distributions (from left to right: MolLogP, BertzCT, and MolWt) of MolGAN-CC-ER-generated (orange), MolGAN-CQ-generated (yellow), MolGAN-generated (blue), and QM9 (gray) molecules.
Figure 3
Figure 3. Example molecules. (a) Example molecules of MolGAN with z_dim = 2. (b) Example molecules of QuMolGAN with z_dim = 2. (c) Example molecules of MolGAN-CQ.
References
This article references 78 other publications.
- 1Hingorani, A. D.; Kuan, V.; Finan, C.; Kruger, F. A.; Gaulton, A.; Chopade, S.; Sofat, R.; MacAllister, R. J.; Overington, J. P.; Hemingway, H.; Denaxas, S.; Prieto, D.; Casas, J. P. Improving the odds of drug development success through human genomics: modelling study. Sci. Rep. 2019, 9, 1– 25, DOI: 10.1038/s41598-019-54849-wThere is no corresponding record for this reference.
- 2Abreu, J. L. Ivermectin for the Prevention of COVID-19 So...WHO is Telling the Truth. Revista Daena (Int. J. Good Conscience) 2020, 15 (2), 1– 30There is no corresponding record for this reference.
- 3Wouters, O. J.; McKee, M.; Luyten, J. Estimated research and development investment needed to bring a new medicine to market, 2009–2018. Jama 2020, 323, 844– 853, DOI: 10.1001/jama.2020.11663https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB387oslyhsA%253D%253D&md5=ff63c2f03e59ef0223585a71fb6d5df0Estimated Research and Development Investment Needed to Bring a New Medicine to Market, 2009-2018Wouters Olivier J; McKee Martin; Luyten JeroenJAMA (2020), 323 (9), 844-853 ISSN:.IMPORTANCE: The mean cost of developing a new drug has been the subject of debate, with recent estimates ranging from $314 million to $2.8 billion. OBJECTIVE: To estimate the research and development investment required to bring a new therapeutic agent to market, using publicly available data. DESIGN AND SETTING: Data were analyzed on new therapeutic agents approved by the US Food and Drug Administration (FDA) between 2009 and 2018 to estimate the research and development expenditure required to bring a new medicine to market. Data were accessed from the US Securities and Exchange Commission, Drugs@FDA database, and ClinicalTrials.gov, alongside published data on clinical trial success rates. EXPOSURES: Conduct of preclinical and clinical studies of new therapeutic agents. MAIN OUTCOMES AND MEASURES: Median and mean research and development spending on new therapeutic agents approved by the FDA, capitalized at a real cost of capital rate (the required rate of return for an investor) of 10.5% per year, with bootstrapped CIs. All amounts were reported in 2018 US dollars. RESULTS: The FDA approved 355 new drugs and biologics over the study period. Research and development expenditures were available for 63 (18%) products, developed by 47 different companies. After accounting for the costs of failed trials, the median capitalized research and development investment to bring a new drug to market was estimated at $985.3 million (95% CI, $683.6 million-$1228.9 million), and the mean investment was estimated at $1335.9 million (95% CI, $1042.5 million-$1637.5 million) in the base case analysis. Median estimates by therapeutic area (for areas with ≥5 drugs) ranged from $765.9 million (95% CI, $323.0 million-$1473.5 million) for nervous system agents to $2771.6 million (95% CI, $2051.8 million-$5366.2 million) for antineoplastic and immunomodulating agents. Data were mainly accessible for smaller firms, orphan drugs, products in certain therapeutic areas, first-in-class drugs, therapeutic agents that received accelerated approval, and products approved between 2014 and 2018. Results varied in sensitivity analyses using different estimates of clinical trial success rates, preclinical expenditures, and cost of capital. CONCLUSIONS AND RELEVANCE: This study provides an estimate of research and development costs for new therapeutic agents based on publicly available data. Differences from previous studies may reflect the spectrum of products analyzed, the restricted availability of data in the public domain, and differences in underlying assumptions in the cost calculations.
- 4Aliper, A.; Plis, S.; Artemov, A.; Ulloa, A.; Mamoshina, P.; Zhavoronkov, A. Deep learning applications for predicting pharmacological properties of drugs and drug repurposing using transcriptomic data. Mol. Pharmaceutics 2016, 13, 2524– 2530, DOI: 10.1021/acs.molpharmaceut.6b002484https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xot1ers7w%253D&md5=853efb9b636e4c3c8a15bd4c53ecac99Deep Learning Applications for Predicting Pharmacological Properties of Drugs and Drug Repurposing Using Transcriptomic DataAliper, Alexander; Plis, Sergey; Artemov, Artem; Ulloa, Alvaro; Mamoshina, Polina; Zhavoronkov, AlexMolecular Pharmaceutics (2016), 13 (7), 2524-2530CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Deep learning is rapidly advancing many areas of science and technol. with multiple success stories in image, text, voice and video recognition, robotics, and autonomous driving. In this paper we demonstrate how deep neural networks (DNN) trained on large transcriptional response data sets can classify various drugs to therapeutic categories solely based on their transcriptional profiles. We used the perturbation samples of 678 drugs across A549, MCF-7, and PC-3 cell lines from the LINCS Project and linked those to 12 therapeutic use categories derived from MeSH. To train the DNN, we utilized both gene level transcriptomic data and transcriptomic data processed using a pathway activation scoring algorithm, for a pooled data set of samples perturbed with different concns. of the drug for 6 and 24 h. In both pathway and gene level classification, DNN achieved high classification accuracy and convincingly outperformed the support vector machine (SVM) model on every multiclass classification problem, however, models based on pathway level data performed significantly better. For the first time we demonstrate a deep learning neural net trained on transcriptomic data to recognize pharmacol. properties of multiple drugs across different biol. systems and conditions. We also propose using deep neural net confusion matrixes for drug repositioning. This work is a proof of principle for applying deep learning to drug discovery and development.
- 5Kadurin, A.; Aliper, A.; Kazennov, A.; Mamoshina, P.; Vanhaelen, Q.; Khrabrov, K.; Zhavoronkov, A. The cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncology. Oncotarget 2017, 8, 10883, DOI: 10.18632/oncotarget.140735https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1c%252FpvFKruw%253D%253D&md5=677ef0264494eb8a7ef8c6584c1202abThe cornucopia of meaningful leads: Applying deep adversarial autoencoders for new molecule development in oncologyKadurin Artur; Khrabrov Kuzma; Kadurin Artur; Aliper Alexander; Kazennov Andrey; Mamoshina Polina; Vanhaelen Quentin; Zhavoronkov Alex; Kadurin Artur; Kadurin Artur; Kazennov Andrey; Zhavoronkov Alex; Mamoshina Polina; Zhavoronkov AlexOncotarget (2017), 8 (7), 10883-10890 ISSN:.Recent advances in deep learning and specifically in generative adversarial networks have demonstrated surprising results in generating new images and videos upon request even using natural language as input. In this paper we present the first application of generative adversarial autoencoders (AAE) for generating novel molecular fingerprints with a defined set of parameters. We developed a 7-layer AAE architecture with the latent middle layer serving as a discriminator. As an input and output the AAE uses a vector of binary fingerprints and concentration of the molecule. In the latent layer we also introduced a neuron responsible for growth inhibition percentage, which when negative indicates the reduction in the number of tumor cells after the treatment. To train the AAE we used the NCI-60 cell line assay data for 6252 compounds profiled on MCF-7 cell line. The output of the AAE was used to screen 72 million compounds in PubChem and select candidate molecules with potential anti-cancer properties. This approach is a proof of concept of an artificially-intelligent drug discovery engine, where AAEs are used to generate new molecular fingerprints with the desired molecular properties.
- 6Zhavoronkov, A. Artificial intelligence for drug discovery, biomarker development, and generation of novel chemistry. Mol. Pharmaceutics 2018, 15, 4311, DOI: 10.1021/acs.molpharmaceut.8b009306https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslOgtrjE&md5=2c51295d4e682eeaf8069eddc09ed3e8Artificial Intelligence for Drug Discovery, Biomarker Development, and Generation of Novel ChemistryZhavoronkov, AlexMolecular Pharmaceutics (2018), 15 (10), 4311-4313CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The potential of artificial intelligence in drug discovery is discussed and reviewed.
- 7Carracedo-Reboredo, P.; Liñares-Blanco, J.; Rodríguez-Fernández, N.; Cedrón, F.; Novoa, F. J.; Carballal, A.; Maojo, V.; Pazos, A.; Fernandez-Lozano, C. A review on machine learning approaches and trends in drug discovery. Comput. Struct. Biotechnol. J. 2021, 19, 4538– 4558, DOI: 10.1016/j.csbj.2021.08.0117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisF2gs7zM&md5=eccac145867b8821f0719463ec1be641A review on machine learning approaches and trends in drug discoveryCarracedo-Reboredo, Paula; Linares-Blanco, Jose; Rodriguez-Fernandez, Nereida; Cedron, Francisco; Novoa, Francisco J.; Carballal, Adrian; Maojo, Victor; Pazos, Alejandro; Fernandez-Lozano, CarlosComputational and Structural Biotechnology Journal (2021), 19 (), 4538-4558CODEN: CSBJAC; ISSN:2001-0370. (Elsevier B.V.)A review. Drug discovery aims at finding new compds. with specific chem. properties for the treatment of diseases. In the last years, the approach used in this search presents an important component in computer science with the skyrocketing of machine learning techniques due to its democratization. With the objectives set by the Precision Medicine initiative and the new challenges generated, it is necessary to establish robust, std. and reproducible computational methodologies to achieve the objectives set. Currently, predictive models based on Machine Learning have gained great importance in the step prior to preclin. studies. This stage manages to drastically reduce costs and research times in the discovery of new drugs. This review article focuses on how these new methodologies are being used in recent years of research. Analyzing the state of the art in this field will give us an idea of where cheminformatics will be developed in the short term, the limitations it presents and the pos. results it has achieved. This review will focus mainly on the methods used to model the mol. data, as well as the biol. problems addressed and the Machine Learning algorithms used for drug discovery in recent years.
