10 Cutting-Edge Applications of AI in Drug Discovery

July 21, 2023

Human bodies are phenomenally complex. It takes many years to discover just one new medicine to successfully treat an illness. Can artificial intelligence help speed up this process?

In this blog, we will examine the use of AI in drug discovery, including its applications, benefits and challenges.

1. Accelerating Target Identification

AI can analyze vast amounts of biological and chemical data to identify potential drug targets more efficiently. Machine learning algorithms can learn patterns and predict targets, allowing researchers to focus their efforts on the most promising areas, reducing time and cost. For example, AI is an advanced method to identify novel anti-cancer targets and discover novel drugs from biology networks because the networks can effectively preserve and quantify the interaction between components of cell systems underlying human diseases such as cancer.

2. Virtual Screening

AI-powered virtual screening techniques can efficiently analyze massive chemical libraries to identify molecules that have a high probability of interacting with the target. This saves time and reduces the number of compounds that need to be physically tested in the lab, enabling researchers to focus on the most promising candidates. For example, AI-based Virtual Screening plays a crucial role in screening, enabling efficient analysis and prediction. Here are a few examples of AI-based virtual screening methods:

Machine Learning (ML) Approaches: Machine learning algorithms can be trained on large datasets of known active and inactive compounds to develop predictive models. These models can then be used to screen virtual compound libraries and identify potential drug candidates. Various ML algorithms, such as support vector machines (SVM), random forests, and neural networks, have been employed for virtual screening tasks.

Deep Learning: Deep learning techniques, specifically deep neural networks, have shown promise in virtual screening. Convolutional neural networks (CNNs) can be utilized to analyze molecular structures and extract meaningful features for compound classification. Recurrent neural networks (RNNs) have been employed to model sequential data, such as protein-ligand interactions, for accurate virtual screening predictions.

3. De Novo Drug Design

AI algorithms can generate entirely new molecules with desired properties, such as binding affinity and specificity. By combining deep learning with reinforcement learning, AI can optimize molecular structures to create novel drug candidates, potentially opening up new avenues for therapeutic interventions.

Let's consider the hypothetical scenario of designing a new drug to treat a specific type of cancer.

Step 1: Target Identification

The first step in de novo drug design is to identify a specific target in the cancer cells that plays a crucial role in their growth or survival. In this example, let's say we choose a protein called ‘XYZ’ that is overexpressed in the cancer cells and is known to promote tumor growth.

Step 2: Target Characterization

Next, we need to gather information about the structure and function of the target protein XYZ. This can be done through various experimental techniques such as X-ray crystallography or nuclear magnetic resonance (NMR) spectroscopy. These techniques provide insights into the 3D structure and active sites of the protein.

Step 3: Virtual Screening

Using computational tools, we can perform virtual screening to search for small molecules or compounds that have the potential to bind to the active site of the target protein XYZ. This involves analyzing large databases of chemical compounds and predicting their binding affinity and interactions with the target protein using molecular docking or other computational techniques.

Step 4: Lead Optimization

From the virtual screening, we identify a set of promising compounds that show good binding affinity and interactions with the target protein XYZ. These compounds are referred to as ‘leads’. The next step is to optimize these leads by making modifications to their chemical structure to improve their potency, selectivity, and pharmacokinetic properties. This can be achieved through computer-aided drug design techniques, such as molecular dynamics simulations and quantitative structure-activity relationship (QSAR) analysis.

Step 5: Experimental Validation

The optimized lead compounds are synthesized in the laboratory and tested in vitro and in vivo to evaluate their efficacy, toxicity, and pharmacokinetic profiles. In the case of cancer, they may be tested on cancer cell lines and animal models to assess their ability to inhibit tumor growth and minimize side effects.

Step 6: Clinical Trials

If the lead compounds show promising results in preclinical studies, they can proceed to clinical trials. Clinical trials involve testing the drug candidate on human subjects to determine its safety and effectiveness. The trials are typically conducted in three phases, starting with a small group of healthy volunteers and progressing to larger groups of patients with the target disease.

Step 7: Approval and Market Launch

If the drug candidate successfully completes all phases of clinical trials and demonstrates significant therapeutic benefits with an acceptable safety profile, it can be submitted to regulatory authorities for approval. If approved, the drug can be launched in the market and made available for patients with the specific type of cancer it was designed to treat.

This example provides a general overview of the de novo drug design process. It's important to note that the actual process can be much more complex and may involve additional steps and iterations to optimize the drug candidate before it reaches the market.

4. Predicting Drug-Target Interactions

AI algorithms can predict and model the interactions between drugs and their target proteins. By analyzing large-scale biological data, AI can identify potential side effects, drug-drug interactions, and optimize drug dosages, leading to safer and more effective treatments.

Predicting drug-target interactions is a crucial step in drug discovery and development. There are several computational methods and tools available for this purpose. Here are a few examples of approaches used to predict drug-target interactions:

1. Ligand-based Approaches

   - Quantitative Structure-Activity Relationship (QSAR): This method predicts the activity of a drug based on its chemical structure and compares it to known activity data.

   - Similarity-based Methods: These methods compare the chemical similarity between a drug and known ligands to predict potential targets.

   - Pharmacophore Modeling: It identifies the essential features or spatial arrangement of atoms necessary for a molecule to interact with a specific target.

2. Structure-based Approaches

   - Molecular Docking: It predicts the binding orientation and affinity of a drug molecule with a target protein by analyzing their 3D structures.

   - Molecular Dynamics (MD) Simulations: These simulations model the dynamic behavior of the drug-target complex over time, providing insights into the stability and interactions.

   - Virtual Screening: It involves screening large databases of drug compounds against a target protein using molecular docking or other computational methods.

3. Network-based Approaches

   - Protein-Protein Interaction (PPI) Networks: These networks analyze the interaction patterns between proteins to predict potential drug targets based on their association with disease-related proteins.

   - Network Propagation: It utilizes network topology and known drug-target interactions to propagate information and predict potential interactions.

4. Machine Learning Approaches

   - Supervised Learning: It trains a model using known drug-target interaction data to predict interactions for new drug-target pairs.

   - Deep Learning: Deep neural networks can learn complex patterns from drug and target features to predict interactions.

   - Ensemble Methods: Combining multiple machine learning models can improve prediction accuracy.

It's worth noting that predicting drug-target interactions is a challenging task due to the complexity and diversity of biological systems. Different methods may have their strengths and limitations, and a combination of approaches often yields more accurate predictions. Experimental validation is crucial to confirm the predicted interactions and guide the drug discovery process.

5. Repurposing Existing Drugs

AI can sift through vast amounts of data, including scientific literature and clinical trial results, to identify potential new uses for existing drugs. This can significantly reduce the time and cost required to bring a drug to market since the safety profiles and pharmacokinetics of these drugs are already well understood.

One example of repurposing existing drugs through AI is the identification of potential treatments for COVID-19. When the COVID-19 pandemic hit, researchers and scientists around the world quickly turned to existing drugs to find potential candidates that could be repurposed for the treatment of the virus.

Using AI techniques, researchers were able to analyze vast amounts of data and identify existing drugs that had the potential to target the virus or alleviate its symptoms. By leveraging machine learning algorithms and predictive modeling, researchers could predict how existing drugs might interact with the virus and identify promising candidates for further investigation.

For instance, a study published in the journal Nature in February 2020 used AI algorithms to screen a database of approved drugs and identify 28 potential drug candidates for the treatment of COVID-19. The researchers used a combination of computational models to predict how these drugs might interact with the virus and inhibit its replication.