- 8Kao, P.-Y.; Kao, S.-M.; Huang, N.-L.; Lin, Y.-C. Toward Drug-Target Interaction Prediction via Ensemble Modeling and Transfer Learning. In 2021 IEEE International Conference on Bioinformatics and Biomedicine (BIBM) , 2021; pp 2384– 2391.There is no corresponding record for this reference.
- 9Kolluri, S.; Lin, J.; Liu, R.; Zhang, Y.; Zhang, W. Machine Learning and Artificial Intelligence in Pharmaceutical Research and Development: a Review. AAPS J. 2022, 24, 1– 10, DOI: 10.1208/s12248-021-00644-3There is no corresponding record for this reference.
- 10Zhavoronkov, A.; Ivanenkov, Y. A.; Aliper, A.; Veselov, M. S.; Aladinskiy, V. A.; Aladinskaya, A. V.; Terentiev, V. A.; Polykovskiy, D. A.; Kuznetsov, M. D.; Asadulaev, A.; Volkov, Y.; Zholus, A.; Shayakhmetov, R. R.; Zhebrak, A.; Minaeva, L. I.; Zagribelnyy, B. A.; Lee, L. H.; Soll, R.; Madge, D.; Xing, L.; Guo, T.; Aspuru-Guzik, A. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat. Biotechnol. 2019, 37, 1038– 1040, DOI: 10.1038/s41587-019-0224-x10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12gurnM&md5=b15262b61b9172ab2bc37e534a70f010Deep learning enables rapid identification of potent DDR1 kinase inhibitorsZhavoronkov, Alex; Ivanenkov, Yan A.; Aliper, Alex; Veselov, Mark S.; Aladinskiy, Vladimir A.; Aladinskaya, Anastasiya V.; Terentiev, Victor A.; Polykovskiy, Daniil A.; Kuznetsov, Maksim D.; Asadulaev, Arip; Volkov, Yury; Zholus, Artem; Shayakhmetov, Rim R.; Zhebrak, Alexander; Minaeva, Lidiya I.; Zagribelnyy, Bogdan A.; Lee, Lennart H.; Soll, Richard; Madge, David; Xing, Li; Guo, Tao; Aspuru-Guzik, AlanNature Biotechnology (2019), 37 (9), 1038-1040CODEN: NABIF9; ISSN:1087-0156. (Nature Research)We have developed a deep generative model, generative tensorial reinforcement learning (GENTRL), for de novo small-mol. design. GENTRL optimizes synthetic feasibility, novelty, and biol. activity. We used GENTRL to discover potent inhibitors of discoidin domain receptor 1 (DDR1), a kinase target implicated in fibrosis and other diseases, in 21 days. Four compds. were active in biochem. assays, and two were validated in cell-based assays. One lead candidate was tested and demonstrated favorable pharmacokinetics in mice.
- 11Chan, H. S.; Shan, H.; Dahoun, T.; Vogel, H.; Yuan, S. Advancing drug discovery via artificial intelligence. Trends Pharmacol. Sci. 2019, 40, 592– 604, DOI: 10.1016/j.tips.2019.06.00411https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlalsbbL&md5=03ebb3bc60278061eb29feb531b92d02Advancing Drug Discovery via Artificial IntelligenceChan, H. C. Stephen; Shan, Hanbin; Dahoun, Thamani; Vogel, Horst; Yuan, ShuguangTrends in Pharmacological Sciences (2019), 40 (8), 592-604CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)Drug discovery and development are among the most important translational science activities that contribute to human health and wellbeing. However, the development of a new drug is a very complex, expensive, and long process which typically costs 2.6 billion USD and takes 12 years on av. How to decrease the costs and speed up new drug discovery has become a challenging and urgent question in industry. Artificial intelligence (AI) combined with new exptl. technologies is expected to make the hunt for new pharmaceuticals quicker, cheaper, and more effective. We discuss here emerging applications of AI to improve the drug discovery process.
- 12Schneider, G.; Fechner, U. Computer-based de novo design of drug-like molecules. Nat. Rev. Drug Discovery 2005, 4, 649– 663, DOI: 10.1038/nrd179912https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmvVOqtro%253D&md5=a30dbc58ed81e0b7fe3f7d41a668e9acComputer-based de novo design of drug-like moleculesSchneider, Gisbert; Fechner, UliNature Reviews Drug Discovery (2005), 4 (8), 649-663CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review with refs. Ever since the first automated de novo design techniques were conceived only 15 years ago, the computer-based design of hit and lead structure candidates has emerged as a complementary approach to high-throughput screening. Although many challenges remain, de novo design supports drug discovery projects by generating novel pharmaceutically active agents with desired properties in a cost- and time-efficient manner. In this review, we outline the various design concepts and highlight current developments in computer-based de novo design.
- 13Fischer, T.; Gazzola, S.; Riedl, R. Approaching target selectivity by de novo drug design. Expert Opin. Drug Discovery 2019, 14, 791– 803, DOI: 10.1080/17460441.2019.161543513https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFeqtrbJ&md5=fd845eb91cde2a8042ec28fc2b97dd32Approaching Target Selectivity by De Novo Drug DesignFischer, Thomas; Gazzola, Silvia; Riedl, RainerExpert Opinion on Drug Discovery (2019), 14 (8), 791-803CODEN: EODDBX; ISSN:1746-0441. (Taylor & Francis Ltd.)A review. The development of drug candidates with a defined selectivity profile and a unique mol. structure is of fundamental interest for drug discovery. In contrast to the costly screening of large substance libraries, the targeted de novo design of a drug by using structural information of either the biol. target and/or structure-activity relationship data of active modulators offers an efficient and intellectually appealing alternative.: This review provides an overview on the different techniques of de novo drug design (ligand-based drug design, structure-based drug design, and fragment-based drug design) and highlights successful examples of this targeted approach toward selective modulators of therapeutically relevant targets.: De novo drug design has established itself as a very efficient method for the development of potent and selective modulators for a variety of different biol. target classes. The ever-growing wealth of structural data on therapeutic targets will certainly further enhance the importance of de novo design for the drug discovery process in the future. However, a consistent use of the terminol. of de novo drug design in the scientific literature should be sought.
- 14Mouchlis, V. D.; Afantitis, A.; Serra, A.; Fratello, M.; Papadiamantis, A. G.; Aidinis, V.; Lynch, I.; Greco, D.; Melagraki, G. Advances in de novo drug design: from conventional to machine learning methods. Int. J. Mol. Sci. 2021, 22, 1676, DOI: 10.3390/ijms22041676There is no corresponding record for this reference.
- 15Speck-Planche, A. Recent advances in fragment-based computational drug design: tackling simultaneous targets/biological effects. Future Med. Chem. 2018, 10, 2021– 2024, DOI: 10.4155/fmc-2018-021315https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsF2gsLzJ&md5=a6a28a9e6dde2e290064bdf0f646b7e1Recent advances in fragment-based computational drug design: tackling simultaneous targets/biological effectsSpeck-Planche, AlejandroFuture Medicinal Chemistry (2018), 10 (17), 2021-2024CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)There is no expanded citation for this reference.
- 16Mamoshina, P.; Ojomoko, L.; Yanovich, Y.; Ostrovski, A.; Botezatu, A.; Prikhodko, P.; Izumchenko, E.; Aliper, A.; Romantsov, K.; Zhebrak, A.; Ogu, I. O.; Zhavoronkov, A. Converging blockchain and next-generation artificial intelligence technologies to decentralize and accelerate biomedical research and healthcare. Oncotarget 2018, 9, 5665, DOI: 10.18632/oncotarget.2234516https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mrjs1Ontw%253D%253D&md5=d2ee7d11f2b610408b9ad2007d0d022bConverging blockchain and next-generation artificial intelligence technologies to decentralize and accelerate biomedical research and healthcareMamoshina Polina; Ojomoko Lucy; Aliper Alexander; Romantsov Konstantin; Zhebrak Alexander; Zhavoronkov Alex; Mamoshina Polina; Yanovich Yury; Ostrovski Alex; Botezatu Alex; Prikhodko Pavel; Izumchenko Eugene; Ogu Iraneus Obioma; Zhavoronkov AlexOncotarget (2018), 9 (5), 5665-5690 ISSN:.The increased availability of data and recent advancements in artificial intelligence present the unprecedented opportunities in healthcare and major challenges for the patients, developers, providers and regulators. The novel deep learning and transfer learning techniques are turning any data about the person into medical data transforming simple facial pictures and videos into powerful sources of data for predictive analytics. Presently, the patients do not have control over the access privileges to their medical records and remain unaware of the true value of the data they have. In this paper, we provide an overview of the next-generation artificial intelligence and blockchain technologies and present innovative solutions that may be used to accelerate the biomedical research and enable patients with new tools to control and profit from their personal data as well with the incentives to undergo constant health monitoring. We introduce new concepts to appraise and evaluate personal records, including the combination-, time- and relationship-value of the data. We also present a roadmap for a blockchain-enabled decentralized personal health data ecosystem to enable novel approaches for drug discovery, biomarker development, and preventative healthcare. A secure and transparent distributed personal data marketplace utilizing blockchain and deep learning technologies may be able to resolve the challenges faced by the regulators and return the control over personal data including medical records back to the individuals.
- 17Zhavoronkov, A.; Zagribelnyy, B.; Zhebrak, A.; Aladinskiy, V.; Terentiev, V.; Vanhaelen, Q.; Bezrukov, D. S.; Polykovskiy, D.; Shayakhmetov, R.; Filimonov, A.; Filimonov, A.; Bishop, M.; McCloskey, S.; Lejia, E.; Bright, D.; Funakawa, K.; Lin, Y.-C.; Huang, S.-H.; Liao, H.-J.; Aliper, A.; Ivanenkov, Y. Potential non-covalent SARS-CoV-2 3C-like protease inhibitors designed using generative deep learning approaches and reviewed by human medicinal chemist in virtual reality. ChemRxiv , 2020.There is no corresponding record for this reference.
- 18Paul, D.; Sanap, G.; Shenoy, S.; Kalyane, D.; Kalia, K.; Tekade, R. K. Artificial intelligence in drug discovery and development. Drug Discovery Today 2021, 26, 80, DOI: 10.1016/j.drudis.2020.10.01018https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlentrbI&md5=092fc22fb6e29075f3ccd8867f0c65f1Artificial intelligence in drug discovery and developmentPaul, Debleena; Sanap, Gaurav; Shenoy, Snehal; Kalyane, Dnyaneshwar; Kalia, Kiran; Tekade, Rakesh K.Drug Discovery Today (2021), 26 (1), 80-93CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. Artificial Intelligence (AI) has recently started to gear-up its application in various sectors of the society with the pharmaceutical industry as a front-runner beneficiary. This review highlights the impactful use of AI in diverse areas of the pharmaceutical sectors viz., drug discovery and development, drug repurposing, improving pharmaceutical productivity, clin. trials, etc. to name a few, thus reducing the human workload as well as achieving targets in a short period. Crosstalk on the tools and techniques utilized in enforcing AI, ongoing challenges, and ways to overcome them, along with the future of AI in the pharmaceutical industry, is also discussed.
- 19Martinelli, D. Generative machine learning for de novo drug discovery: A systematic review. Comput. Biol. Med. 2022, 145, 105403, DOI: 10.1016/j.compbiomed.2022.10540319https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2MzptFantQ%253D%253D&md5=01320d8d3a5987a1634dc2142343cd39Generative machine learning for de novo drug discovery: A systematic reviewMartinelli Dominic DComputers in biology and medicine (2022), 145 (), 105403 ISSN:.Recent research on artificial intelligence indicates that machine learning algorithms can auto-generate novel drug-like molecules. Generative models have revolutionized de novo drug discovery, rendering the explorative process more efficient. Several model frameworks and input formats have been proposed to enhance the performance of intelligent algorithms in generative molecular design. In this systematic literature review of experimental articles and reviews over the last five years, machine learning models, challenges associated with computational molecule design along with proposed solutions, and molecular encoding methods are discussed. A query-based search of the PubMed, ScienceDirect, Springer, Wiley Online Library, arXiv, MDPI, bioRxiv, and IEEE Xplore databases yielded 87 studies. Twelve additional studies were identified via citation searching. Of the articles in which machine learning was implemented, six prominent algorithms were identified: long short-term memory recurrent neural networks (LSTM-RNNs), variational autoencoders (VAEs), generative adversarial networks (GANs), adversarial autoencoders (AAEs), evolutionary algorithms, and gated recurrent unit (GRU-RNNs). Furthermore, eight central challenges were designated: homogeneity of generated molecular libraries, deficient synthesizability, limited assay data, model interpretability, incapacity for multi-property optimization, incomparability, restricted molecule size, and uncertainty in model evaluation. Molecules were encoded either as strings, which were occasionally augmented using randomization, as 2D graphs, or as 3D graphs. Statistical analysis and visualization are performed to illustrate how approaches to machine learning in de novo drug design have evolved over the past five years. Finally, future opportunities and reservations are discussed.
- 20Gao, K.; Nguyen, D. D.; Tu, M.; Wei, G.-W. Generative Network Complex for the Automated Generation of Drug-like Molecules. J. Chem. Inf. Model. 2020, 60, 5682– 5698, DOI: 10.1021/acs.jcim.0c0059920https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVSms7jL&md5=d19c56b0b0752814fc8b8c5d2e50144cGenerative Network Complex for the Automated Generation of Drug-like MoleculesGao, Kaifu; Nguyen, Duc Duy; Tu, Meihua; Wei, Guo-WeiJournal of Chemical Information and Modeling (2020), 60 (12), 5682-5698CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Current drug discovery is expensive and time-consuming. It remains a challenging task to create a wide variety of novel compds. that not only have desirable pharmacol. properties but also are cheaply available to low-income people. In this work, we develop a generative network complex (GNC) to generate new drug-like mols. based on the multiproperty optimization via the gradient descent in the latent space of an autoencoder. In our GNC, both multiple chem. properties and similarity scores are optimized to generate drug-like mols. with desired chem. properties. To further validate the reliability of the predictions, these mols. are reevaluated and screened by independent 2D fingerprint-based predictors to come up with a few hundreds of new drug candidates. As a demonstration, we apply our GNC to generate a large no. of new BACE1 inhibitors, as well as thousands of novel alternative drug candidates for eight existing market drugs, including Ceritinib, Ribociclib, Acalabrutinib, Idelalisib, Dabrafenib, Macimorelin, Enzalutamide, and Panobinostat.
- 21Goodfellow, I.; Pouget-Abadie, J.; Mirza, M.; Xu, B.; Warde-Farley, D.; Ozair, S.; Courville, A.; Bengio, Y. Generative adversarial nets. In Advances in Neural Information Processing Systems 27 (NIPS 2014); Ghahramani, Z.; Welling, M.; Cortes, C.; Lawrence, N.; Weinberger, K. Q., Eds.; 2014.There is no corresponding record for this reference.
- 22Karras, T.; Aila, T.; Laine, S.; Lehtinen, J. Progressive growing of gans for improved quality, stability, and variation. arXiv preprint , 2017, arXiv:1710.10196.There is no corresponding record for this reference.
- 23Isola, P.; Zhu, J.-Y.; Zhou, T.; Efros, A. A. Image-to-image translation with conditional adversarial networks. In Proceedings of the IEEE Conference on Computer Vision and Pattern Recognition , 2017; pp 1125– 1134.There is no corresponding record for this reference.
- 24Zhu, J.-Y.; Park, T.; Isola, P.; Efros, A. A. Unpaired image-to-image translation using cycle-consistent adversarial networks. In Proceedings of the IEEE International Conference on Computer Vision , 2017; pp 2223– 2232.There is no corresponding record for this reference.
- 25Karras, T.; Laine, S.; Aila, T. A style-based generator architecture for generative adversarial networks. Proceedings of the IEEE/CVF Conference on Computer Vision and Pattern Recognition , 2019; pp 4401– 4410.There is no corresponding record for this reference.
- 26Jangid, D. K.; Brodnik, N. R.; Khan, A.; Goebel, M. G.; Echlin, M. P.; Pollock, T. M.; Daly, S. H.; Manjunath, B. 3D Grain Shape Generation in Polycrystals Using Generative Adversarial Networks. Integr. Mater. Manuf. Innov. 2022, 11, 71– 84, DOI: 10.1007/s40192-021-00244-1There is no corresponding record for this reference.
- 27Kadurin, A.; Nikolenko, S.; Khrabrov, K.; Aliper, A.; Zhavoronkov, A. druGAN: an advanced generative adversarial autoencoder model for de novo generation of new molecules with desired molecular properties in silico. Mol. Pharmaceutics 2017, 14, 3098– 3104, DOI: 10.1021/acs.molpharmaceut.7b0034627https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFGitbbF&md5=6fc20afd4c6a8188e830aa90a7875dd6druGAN: An Advanced Generative Adversarial Autoencoder Model for de Novo Generation of New Molecules with Desired Molecular Properties in SilicoKadurin, Artur; Nikolenko, Sergey; Khrabrov, Kuzma; Aliper, Alex; Zhavoronkov, AlexMolecular Pharmaceutics (2017), 14 (9), 3098-3104CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Deep generative adversarial networks (GANs) are the emerging technol. in drug discovery and biomarker development. In our recent work, we demonstrated a proof-of-concept of implementing deep generative adversarial autoencoder (AAE) to identify new mol. fingerprints with predefined anticancer properties. Another popular generative model is the variational autoencoder (VAE), which is based on deep neural architectures. In this work, we developed an advanced AAE model for mol. feature extn. problems, and demonstrated its advantages compared to VAE in terms of (a) adjustability in generating mol. fingerprints; (b) capacity of processing very large mol. data sets; and (c) efficiency in unsupervised pretraining for regression model. Our results suggest that the proposed AAE model significantly enhances the capacity and efficiency of development of the new mols. with specific anticancer properties using the deep generative models.