Another example comes from a UK-based company that uses AI and machine learning to accelerate drug discovery and development. In early 2020, an AI platform analyzed existing drugs to identify potential treatments for COVID-19. Their analysis led to the identification of Baricitinib, an FDA-approved drug used to treat rheumatoid arthritis, as a potential candidate for repurposing against COVID-19. Subsequent clinical trials have shown promising results in using Baricitinib for the treatment of COVID-19 patients.

These examples illustrate how AI can be used to analyze large datasets and identify potential drug candidates for repurposing. By leveraging AI's capabilities for data analysis and predictive modeling, researchers can expedite the drug discovery process and potentially find new uses for existing drugs, leading to faster development of treatments for various diseases.

6. Optimizing Clinical Trials

AI can enhance the design and execution of clinical trials. Machine learning algorithms can analyze patient data to identify suitable participants, predict treatment responses, and optimize trial protocols. This can increase the chances of success and reduce the time and cost associated with clinical trials.

AI can play a crucial role in optimizing clinical trials by improving the efficiency and effectiveness of the trial process. One example of AI in optimizing clinical trials is the use of machine learning algorithms to identify suitable patient populations and streamline the recruitment process.

Patient recruitment is a critical aspect of clinical trials, but it can be challenging and time-consuming. AI algorithms can analyze large amounts of data, such as electronic health records, medical literature, and genetic information, to identify potential participants who meet specific criteria for a clinical trial. By automating the screening process, AI can significantly reduce the time and resources required to identify eligible patients, thus accelerating the recruitment phase.

Additionally, AI can help optimize the trial design and sample size determination. Machine learning algorithms can analyze historical clinical trial data, patient characteristics, and treatment outcomes to generate predictive models. These models can estimate the optimal sample size required for a clinical trial, leading to more efficient trial designs and cost savings.

Another example is the use of AI for real-time monitoring and analysis of trial data. Traditional monitoring of clinical trial data is often done manually and periodically, which can result in delays in detecting adverse events or deviations from the protocol. AI can automate the monitoring process and continuously analyze incoming data, flagging any anomalies or safety concerns in real-time. This enables researchers to respond promptly, ensure participant safety, and make data-driven decisions during the trial.

Furthermore, AI can support the analysis of complex and diverse data sets generated during clinical trials. For example, AI algorithms can analyze genomic data, biomarkers, medical imaging, and patient-reported outcomes to identify patterns, correlations, and treatment responses. This deeper analysis can provide valuable insights into treatment efficacy, patient subgroups, and potential adverse events.

Overall, AI's application in optimizing clinical trials has the potential to enhance patient recruitment, improve trial design, monitor data in real-time, and facilitate data analysis. By leveraging AI's capabilities, researchers can accelerate the drug development process, reduce costs, and bring new treatments to patients more efficiently.

7. Personalized Medicine

AI can analyze individual patient data, including genomics, proteomics, and clinical records, to identify specific biomarkers and develop personalized treatment plans. This targeted approach can lead to more effective and tailored therapies, improving patient outcomes.

Personalized medicine, also known as precision medicine, aims to tailor medical treatments and interventions to individual patients based on their unique characteristics. AI plays a crucial role in enabling personalized medicine by analyzing vast amounts of patient data and generating insights that guide personalized treatment decisions. Here's an example of how AI can facilitate personalized medicine:

Cancer treatment is an area where personalized medicine has shown significant promise, and AI has been instrumental in driving advancements in this field. For instance, AI algorithms can analyze large-scale genomic data from cancer patients, including DNA sequencing and gene expression profiles. By comparing these data with known patterns and databases, AI can identify genetic alterations and mutations associated with specific cancer types or treatment responses.

With this information, AI can help oncologists make more informed decisions about personalized treatment options. For example, AI algorithms can predict the likelihood of a patient's response to a particular chemotherapy drug or targeted therapy based on their genomic profile. This information can guide treatment decisions, such as selecting the most effective drug or combination of drugs for an individual patient.