- 28Vanhaelen, Q.; Lin, Y.-C.; Zhavoronkov, A. The advent of generative chemistry. ACS Med. Chem. Lett. 2020, 11, 1496– 1505, DOI: 10.1021/acsmedchemlett.0c0008828https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtl2jtbfF&md5=847e493bb1da64aeae859a40b031d9c2The Advent of Generative ChemistryVanhaelen, Quentin; Lin, Yen-Chu; Zhavoronkov, AlexACS Medicinal Chemistry Letters (2020), 11 (8), 1496-1505CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)A review. Generative adversarial networks (GANs), first published in 2014, are among the most important concepts in modern artificial intelligence (AI). Bridging deep learning and game theory, GANs are used to generate or "imagine" new objects with desired properties. Since 2016, multiple GANs with reinforcement learning (RL) have been successfully applied in pharmacol. for de novo mol. design. Those techniques aim at a more efficient use of the data and a better exploration of the chem. space. We review recent advances for the generation of novel mols. with desired properties with a focus on the applications of GANs, RL, and related techniques. We also discuss the current limitations and challenges in the new growing field of generative chem.
- 29Xu, M.; Cheng, J.; Liu, Y.; Huang, W. DeepGAN: Generating Molecule for Drug Discovery Based on Generative Adversarial Network. In 2021 IEEE Symposium on Computers and Communications (ISCC) , 2021; pp 1– 6.There is no corresponding record for this reference.
- 30Guimaraes, G. L.; Sanchez-Lengeling, B.; Outeiral, C.; Farias, P. L. C.; Aspuru-Guzik, A. Objective-reinforced generative adversarial networks (ORGAN) for sequence generation models. arXiv preprint , arXiv:1705.10843, 2017.There is no corresponding record for this reference.
- 31Yu, L.; Zhang, W.; Wang, J.; Yu, Y. SeqGAN: sequence generative adversarial nets with policy gradient. In Proceedings of the Thirty-First AAAI Conference on Artificial Intelligence , 2017; pp 2852– 2858.There is no corresponding record for this reference.
- 32Prykhodko, O.; Johansson, S. V.; Kotsias, P.-C.; Arús-Pous, J.; Bjerrum, E. J.; Engkvist, O.; Chen, H. A de novo molecular generation method using latent vector based generative adversarial network. J. Cheminf. 2019, 11, 1– 13, DOI: 10.1186/s13321-019-0397-9There is no corresponding record for this reference.
- 33Kotsias, P.-C.; Arús-Pous, J.; Chen, H.; Engkvist, O.; Tyrchan, C.; Bjerrum, E. J. Direct steering of de novo molecular generation with descriptor conditional recurrent neural networks. Nat. Mach. Intell. 2020, 2, 254– 265, DOI: 10.1038/s42256-020-0174-5There is no corresponding record for this reference.
- 34De Cao, N.; Kipf, T. MolGAN: An implicit generative model for small molecular graphs. arXiv preprint arXiv:1805.11973 2018,.There is no corresponding record for this reference.
- 35Neukart, F.; Compostella, G.; Seidel, C.; Von Dollen, D.; Yarkoni, S.; Parney, B. Traffic flow optimization using a quantum annealer. Front. ICT 2017, 4, 29, DOI: 10.3389/fict.2017.00029There is no corresponding record for this reference.
- 36Harwood, S.; Gambella, C.; Trenev, D.; Simonetto, A.; Bernal Neira, D.; Greenberg, D. Formulating and solving routing problems on quantum computers. IEEE Trans. Quantum Eng. 2021, 2, 1– 17, DOI: 10.1109/TQE.2021.3049230There is no corresponding record for this reference.
- 37Orus, R.; Mugel, S.; Lizaso, E. Quantum computing for finance: Overview and prospects. Rev. Phys. 2019, 4, 100028, DOI: 10.1016/j.revip.2019.100028There is no corresponding record for this reference.
- 38Liu, G.; Ma, W. A quantum artificial neural network for stock closing price prediction. Inf. Sci. (N.Y.) 2022, 598, 75– 85, DOI: 10.1016/j.ins.2022.03.064There is no corresponding record for this reference.
- 39Du, Y.; Yang, Y.; Tao, D.; Hsieh, M.-H. Demystify Problem-Dependent Power of Quantum Neural Networks on Multi-Class Classification. arXiv preprint , arXiv:2301.01597, 2022.There is no corresponding record for this reference.
- 40Yin, X.-F.; Du, Y.; Fei, Y.-Y.; Zhang, R.; Liu, L.-Z.; Mao, Y.; Liu, T.; Hsieh, M.-H.; Li, L.; Liu, N.-L.; Tao, D.; Chen, Y.-A.; Pan, J.-W. Efficient Bipartite Entanglement Detection Scheme with a Quantum Adversarial Solver. Phys. Rev. Lett. 2022, 128, 110501, DOI: 10.1103/PhysRevLett.128.11050140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XpsFKgu74%253D&md5=d13b175a37f194b174270568ff59afc4Efficient Bipartite Entanglement Detection Scheme with a Quantum Adversarial SolverYin, Xu-Fei; Du, Yuxuan; Fei, Yue-Yang; Zhang, Rui; Liu, Li-Zheng; Mao, Yingqiu; Liu, Tongliang; Hsieh, Min-Hsiu; Li, Li; Liu, Nai-Le; Tao, Dacheng; Chen, Yu-Ao; Pan, Jian-WeiPhysical Review Letters (2022), 128 (11), 110501CODEN: PRLTAO; ISSN:1079-7114. (American Physical Society)The recognition of entanglement states is a notoriously difficult problem when no prior information is available. Here, we propose an efficient quantum adversarial bipartite entanglement detection scheme to address this issue. Our proposal reformulates the bipartite entanglement detection as a two-player zero-sum game completed by parameterized quantum circuits, where a two-outcome measurement can be used to query a classical binary result about whether the input state is bipartite entangled or not. In principle, for an N-qubit quantum state, the runtime complexity of our proposal is O(poly(N)T) with T being the no. of iterations. We exptl. implement our protocol on a linear optical network and exhibit its effectiveness to accomplish the bipartite entanglement detection for 5-qubit quantum pure states and 2-qubit quantum mixed states. Our work paves the way for using near-term quantum machines to tackle entanglement detection on multipartite entangled quantum systems.
- 41Rudolph, M. S.; Toussaint, N. B.; Katabarwa, A.; Johri, S.; Peropadre, B.; Perdomo-Ortiz, A. Generation of high-resolution handwritten digits with an ion-trap quantum computer. Phys. Rev. X 2022, 12, 031010, DOI: 10.1103/PhysRevX.12.03101041https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xis1ymurzM&md5=2c6059f94ef9b16a6ea891ad14559e4aGeneration of High-Resolution Handwritten Digits with an Ion-Trap Quantum ComputerRudolph, Manuel S.; Toussaint, Ntwali Bashige; Katabarwa, Amara; Johri, Sonika; Peropadre, Borja; Perdomo-Ortiz, AlejandroPhysical Review X (2022), 12 (3), 031010CODEN: PRXHAE; ISSN:2160-3308. (American Physical Society)Generating high-quality data (e.g., images or video) is one of the most exciting and challenging frontiers in unsupervised machine learning. Utilizing quantum computers in such tasks to potentially enhance conventional machine-learning algorithms has emerged as a promising application but poses big challenges due to the limited no. of qubits and the level of gate noise in available devices. In this work, we provide the first practical and exptl. implementation of a quantum-classical generative algorithm capable of generating high-resoln. images of handwritten digits with state-of-the-art gate-based quantum computers. In our quantum-assisted machine-learning framework, we implement a quantum-circuit-based generative model to learn and sample the prior distribution of a generative adversarial network. We introduce a multibasis technique which leverages the unique possibility of measuring quantum states in different bases, hence enhancing the expressivity of the prior distribution. We train this hybrid algorithm on an ion-trap device based on Yb+171 ion qubits to generate high-quality images and quant. outperform comparable classical generative adversarial networks trained on the popular MNIST dataset for handwritten digits.
- 42Aspuru-Guzik, A.; Dutoi, A. D.; Love, P. J.; Head-Gordon, M. Simulated quantum computation of molecular energies. Science 2005, 309, 1704– 1707, DOI: 10.1126/science.111347942https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpvFCisrg%253D&md5=c11f20a70de762c9a4b933e4a35d0af3Simulated Quantum Computation of Molecular EnergiesAspuru-Guzik, Alan; Dutoi, Anthony D.; Love, Peter J.; Head-Gordon, MartinScience (Washington, DC, United States) (2005), 309 (5741), 1704-1707CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The calcn. time for the energy of atoms and mols. scales exponentially with system size on a classical computer but polynomially using quantum algorithms. We demonstrate that such algorithms can be applied to problems of chem. interest using modest nos. of quantum bits. Calcns. of the water and lithium hydride mol. ground-state energies have been carried out on a quantum computer simulator using a recursive phase-estn. algorithm. The recursive algorithm reduces the no. of quantum bits required for the readout register from about 20 to 4. Mappings of the mol. wave function to the quantum bits are described. An adiabatic method for the prepn. of a good approx. ground-state wave function is described and demonstrated for a stretched hydrogen mol. The no. of quantum bits required scales linearly with the no. of basis functions, and the no. of gates required grows polynomially with the no. of quantum bits.