Furthermore, AI can assist in identifying potential drug targets and developing new targeted therapies. By analyzing molecular and genetic data, AI algorithms can identify specific genetic alterations or molecular pathways that are driving a patient's cancer growth. This information can guide the development of targeted therapies that specifically inhibit those pathways, offering more effective and personalized treatment options.

AI can also help in predicting and managing treatment side effects. By analyzing patient characteristics, treatment history, and other relevant data, AI algorithms can generate models that predict the likelihood and severity of side effects for individual patients. This information can help clinicians proactively adjust treatment plans, mitigate risks, and improve patient outcomes.

Overall, the integration of AI in personalized medicine allows for a more precise and tailored approach to patient care. By leveraging AI's capabilities in analyzing complex and diverse data, clinicians can make more informed decisions about treatment options, identify potential drug targets, and predict treatment outcomes and side effects. This ultimately leads to more effective and personalized treatments that maximize patient outcomes and minimize adverse effects.

8. Side Effect Prediction

AI algorithms can predict the likelihood and severity of side effects associated with drug candidates, aiding in early-stage drug development. By analyzing diverse datasets, including genetics and drug-target interactions, AI can help identify potential safety concerns before they arise during clinical trials.

Side effect prediction is an important aspect of drug development and patient care, as it helps identify potential adverse reactions to medications before they occur. AI can assist in side effect prediction by analyzing large datasets of drug information, patient characteristics, and reported adverse events. Here's an example of how AI can be used for side effect prediction:

In a study published in the journal ‘Science Translational Medicine’, researchers used AI algorithms to predict potential side effects of drugs. They collected data from various sources, including electronic health records, scientific literature, and drug labels. This dataset consisted of information on drugs, their molecular structures, biological targets, and reported adverse events.

The AI algorithms analyzed this extensive dataset to identify patterns and associations between drugs and side effects. By leveraging machine learning techniques, the algorithms learned from the data and developed models to predict the likelihood of specific side effects for a given drug.

For example, the study found that an antidepressant called fluvoxamine had potential anti-inflammatory effects. Based on this discovery, the researchers hypothesized that fluvoxamine might be repurposed for the treatment of inflammatory diseases like sepsis. They conducted a clinical trial and confirmed that fluvoxamine reduced the risk of clinical deterioration in patients with COVID-19.

This example demonstrates how AI can uncover unexpected associations between drugs and side effects, leading to new therapeutic possibilities. By analyzing vast amounts of data and identifying patterns that might not be immediately apparent to humans, AI algorithms can predict potential side effects and inform decision-making in drug development and patient care.

AI-based side effect prediction can also be used to improve medication safety and personalize treatment plans. By analyzing a patient's medical history, genetic information, and other relevant data, AI algorithms can assess the individual's risk factors for specific side effects. This information enables clinicians to make more informed decisions about medication selection, dosages, and monitoring strategies, thus minimizing the occurrence of adverse events and enhancing patient safety.

In summary, AI-driven side effect prediction can aid in identifying potential adverse reactions to drugs, uncover new therapeutic opportunities, and enhance medication safety by personalizing treatment plans. By leveraging AI's capabilities in data analysis and pattern recognition, researchers and clinicians can make more informed decisions and improve patient outcomes.

9. Drug Formulation Optimization

AI can assist in optimizing drug formulations to enhance bioavailability, stability, and efficacy. Machine learning algorithms can analyze the physicochemical properties of molecules to predict their behavior in various formulations, enabling researchers to develop optimal drug delivery systems.

AI can be utilized to optimize drug formulation, enhancing drug efficacy, safety, and patient experience. Here's an example of how AI can be applied in drug formulation optimization:

One area where AI has shown promise is in optimizing drug formulations to improve solubility, stability, and bioavailability. Poor solubility and stability can hinder a drug's effectiveness, while low bioavailability can limit its therapeutic impact. AI algorithms can analyze large databases of chemical and formulation data to identify patterns and correlations that can guide formulation optimization.