- 43Lanyon, B. P.; Whitfield, J. D.; Gillett, G. G.; Goggin, M. E.; Almeida, M. P.; Kassal, I.; Biamonte, J. D.; Mohseni, M.; Powell, B. J.; Barbieri, M.; Aspuru-Guzik, A.; White, A. G. Towards quantum chemistry on a quantum computer. Nat. Chem. 2010, 2, 106– 111, DOI: 10.1038/nchem.48343https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXos1Crug%253D%253D&md5=3434a0ba7b2b832ac19cd0e792e7af2fTowards quantum chemistry on a quantum computerLanyon, B. P.; Whitfield, J. D.; Gillett, G. G.; Goggin, M. E.; Almeida, M. P.; Kassal, I.; Biamonte, J. D.; Mohseni, M.; Powell, B. J.; Barbieri, M.; Aspuru-Guzik, A.; White, A. G.Nature Chemistry (2010), 2 (2), 106-111CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Exact first-principles calcns. of mol. properties are currently intractable because their computational cost grows exponentially with both the no. of atoms and basis set size. A soln. is to move to a radically different model of computing by building a quantum computer, which is a device that uses quantum systems themselves to store and process data. Here we report the application of the latest photonic quantum computer technol. to calc. properties of the smallest mol. system: the mol. in a minimal basis. We calc. the complete energy spectrum to 20 bits of precision and discuss how the technique can be expanded to solve large-scale chem. problems that lie beyond the reach of modern supercomputers. These results represent an early practical step toward a powerful tool with a broad range of quantum-chem. applications.
- 44Peruzzo, A.; McClean, J.; Shadbolt, P.; Yung, M.-H.; Zhou, X.-Q.; Love, P. J.; Aspuru-Guzik, A.; O’brien, J. L. A variational eigenvalue solver on a photonic quantum processor. Nat. Commun. 2014, 5, 1– 7, DOI: 10.1038/ncomms5213There is no corresponding record for this reference.
- 45O’Malley, P. J. J.; Babbush, R.; Kivlichan, I. D.; Romero, J.; McClean, J. R.; Barends, R.; Kelly, J.; Roushan, P.; Tranter, A.; Ding, N.; Campbell, B.; Chen, Y.; Chen, Z.; Chiaro, B.; Dunsworth, A.; Fowler, A. G.; Jeffrey, E.; Lucero, E.; Megrant, A.; Mutus, J. Y.; Neeley, M.; Neill, C.; Quintana, C.; Sank, D.; Vainsencher, A.; Wenner, J.; White, T. C.; Coveney, P. V.; Love, P. J.; Neven, H.; Aspuru-Guzik, A.; Martinis, J. M. Scalable quantum simulation of molecular energies. Phys. Rev. X 2016, 6, 031007, DOI: 10.1103/PhysRevX.6.03100745https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslyrtbvF&md5=750ce9fcb52cba1233a0b592d09142e3Scalable quantum simulation of molecular energiesO'Malley, P. J. J.; Babbush, R.; Kivlichan, I. D.; Romero, J.; McClean, J. R.; Barends, R.; Kelly, J.; Roushan, P.; Tranter, A.; Ding, N.; Campbell, B.; Chen, Y.; Chen, Z.; Chiaro, B.; Dunsworth, A.; Fowler, A. G.; Jeffrey, E.; Lucero, E.; Megrant, A.; Mutus, J. Y.; Neeley, M.; Neill, C.; Quintana, C.; Sank, D.; Vainsencher, A.; Wenner, J.; White, T. C.; Coveney, P. V.; Love, P. J.; Neven, H.; Aspuru-Guzik, A.; Martinis, J. M.Physical Review X (2016), 6 (3), 031007/1-031007/13CODEN: PRXHAE; ISSN:2160-3308. (American Physical Society)We report the first electronic structure calcn. performed on a quantum computer without exponentially costly precompilation. We use a programmable array of superconducting qubits to compute the energy surface of mol. hydrogen using two distinct quantum algorithms. First, we exptl. execute the unitary coupled cluster method using the variational quantum eigensolver. Our efficient implementation predicts the correct dissocn. energy to within chem. accuracy of the numerically exact result. Second, we exptl. demonstrate the canonical quantum algorithm for chem., which consists of Trotterization and quantum phase estn. We compare the exptl. performance of these approaches to show clear evidence that the variational quantum eigensolver is robust to certain errors. This error tolerance inspires hope that variational quantum simulations of classically intractable mols. may be viable in the near future.
- 46Kandala, A.; Mezzacapo, A.; Temme, K.; Takita, M.; Brink, M.; Chow, J. M.; Gambetta, J. M. Hardware-efficient variational quantum eigensolver for small molecules and quantum magnets. Nature 2017, 549, 242– 246, DOI: 10.1038/nature2387946https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsV2isLjP&md5=4315a0cb0cda7fb07584cacd895b576fHardware-efficient variational quantum eigensolver for small molecules and quantum magnetsKandala, Abhinav; Mezzacapo, Antonio; Temme, Kristan; Takita, Maika; Brink, Markus; Chow, Jerry M.; Gambetta, Jay M.Nature (London, United Kingdom) (2017), 549 (7671), 242-246CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Quantum computers can be used to address electronic-structure problems and problems in materials science and condensed matter physics that can be formulated as interacting fermionic problems, problems which stretch the limits of existing high-performance computers. Finding exact solns. to such problems numerically has a computational cost that scales exponentially with the size of the system, and Monte Carlo methods are unsuitable owing to the fermionic sign problem. These limitations of classical computational methods have made solving even few-atom electronic-structure problems interesting for implementation using medium-sized quantum computers. Yet exptl. implementations have so far been restricted to mols. involving only hydrogen and helium. Here we demonstrate the exptl. optimization of Hamiltonian problems with up to six qubits and more than one hundred Pauli terms, detg. the ground-state energy for mols. of increasing size, up to BeH2. We achieve this result by using a variational quantum eigenvalue solver (eigensolver) with efficiently prepd. trial states that are tailored specifically to the interactions that are available in our quantum processor, combined with a compact encoding of fermionic Hamiltonians and a robust stochastic optimization routine. We demonstrate the flexibility of our approach by applying it to a problem of quantum magnetism, an antiferromagnetic Heisenberg model in an external magnetic field. In all cases, we find agreement between our expts. and numerical simulations using a model of the device with noise. Our results help to elucidate the requirements for scaling the method to larger systems and for bridging the gap between key problems in high-performance computing and their implementation on quantum hardware.
- 47Cao, Y.; Romero, J.; Olson, J. P.; Degroote, M.; Johnson, P. D.; Kieferová, M.; Kivlichan, I. D.; Menke, T.; Peropadre, B.; Sawaya, N. P. D.; Sim, S.; Veis, L.; Aspuru-Guzik, A. Quantum Chemistry in the Age of Quantum Computing. Chem. Rev. 2019, 119, 10856– 10915, DOI: 10.1021/acs.chemrev.8b0080347https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1Krtb7K&md5=42620699778f2ef25d3f6958b8d4e776Quantum Chemistry in the Age of Quantum ComputingCao, Yudong; Romero, Jonathan; Olson, Jonathan P.; Degroote, Matthias; Johnson, Peter D.; Kieferova, Maria; Kivlichan, Ian D.; Menke, Tim; Peropadre, Borja; Sawaya, Nicolas P. D.; Sim, Sukin; Veis, Libor; Aspuru-Guzik, AlanChemical Reviews (Washington, DC, United States) (2019), 119 (19), 10856-10915CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Practical challenges in simulating quantum systems on classical computers have been widely recognized in the quantum physics and quantum chem. communities over the past century. Although many approxn. methods have been introduced, the complexity of quantum mechanics remains hard to appease. The advent of quantum computation brings new pathways to navigate this challenging complexity landscape. By manipulating quantum states of matter and taking advantage of their unique features such as superposition and entanglement, quantum computers promise to efficiently deliver accurate results for many important problems in quantum chem. such as the electronic structure of mols. In the past two decades significant advances have been made in developing algorithms and phys. hardware for quantum computing, heralding a revolution in simulation of quantum systems. This article is an overview of the algorithms and results that are relevant for quantum chem. The intended audience is both quantum chemists who seek to learn more about quantum computing, and quantum computing researchers who would like to explore applications in quantum chem.
- 48Quantum, G. A.; Collaborators*†; Arute, F.; Arya, K.; Babbush, R.; Bacon, D.; Bardin, J. C.; Barends, R.; Boixo, S.; Broughton, M. Hartree-Fock on a superconducting qubit quantum computer. Science 2020, 369, 1084– 1089, DOI: 10.1126/science.abb9811There is no corresponding record for this reference.
- 49Gao, Q.; Nakamura, H.; Gujarati, T. P.; Jones, G. O.; Rice, J. E.; Wood, S. P.; Pistoia, M.; Garcia, J. M.; Yamamoto, N. Computational investigations of the lithium superoxide dimer rearrangement on noisy quantum devices. J. Phys. Chem. A 2021, 125, 1827– 1836, DOI: 10.1021/acs.jpca.0c0953049https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXlt1ShsbY%253D&md5=233ec03e41ecf3978d94c885afcf1e1cComputational Investigations of the Lithium Superoxide Dimer Rearrangement on Noisy Quantum DevicesGao, Qi; Nakamura, Hajime; Gujarati, Tanvi P.; Jones, Gavin O.; Rice, Julia E.; Wood, Stephen P.; Pistoia, Marco; Garcia, Jeannette M.; Yamamoto, NaokiJournal of Physical Chemistry A (2021), 125 (9), 1827-1836CODEN: JPCAFH; ISSN:1089-5639. (American Chemical Society)Quantum chem. studies of biradical systems are challenging due to the required multiconfigurational nature of the wavefunction. In this work, Variational Quantum Eigensolver (VQE) is used to compute the energy profile for the lithium superoxide dimer rearrangement, involving biradical species, on quantum simulators and devices. Considering that current quantum devices can only handle limited no. of qubits, we present guidelines for selecting an appropriate active space to perform computations on chem. systems that require many qubits. We show that with VQE performed with a quantum simulator reproduces results obtained with full-CI (Full CI) for the chosen active space. However, results deviate from exact values by about 39 mHa for calcns. on a quantum device. This deviation can be improved to about 4 mHa using the readout mitigation approach and can be further improved to 2 mHa, approaching chem. accuracy, using the state tomog. technique to purify the calcd. quantum state.
- 50Shee, Y.; Yeh, T.-L.; Hsiao, J.-Y.; Yang, A.; Lin, Y.-C.; Hsieh, M.-H. Quantum Simulation of Preferred Tautomeric State Prediction. arXiv preprint , arXiv:2210.02977, 2022.There is no corresponding record for this reference.
- 51Cao, Y.; Romero, J.; Aspuru-Guzik, A. Potential of quantum computing for drug discovery. IBM J. Res. Dev. 2018, 62, 1– 20, DOI: 10.1147/JRD.2018.2888987There is no corresponding record for this reference.
- 52Li, J.; Topaloglu, R. O.; Ghosh, S. Quantum generative models for small molecule drug discovery. IEEE Trans. Quantum Eng. 2021, 2, 1– 8, DOI: 10.1109/TQE.2021.3104804There is no corresponding record for this reference.
- 53Bharti, K.; Cervera-Lierta, A.; Kyaw, T. H.; Haug, T.; Alperin-Lea, S.; Anand, A.; Degroote, M.; Heimonen, H.; Kottmann, J. S.; Menke, T.; Mok, W.-K.; Sim, S.; Kwek, L.-C.; Aspuru-Guzik, A. Noisy intermediate-scale quantum algorithms. Rev. Mod. Phys. 2022, 94, 015004, DOI: 10.1103/RevModPhys.94.01500453https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtVOnsLrE&md5=1d31a740165d58c2a52b3baa15f9f9a1Noisy intermediate-scale quantum algorithmsBharti, Kishor; Cervera-Lierta, Alba; Kyaw, Thi Ha; Haug, Tobias; Alperin-Lea, Sumner; Anand, Abhinav; Degroote, Matthias; Heimonen, Hermanni; Kottmann, Jakob S.; Menke, Tim; Mok, Wai-Keong; Sim, Sukin; Kwek, Leong-Chuan; Aspuru-Guzik, AlanReviews of Modern Physics (2022), 94 (1), 015004CODEN: RMPHAT; ISSN:1539-0756. (American Physical Society)A review. A universal fault-tolerant quantum computer that can efficiently solve problems such as integer factorization and unstructured database search requires millions of qubits with low error rates and long coherence times. While the exptl. advancement toward realizing such devices will potentially take decades of research, noisy intermediate-scale quantum (NISQ) computers already exist. These computers are composed of hundreds of noisy qubits, i.e., qubits that are not error cor., and therefore perform imperfect operations within a limited coherence time. In the search for achieving quantum advantage with these devices, algorithms have been proposed for applications in various disciplines spanning physics, machine learning, quantum chem., and combinatorial optimization. The overarching goal of such algorithms is to leverage the limited available resources to perform classically challenging tasks. In this review, a thorough summary of NISQ computational paradigms and algorithms is provided. The key structure of these algorithms and their limitations and advantages are discussed. A comprehensive overview of various benchmarking and software tools useful for programming and testing NISQ devices is addnl. provided.
- 54Du, Y.; Hsieh, M.-H.; Liu, T.; Tao, D. Expressive power of parametrized quantum circuits. Phys. Rev. Res. 2020, 2, 033125, DOI: 10.1103/PhysRevResearch.2.03312554https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitV2gs7nE&md5=288c5ca7b2455ce8389ffe97a441b449Expressive power of parametrized quantum circuitsDu, Yuxuan; Hsieh, Min-Hsiu; Liu, Tongliang; Tao, DachengPhysical Review Research (2020), 2 (3), 033125CODEN: PRRHAI; ISSN:2643-1564. (American Physical Society)Parametrized quantum circuits (PQCs) have been broadly used as a hybrid quantum-classical machine learning scheme to accomplish generative tasks. However, whether PQCs have better expressive power than classical generative neural networks, such as restricted or deep Boltzmann machines, remains an open issue. In this paper, we prove that PQCs with a simple structure already outperform any classical neural network for generative tasks, unless the polynomial hierarchy collapses. Our proof builds on known results from tensor networks and quantum circuits (in particular, instantaneous quantum polynomial circuits). In addn., PQCs equipped with ancillary qubits for postselection may possess expressive power stronger than that of those without postselection. We employ them as an application for Bayesian learning, since it is possible to learn prior probabilities rather than assuming they are known. We expect that it will find many more applications in semisupervised learning where prior distributions are normally assumed to be unknown. Lastly, we conduct several numerical expts. using the Rigetti Forest platform to demonstrate the performance of the proposed Bayesian quantum circuit.
- 55Du, Y.; Hsieh, M.-H.; Liu, T.; You, S.; Tao, D. Learnability of quantum neural networks. PRX Quantum 2021, 2, 040337, DOI: 10.1103/PRXQuantum.2.040337There is no corresponding record for this reference.
- 56Du, Y.; Hsieh, M.-H.; Liu, T.; Tao, D.; Liu, N. Quantum noise protects quantum classifiers against adversaries. Phys. Rev. Res. 2021, 3, 023153, DOI: 10.1103/PhysRevResearch.3.02315356https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFSgtbvI&md5=56ed26f0c7b485611e974308cddcbee8Quantum noise protects quantum classifiers against adversariesDu, Yuxuan; Hsieh, Min-Hsiu; Liu, Tongliang; Tao, Dacheng; Liu, NanaPhysical Review Research (2021), 3 (2), 023153CODEN: PRRHAI; ISSN:2643-1564. (American Physical Society)Noise in quantum information processing is often viewed as a disruptive and difficult-to-avoid feature, esp. in near-term quantum technologies. However, noise has often played beneficial roles, from enhancing weak signals in stochastic resonance to protecting the privacy of data in differential privacy. It is then natural to ask: Can we harness the power of quantum noise that is beneficial to quantum computing. An important current direction for quantum computing is its application to machine learning, such as classification problems. One outstanding problem in machine learning for classification is its sensitivity to adversarial examples. These are small, undetectable perturbations from the original data where the perturbed data is completely misclassified in otherwise extremely accurate classifiers. They can also be considered as worst-case perturbations by unknown noise sources. We show that by taking advantage of depolarization noise in quantum circuits for classification, a robustness bound against adversaries can be derived where the robustness improves with increasing noise. This robustness property is intimately connected with an important security concept called differential privacy, which can be extended to quantum differential privacy. For the protection of quantum data, this quantum protocol can be used against the most general adversaries. Furthermore, we show how the robustness in the classical case can be sensitive to the details of the classification model, but in the quantum case the details of the classification model are absent, thus also providing a potential quantum advantage for classical data. This opens the opportunity to explore other ways in which quantum noise can be used in our favor, as well as identifying other ways quantum algorithms can be helpful in a way which is distinct from quantum speedups.
- 57Lloyd, S.; Weedbrook, C. Quantum generative adversarial learning. Phys. Rev. Lett. 2018, 121, 040502, DOI: 10.1103/PhysRevLett.121.04050257https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXltFSjs7g%253D&md5=c7b5965318a3af921f7e97e9e243cbcaQuantum Generative Adversarial LearningLloyd, Seth; Weedbrook, ChristianPhysical Review Letters (2018), 121 (4), 040502CODEN: PRLTAO; ISSN:1079-7114. (American Physical Society)Generative adversarial networks represent a powerful tool for classical machine learning: a generator tries to create statistics for data that mimics those of a true data set, while a discriminator tries to discriminate between the true and fake data. The learning process for generator and discriminator can be thought of as an adversarial game, and under reasonable assumptions, the game converges to the point where the generator generates the same statistics as the true data and the discriminator is unable to discriminate between the true and the generated data. This Letter introduces the notion of quantum generative adversarial networks, where the data consist either of quantum states or of classical data, and the generator and discriminator are equipped with quantum information processors. We show that the unique fixed point of the quantum adversarial game also occurs when the generator produces the same statistics as the data. Neither the generator nor the discriminator perform quantum tomog.; linear programing drives them to the optimal. Since quantum systems are intrinsically probabilistic, the proof of the quantum case is different from-and simpler than-the classical case. We show that, when the data consist of samples of measurements made on high-dimensional spaces, quantum adversarial networks may exhibit an exponential advantage over classical adversarial networks.