For instance, a pharmaceutical company could use AI to analyze the chemical structure of a drug candidate along with information on different excipients and formulation approaches. By considering factors such as molecular interactions, solubility, and stability, AI algorithms can predict the most suitable formulation parameters and identify potential challenges in drug delivery.

Furthermore, AI can help in designing and optimizing drug delivery systems. By considering factors such as drug release profiles, absorption mechanisms, and pharmacokinetics, AI algorithms can guide the development of novel drug delivery systems that improve drug efficacy and patient adherence.

An example of AI-driven formulation optimization is the work conducted by Insilico Medicine, a biotechnology company. They use AI algorithms to design new chemical entities and optimize drug formulations. In one study, Insilico Medicine used AI to develop a new compound that targeted a specific protein involved in cancer growth. The AI algorithms analyzed millions of chemical structures to identify the most promising candidates, which were then synthesized and tested in the laboratory. The resulting compound showed improved efficacy and safety compared to existing drugs.

Moreover, AI can also assist in the optimization of dosage forms, such as tablets or capsules, by considering factors like dissolution profiles, tablet disintegration, and controlled release mechanisms. AI algorithms can analyze formulation data, process parameters, and historical manufacturing data to optimize the formulation process, ensuring consistent quality and performance of the dosage form.

By leveraging AI's capabilities in data analysis, pattern recognition, and predictive modeling, drug formulation optimization can be streamlined and accelerated. AI algorithms can guide the selection of excipients, predict formulation challenges, design novel drug delivery systems, and optimize dosage forms. This enables pharmaceutical companies to develop more effective and patient-friendly formulations, ultimately improving drug efficacy and patient outcomes.

10. Data Integration and Knowledge Management

AI can aid in integrating and analyzing diverse datasets from various sources, including scientific literature, patents, and databases. By extracting valuable insights and patterns from this vast amount of information, AI can facilitate knowledge management, enabling researchers to make more informed decisions.

AI holds immense potential in revolutionizing the drug discovery process. From accelerating target identification to optimizing clinical trials and enabling personalized medicine, the possibilities are vast. Developers, CTOs, and technology enthusiasts can leverage AI to streamline drug discovery, reduce costs, and bring safer and more effective treatments to patients faster than ever before. Embracing AI in the pharmaceutical industry will undoubtedly drive innovation and reshape the future of healthcare.

Data integration and knowledge management are crucial components of AI systems, enabling the organization, integration, and utilization of vast amounts of data from various sources. Here's an example of how AI can facilitate data integration and knowledge management:

In the healthcare industry, there is a wealth of data generated from diverse sources such as electronic health records, medical imaging, genomics, research studies, and scientific literature. Integrating and synthesizing this data is essential for generating meaningful insights and driving evidence-based decision-making.

AI algorithms can analyze and extract information from these disparate data sources, facilitating data integration and knowledge management. For example, natural language processing (NLP) techniques can be used to extract relevant information from medical literature and clinical notes, converting unstructured text into structured data that can be easily analyzed.

By applying machine learning algorithms, AI can learn from and make connections between various data sources, enabling comprehensive knowledge management. For instance, AI systems can analyze patient data, treatment outcomes, and genomics data to identify patterns and correlations that may inform personalized treatment decisions or contribute to medical research.

An example of AI-enabled data integration and knowledge management is the use of AI platforms in drug discovery and development. Companies like Atomwise and BenevolentAI leverage AI to integrate diverse datasets, including molecular structures, chemical properties, and biological assays, to identify potential drug candidates. By integrating and analyzing this data, AI can suggest novel compounds for further investigation, optimizing the drug discovery process.

Furthermore, AI can facilitate knowledge management by providing decision support systems and expert systems. These systems can capture and organize expert knowledge, clinical guidelines, and best practices, making them accessible to healthcare professionals for informed decision-making. AI algorithms can continuously update and refine this knowledge base as new data and insights become available.