- 58Huang, H.-L.; Du, Y.; Gong, M.; Zhao, Y.; Wu, Y.; Wang, C.; Li, S.; Liang, F.; Lin, J.; Xu, Y.; Yang, R.; Liu, T.; Hsieh, M.-H.; Deng, H.; Rong, H.; Peng, C.-Z.; Lu, C.-Y.; Chen, Y.-A.; Tao, D.; Zhu, X.; Pan, J.-W. Experimental quantum generative adversarial networks for image generation. Phys. Rev. Appl. 2021, 16, 024051, DOI: 10.1103/PhysRevApplied.16.02405158https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVekurvF&md5=f8aa2cb8ed6846dc955f570486b24cdaExperimental Quantum Generative Adversarial Networks for Image GenerationHuang, He-Liang; Du, Yuxuan; Gong, Ming; Zhao, Youwei; Wu, Yulin; Wang, Chaoyue; Li, Shaowei; Liang, Futian; Lin, Jin; Xu, Yu; Yang, Rui; Liu, Tongliang; Hsieh, Min-Hsiu; Deng, Hui; Rong, Hao; Peng, Cheng-Zhi; Lu, Chao-Yang; Chen, Yu-Ao; Tao, Dacheng; Zhu, Xiaobo; Pan, Jian-WeiPhysical Review Applied (2021), 16 (2), 024051CODEN: PRAHB2; ISSN:2331-7019. (American Physical Society)Quantum machine learning is expected to be one of the first practical applications of near-term quantum devices. Pioneer theor. works suggest that quantum generative adversarial networks (GANs) may exhibit a potential exponential advantage over classical GANs, thus attracting widespread attention. However, it remains elusive whether quantum GANs implemented on near-term quantum devices can actually solve real-world learning tasks. Here, we devise a flexible quantum GAN scheme to narrow this knowledge gap. In principle, this scheme has the ability to complete image generation with high-dimensional features and could harness quantum superposition to train multiple examples in parallel. We exptl. achieve the learning and generating of real-world handwritten digit images on a superconducting quantum processor. Moreover, we utilize a gray-scale bar dataset to exhibit competitive performance between quantum GANs and the classical GANs based on multilayer perceptron and convolutional neural network architectures, resp., benchmarked by the Fr´echet distance score. Our work provides guidance for developing advanced quantum generative models on near-term quantum devices and opens up an avenue for exploring quantum advantages in various GAN-related learning tasks.
- 59Dallaire-Demers, P.-L.; Killoran, N. Quantum generative adversarial networks. Phys. Rev. A 2018, 98, 012324, DOI: 10.1103/PhysRevA.98.01232459https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXlsV2ht7Y%253D&md5=9c61c6048cbf7251849f2fb0d689c3bbQuantum generative adversarial networksDallaire-Demers, Pierre-Luc; Killoran, NathanPhysical Review A (2018), 98 (1), 012324CODEN: PRAHC3; ISSN:2469-9934. (American Physical Society)Quantum machine learning is expected to be one of the first potential general-purpose applications of near-term quantum devices. A major recent breakthrough in classical machine learning is the notion of generative adversarial training, where the gradients of a discriminator model are used to train a sep. generative model. In this work and a companion paper, we extend adversarial training to the quantum domain and show how to construct generative adversarial networks using quantum circuits. Furthermore, we also show how to compute gradients-a key element in generative adversarial network training-using another quantum circuit. We give an example of a simple practical circuit ansatz to parametrize quantum machine learning models and perform a simple numerical expt. to demonstrate that quantum generative adversarial networks can be trained successfully.
- 60Romero, J.; Aspuru-Guzik, A. Variational quantum generators: Generative adversarial quantum machine learning for continuous distributions. Adv. Quantum Technol. 2021, 4, 2000003, DOI: 10.1002/qute.202000003There is no corresponding record for this reference.
- 61Li, J. Quantum GAN with Hybrid Generator. https://github.com/jundeli/quantum-gan, accessed Aug. 2, 2021.There is no corresponding record for this reference.
- 62Ivanenkov, Y. A.; Polykovskiy, D.; Bezrukov, D.; Zagribelnyy, B.; Aladinskiy, V.; Kamya, P.; Aliper, A.; Ren, F.; Zhavoronkov, A. Chemistry42: An AI-Driven Platform for Molecular Design and Optimization. J. Chem. Inf. Model. 2023, 63, 695– 701, DOI: 10.1021/acs.jcim.2c0119162https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXit1yjsrc%253D&md5=04066dad851fb49f198d9c7e4007fc02Chemistry42: An AI-Driven Platform for Molecular Design and OptimizationIvanenkov, Yan A.; Polykovskiy, Daniil; Bezrukov, Dmitry; Zagribelnyy, Bogdan; Aladinskiy, Vladimir; Kamya, Petrina; Aliper, Alex; Ren, Feng; Zhavoronkov, AlexJournal of Chemical Information and Modeling (2023), 63 (3), 695-701CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Chem.42 is a software platform for de novo small mol. design and optimization that integrates Artificial Intelligence (AI) techniques with computational and medicinal chem. methodologies. Chem.42 efficiently generates novel mol. structures with optimized properties validated in both in vitro and in vivo studies and is available through licensing or collaboration. Chem.42 is the core component of Insilico Medicine's Pharma.ai drug discovery suite. Pharma.ai also includes PandaOmics for target discovery and multiomics data anal., and inClinico-a data-driven multimodal forecast of a clin. trial's probability of success (PoS). In this paper, we demonstrate how the platform can be used to efficiently find novel mol. structures against DDR1 and CDK20.
- 63Bergholm, V.; Izaac, J.; Schuld, M.; Gogolin, C.; Ahmed, S.; Ajith, V.; Alam, M. S.; Alonso-Linaje, G.; AkashNarayanan, B.; Asadi, A.; Arrazola, J. M.; Azad, U.; Banning, S.; Blank, C.; Bromley, T. R.; Cordier, B. A.; Ceroni, J.; Delgado, A.; Di Matteo, O.; Dusko, A.; Garg, T.; Guala, D.; Hayes, A.; Hill, R.; Ijaz, A.; Isacsson, T.; Ittah, D.; Jahangiri, S.; Jain, P.; Jiang, E.; Khandelwal, A.; Kottmann, K.; Lang, R. A.; Lee, C.; Loke, T.; Lowe, A.; McKiernan, K.; Meyer, J. J.; Montañez-Barrera, J. A.; Moyard, R.; Niu, Z.; O’Riordan, L. J.; Oud, S.; Panigrahi, A.; Park, C.-Y.; Polatajko, D.; Quesada, N.; Roberts, C.; Sá, N.; Schoch, I.; Shi, B.; Shu, S.; Sim, S.; Singh, A.; Strandberg, I.; Soni, J.; Száva, A.; Thabet, S.; Vargas-Hernández, R. A.; Vincent, T.; Vitucci, N.; Weber, M.; Wierichs, D.; Wiersema, R.; Willmann, M.; Wong, V.; Zhang, S.; Killoran, N. Pennylane: Automatic differentiation of hybrid quantum-classical computations. arXiv preprint , arXiv:1811.04968, 2018.There is no corresponding record for this reference.
- 64Paszke, A.; Gross, S.; Chintala, S.; Chanan, G.; Yang, E.; DeVito, Z.; Lin, Z.; Desmaison, A.; Antiga, L.; Lerer, A. Automatic differentiation in pytorch. Presented at the NIPS 2017 Autodiff Workshop , 2017.There is no corresponding record for this reference.
- 65Arjovsky, M.; Chintala, S.; Bottou, L. Wasserstein generative adversarial networks. In International Conference on Machine Learning , 2017; pp 214 223.There is no corresponding record for this reference.
- 66Brown, N.; Fiscato, M.; Segler, M. H.; Vaucher, A. C. GuacaMol: benchmarking models for de novo molecular design. J. Chem. Inf. Model. 2019, 59, 1096– 1108, DOI: 10.1021/acs.jcim.8b0083966https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXltVWrsbY%253D&md5=d3fb616b81a4b146cf77950a1c92e4d1GuacaMol: Benchmarking Models for de Novo Molecular DesignBrown, Nathan; Fiscato, Marco; Segler, Marwin H. S.; Vaucher, Alain C.Journal of Chemical Information and Modeling (2019), 59 (3), 1096-1108CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)De novo design seeks to generate mols. with required property profiles by virtual design-make-test cycles. With the emergence of deep learning and neural generative models in many application areas, models for mol. design based on neural networks appeared recently and show promising results. However, the new models have not been profiled on consistent tasks, and comparative studies to well-established algorithms have only seldom been performed. To standardize the assessment of both classical and neural models for de novo mol. design, we propose an evaluation framework, GuacaMol, based on a suite of standardized benchmarks. The benchmark tasks encompass measuring the fidelity of the models to reproduce the property distribution of the training sets, the ability to generate novel mols., the exploration and exploitation of chem. space, and a variety of single and multiobjective optimization tasks. The benchmarking open-source Python code and a leaderboard can be found on https://benevolent.ai/guacamol.
- 67Kaelbling, L. P.; Littman, M. L.; Moore, A. W. Reinforcement learning: A survey. J. Artif. Intell. Res. 1996, 4, 237– 285, DOI: 10.1613/jair.301There is no corresponding record for this reference.