Overall, AI plays a vital role in data integration and knowledge management by extracting, integrating, and synthesizing data from diverse sources. By leveraging AI's capabilities in NLP, machine learning, and knowledge representation, organizations can efficiently manage and utilize vast amounts of data, leading to enhanced decision-making, improved patient care, and advancements in research and development.

Learn how to create machine learning models for drug discovery on E2E cloud. Write to sales@e2enetworks.com

Latest Blogs
This is a decorative image for: A Complete Guide To Customer Acquisition For Startups
October 18, 2022

A Complete Guide To Customer Acquisition For Startups

Any business is enlivened by its customers. Therefore, a strategy to constantly bring in new clients is an ongoing requirement. In this regard, having a proper customer acquisition strategy can be of great importance.

So, if you are just starting your business, or planning to expand it, read on to learn more about this concept.

The problem with customer acquisition

As an organization, when working in a diverse and competitive market like India, you need to have a well-defined customer acquisition strategy to attain success. However, this is where most startups struggle. Now, you may have a great product or service, but if you are not in the right place targeting the right demographic, you are not likely to get the results you want.

To resolve this, typically, companies invest, but if that is not channelized properly, it will be futile.

So, the best way out of this dilemma is to have a clear customer acquisition strategy in place.

How can you create the ideal customer acquisition strategy for your business?

  • Define what your goals are

You need to define your goals so that you can meet the revenue expectations you have for the current fiscal year. You need to find a value for the metrics –

  • MRR – Monthly recurring revenue, which tells you all the income that can be generated from all your income channels.
  • CLV – Customer lifetime value tells you how much a customer is willing to spend on your business during your mutual relationship duration.  
  • CAC – Customer acquisition costs, which tells how much your organization needs to spend to acquire customers constantly.
  • Churn rate – It tells you the rate at which customers stop doing business.

All these metrics tell you how well you will be able to grow your business and revenue.

  • Identify your ideal customers

You need to understand who your current customers are and who your target customers are. Once you are aware of your customer base, you can focus your energies in that direction and get the maximum sale of your products or services. You can also understand what your customers require through various analytics and markers and address them to leverage your products/services towards them.

  • Choose your channels for customer acquisition

How will you acquire customers who will eventually tell at what scale and at what rate you need to expand your business? You could market and sell your products on social media channels like Instagram, Facebook and YouTube, or invest in paid marketing like Google Ads. You need to develop a unique strategy for each of these channels. 

  • Communicate with your customers

If you know exactly what your customers have in mind, then you will be able to develop your customer strategy with a clear perspective in mind. You can do it through surveys or customer opinion forms, email contact forms, blog posts and social media posts. After that, you just need to measure the analytics, clearly understand the insights, and improve your strategy accordingly.

Combining these strategies with your long-term business plan will bring results. However, there will be challenges on the way, where you need to adapt as per the requirements to make the most of it. At the same time, introducing new technologies like AI and ML can also solve such issues easily. To learn more about the use of AI and ML and how they are transforming businesses, keep referring to the blog section of E2E Networks.

Reference Links

https://www.helpscout.com/customer-acquisition/

https://www.cloudways.com/blog/customer-acquisition-strategy-for-startups/

https://blog.hubspot.com/service/customer-acquisition

This is a decorative image for: Constructing 3D objects through Deep Learning
October 18, 2022

Image-based 3D Object Reconstruction State-of-the-Art and trends in the Deep Learning Era

3D reconstruction is one of the most complex issues of deep learning systems. There have been multiple types of research in this field, and almost everything has been tried on it — computer vision, computer graphics and machine learning, but to no avail. However, that has resulted in CNN or convolutional neural networks foraying into this field, which has yielded some success.