- 68Samanta, B.; De, A.; Ganguly, N.; Gomez-Rodriguez, M. Designing random graph models using variational autoencoders with applications to chemical design. arXiv preprint , arXiv:1802.05283, 2018.There is no corresponding record for this reference.
- 69Polykovskiy, D.; Zhebrak, A.; Sanchez-Lengeling, B.; Golovanov, S.; Tatanov, O.; Belyaev, S.; Kurbanov, R.; Artamonov, A.; Aladinskiy, V.; Veselov, M.; Kadurin, A.; Johansson, S.; Chen, H.; Nikolenko, S.; Aspuru-Guzik, A.; Zhavoronkov, A. Molecular sets (MOSES): a benchmarking platform for molecular generation models. Front. Pharmacol. 2020, 11, 565644, DOI: 10.3389/fphar.2020.56564469https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsl2isr4%253D&md5=bf849713f33d3c7e4b931ca98c5d0b6bMolecular sets (MOSES): a benchmarking platform for molecular generation modelsPolykovskiy, Daniil; Zhebrak, Alexander; Sanchez-Lengeling, Benjamin; Golovanov, Sergey; Tatanov, Oktai; Belyaev, Stanislav; Kurbanov, Rauf; Artamonov, Aleksey; Aladinskiy, Vladimir; Veselov, Mark; Kadurin, Artur; Johansson, Simon; Chen, Hongming; Nikolenko, Sergey; Aspuru-Guzik, Alan; Zhavoronkov, AlexFrontiers in Pharmacology (2020), 11 (), 565644CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)Generative models are becoming a tool of choice for exploring the mol. space. These models learn on a large training dataset and produce novel mol. structures with similar properties. Generated structures can be utilized for virtual screening or training semi-supervized predictive models in the downstream tasks. While there are plenty of generative models, it is unclear how to compare and rank them. In this work, we introduce a benchmarking platform called Mol. Sets (MOSES) to standardize training and comparison of mol. generative models. MOSES provides training and testing datasets, and a set of metrics to evaluate the quality and diversity of generated structures. We have implemented and compared several mol. generation models and suggest to use our results as ref. points for further advancements in generative chem. research. The platform and source code are available at https://github.com/molecularsets/moses.
- 70Bickerton, G. R.; Paolini, G. V.; Besnard, J.; Muresan, S.; Hopkins, A. L. Quantifying the chemical beauty of drugs. Nat. Chem. 2012, 4, 90– 98, DOI: 10.1038/nchem.124370https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1aktLk%253D&md5=7ab125d4ab381924f965d072695f7432Quantifying the chemical beauty of drugsBickerton, G. Richard; Paolini, Gaia V.; Besnard, Jeremy; Muresan, Sorel; Hopkins, Andrew L.Nature Chemistry (2012), 4 (2), 90-98CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Drug-likeness is a key consideration when selecting compds. during the early stages of drug discovery. However, evaluation of drug-likeness in abs. terms does not reflect adequately the whole spectrum of compd. quality. More worryingly, widely used rules may inadvertently foster undesirable mol. property inflation as they permit the encroachment of rule-compliant compds. towards their boundaries. We propose a measure of drug-likeness based on the concept of desirability called the quant. est. of drug-likeness (QED). The empirical rationale of QED reflects the underlying distribution of mol. properties. QED is intuitive, transparent, straightforward to implement in many practical settings and allows compds. to be ranked by their relative merit. We extended the utility of QED by applying it to the problem of mol. target druggability assessment by prioritizing a large set of published bioactive compds. The measure may also capture the abstr. notion of aesthetics in medicinal chem.
- 71Wildman, S. A.; Crippen, G. M. Prediction of physicochemical parameters by atomic contributions. J. Chem. Inf. Model. 1999, 39, 868– 873, DOI: 10.1021/ci990307l71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXlt1WjtbY%253D&md5=5eb46da66f7861906be7078f0b7e1b95Prediction of Physicochemical Parameters by Atomic ContributionsWildman, Scott A.; Crippen, Gordon M.Journal of Chemical Information and Computer Sciences (1999), 39 (5), 868-873CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)We present a new atom type classification system for use in atom-based calcn. of partition coeff. (log P) and molar refractivity (MR) designed in part to address published concerns of previous at. methods. The 68 at. contributions to log P have been detd. by fitting an extensive training set of 9920 mols., with r2 = 0.918 and σ = 0.677. A sep. set of 3412 mols. was used for the detn. of contributions to MR with r2 = 0.997 and σ = 1.43. Both calcns. are shown to have high predictive ability.
- 72Ertl, P.; Schuffenhauer, A. Estimation of synthetic accessibility score of drug-like molecules based on molecular complexity and fragment contributions. J. Cheminf. 2009, 1, 1– 11, DOI: 10.1186/1758-2946-1-8There is no corresponding record for this reference.
- 73Landrum, G.; Tosco, P.; Kelley, B.; Sriniker, R.; Gedeck; ; Vianello, R.; Schneider, N.; Kawashima, E.; Dalke, A.; N, D.; Cosgrove, D.; Cole, B.; Swain, M.; Turk, S.; Savelyev, A.; Jones, G.; Vaucher, A.; Wójcikowski, M.; Take, I.; Probst, D.; Ujihara, K.; Scalfani, V. F.; godin, G.; Pahl, A.; Berenger, F.; Varjo, J. L.; Strets, J. P.; Doliath Gavid rdkit/rdkit: 2022_03_1 (Q1 2022) Release. 2022, DOI: 10.5281/zenodo.6388425 .There is no corresponding record for this reference.
- 74Kullback, S.; Leibler, R. A. On information and sufficiency. Ann. Math. Stat. 1951, 22, 79– 86, DOI: 10.1214/aoms/1177729694There is no corresponding record for this reference.
- 75Ramakrishnan, R.; Dral, P. O.; Rupp, M.; Von Lilienfeld, O. A. Quantum chemistry structures and properties of 134 kilo molecules. Sci. Data 2014, 1, 1– 7, DOI: 10.1038/sdata.2014.22There is no corresponding record for this reference.
- 76Ruddigkeit, L.; Van Deursen, R.; Blum, L. C.; Reymond, J.-L. Enumeration of 166 billion organic small molecules in the chemical universe database GDB-17. J. Chem. Inf. Model. 2012, 52, 2864– 2875, DOI: 10.1021/ci300415d76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFClsL3J&md5=d0bf9a29f3e9ae1e57bb1c953a562cedEnumeration of 166 Billion Organic Small Molecules in the Chemical Universe Database GDB-17Ruddigkeit, Lars; van Deursen, Ruud; Blum, Lorenz C.; Reymond, Jean-LouisJournal of Chemical Information and Modeling (2012), 52 (11), 2864-2875CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Drug mols. consist of a few tens of atoms connected by covalent bonds. How many such mols. are possible in total and what is their structure. This question is of pressing interest in medicinal chem. to help solve the problems of drug potency, selectivity, and toxicity and reduce attrition rates by pointing to new mol. series. To better define the unknown chem. space, we have enumerated 166.4 billion mols. of up to 17 atoms of C, N, O, S, and halogens forming the chem. universe database GDB-17, covering a size range contg. many drugs and typical for lead compds. GDB-17 contains millions of isomers of known drugs, including analogs with high shape similarity to the parent drug. Compared to known mols. in PubChem, GDB-17 mols. are much richer in nonarom. heterocycles, quaternary centers, and stereoisomers, densely populate the third dimension in shape space, and represent many more scaffold types.
- 77Schuld, M.; Petruccione, F. Supervised Learning with Quantum Computers, Vol. 17; Springer, 2018.There is no corresponding record for this reference.
- 78Schuld, M.; Bocharov, A.; Svore, K. M.; Wiebe, N. Circuit-centric quantum classifiers. Phys. Rev. A 2020, 101, 032308, DOI: 10.1103/PhysRevA.101.03230878https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXptVyjsLo%253D&md5=727a05768b62c15944502aaa96467a03Circuit-centric quantum classifiersSchuld, Maria; Bocharov, Alex; Svore, Krysta M.; Wiebe, NathanPhysical Review A (2020), 101 (3), 032308CODEN: PRAHC3; ISSN:2469-9934. (American Physical Society)Variational quantum circuits are becoming tools of choice in quantum optimization and machine learning. In this paper we investigate a class of variational circuits for the purposes of supervised machine learning. We propose a circuit architecture suitable for predicting class labels of quantumly encoded data via measurements of certain observables. We observe that the required depth of a trainable classification circuit is related to the no. of representative principal components of the data distribution. Quantum circuit architectures used in our design are validated by numerical simulation, which shows significant model size redn. compared to classical predictive models. Circuit-based models demonstrate good resilience to noise, which makes then robust and error tolerant.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.3c00562.
Different complexities of generators in MolGAN; different input noise dimensions of the generator in MolGAN-HR; different parametrized layers of QuMolGAN-HR; all the combinations of the classical/quantum noise/generator/discriminator and their corresponding model name; different numbers of qubits in the quantum circuit of QuMolGAN-HR; the details of MolGAN-CC models with the varied size of discriminators; example molecules from the quantum generator; integration of proposed hybrid generative models with Insilico Medicine Chemistry42 (62) platform; example molecules of QM9 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.