The Main Objective of the 3D Object Reconstruction

Developing this deep learning technology aims to infer the shape of 3D objects from 2D images. So, to conduct the experiment, you need the following:

  • Highly calibrated cameras that take a photograph of the image from various angles.
  • Large training datasets can predict the geometry of the object whose 3D image reconstruction needs to be done. These datasets can be collected from a database of images, or they can be collected and sampled from a video.

By using the apparatus and datasets, you will be able to proceed with the 3D reconstruction from 2D datasets.

State-of-the-art Technology Used by the Datasets for the Reconstruction of 3D Objects

The technology used for this purpose needs to stick to the following parameters:

  • Input

Training with the help of one or multiple RGB images, where the segmentation of the 3D ground truth needs to be done. It could be one image, multiple images or even a video stream.

The testing will also be done on the same parameters, which will also help to create a uniform, cluttered background, or both.

  • Output

The volumetric output will be done in both high and low resolution, and the surface output will be generated through parameterisation, template deformation and point cloud. Moreover, the direct and intermediate outputs will be calculated this way.

  • Network architecture used

The architecture used in training is 3D-VAE-GAN, which has an encoder and a decoder, with TL-Net and conditional GAN. At the same time, the testing architecture is 3D-VAE, which has an encoder and a decoder.

  • Training used

The degree of supervision used in 2D vs 3D supervision, weak supervision along with loss functions have to be included in this system. The training procedure is adversarial training with joint 2D and 3D embeddings. Also, the network architecture is extremely important for the speed and processing quality of the output images.

  • Practical applications and use cases

Volumetric representations and surface representations can do the reconstruction. Powerful computer systems need to be used for reconstruction.

Given below are some of the places where 3D Object Reconstruction Deep Learning Systems are used:

  • 3D reconstruction technology can be used in the Police Department for drawing the faces of criminals whose images have been procured from a crime site where their faces are not completely revealed.
  • It can be used for re-modelling ruins at ancient architectural sites. The rubble or the debris stubs of structures can be used to recreate the entire building structure and get an idea of how it looked in the past.
  • They can be used in plastic surgery where the organs, face, limbs or any other portion of the body has been damaged and needs to be rebuilt.
  • It can be used in airport security, where concealed shapes can be used for guessing whether a person is armed or is carrying explosives or not.
  • It can also help in completing DNA sequences.

So, if you are planning to implement this technology, then you can rent the required infrastructure from E2E Networks and avoid investing in it. And if you plan to learn more about such topics, then keep a tab on the blog section of the website

Reference Links

https://tongtianta.site/paper/68922

https://github.com/natowi/3D-Reconstruction-with-Deep-Learning-Methods

This is a decorative image for: Comprehensive Guide to Deep Q-Learning for Data Science Enthusiasts
October 18, 2022

A Comprehensive Guide To Deep Q-Learning For Data Science Enthusiasts

For all data science enthusiasts who would love to dig deep, we have composed a write-up about Q-Learning specifically for you all. Deep Q-Learning and Reinforcement learning (RL) are extremely popular these days. These two data science methodologies use Python libraries like TensorFlow 2 and openAI’s Gym environment.

So, read on to know more.

What is Deep Q-Learning?

Deep Q-Learning utilizes the principles of Q-learning, but instead of using the Q-table, it uses the neural network. The algorithm of deep Q-Learning uses the states as input and the optimal Q-value of every action possible as the output. The agent gathers and stores all the previous experiences in the memory of the trained tuple in the following order:

State> Next state> Action> Reward

The neural network training stability increases using a random batch of previous data by using the experience replay. Experience replay also means the previous experiences stocking, and the target network uses it for training and calculation of the Q-network and the predicted Q-Value. This neural network uses openAI Gym, which is provided by taxi-v3 environments.

Now, any understanding of Deep Q-Learning   is incomplete without talking about Reinforcement Learning.

What is Reinforcement Learning?

Reinforcement is a subsection of ML. This part of ML is related to the action in which an environmental agent participates in a reward-based system and uses Reinforcement Learning to maximize the rewards. Reinforcement Learning is a different technique from unsupervised learning or supervised learning because it does not require a supervised input/output pair. The number of corrections is also less, so it is a highly efficient technique.

Now, the understanding of reinforcement learning is incomplete without knowing about Markov Decision Process (MDP). MDP is involved with each state that has been presented in the results of the environment, derived from the state previously there. The information which composes both states is gathered and transferred to the decision process. The task of the chosen agent is to maximize the awards. The MDP optimizes the actions and helps construct the optimal policy.

For developing the MDP, you need to follow the Q-Learning Algorithm, which is an extremely important part of data science and machine learning.

What is Q-Learning Algorithm?

The process of Q-Learning is important for understanding the data from scratch. It involves defining the parameters, choosing the actions from the current state and also choosing the actions from the previous state and then developing a Q-table for maximizing the results or output rewards.

The 4 steps that are involved in Q-Learning:

  1. Initializing parameters – The RL (reinforcement learning) model learns the set of actions that the agent requires in the state, environment and time.
  2. Identifying current state – The model stores the prior records for optimal action definition for maximizing the results. For acting in the present state, the state needs to be identified and perform an action combination for it.
  3. Choosing the optimal action set and gaining the relevant experience – A Q-table is generated from the data with a set of specific states and actions, and the weight of this data is calculated for updating the Q-Table to the following step.
  4. Updating Q-table rewards and next state determination – After the relevant experience is gained and agents start getting environmental records. The reward amplitude helps to present the subsequent step.  

In case the Q-table size is huge, then the generation of the model is a time-consuming process. This situation requires Deep Q-learning.

Hopefully, this write-up has provided an outline of Deep Q-Learning and its related concepts. If you wish to learn more about such topics, then keep a tab on the blog section of the E2E Networks website.

Reference Links

https://analyticsindiamag.com/comprehensive-guide-to-deep-q-learning-for-data-science-enthusiasts/

https://medium.com/@jereminuerofficial/a-comprehensive-guide-to-deep-q-learning-8aeed632f52f

This is a decorative image for: GAUDI: A Neural Architect for Immersive 3D Scene Generation
October 13, 2022

GAUDI: A Neural Architect for Immersive 3D Scene Generation

The evolution of artificial intelligence in the past decade has been staggering, and now the focus is shifting towards AI and ML systems to understand and generate 3D spaces. As a result, there has been extensive research on manipulating 3D generative models. In this regard, Apple’s AI and ML scientists have developed GAUDI, a method specifically for this job.

An introduction to GAUDI

The GAUDI 3D immersive technique founders named it after the famous architect Antoni Gaudi. This AI model takes the help of a camera pose decoder, which enables it to guess the possible camera angles of a scene. Hence, the decoder then makes it possible to predict the 3D canvas from almost every angle.

What does GAUDI do?

GAUDI can perform multiple functions –

  • The extensions of these generative models have a tremendous effect on ML and computer vision. Pragmatically, such models are highly useful. They are applied in model-based reinforcement learning and planning world models, SLAM is s, or 3D content creation.
  • Generative modelling for 3D objects has been used for generating scenes using graf, pigan, and gsn, which incorporate a GAN (Generative Adversarial Network). The generator codes radiance fields exclusively. Using the 3D space in the scene along with the camera pose generates the 3D image from that point. This point has a density scalar and RGB value for that specific point in 3D space. This can be done from a 2D camera view. It does this by imposing 3D datasets on those 2D shots. It isolates various objects and scenes and combines them to render a new scene altogether.
  • GAUDI also removes GANs pathologies like mode collapse and improved GAN.
  • GAUDI also uses this to train data on a canonical coordinate system. You can compare it by looking at the trajectory of the scenes.

How is GAUDI applied to the content?

The steps of application for GAUDI have been given below:

  • Each trajectory is created, which consists of a sequence of posed images (These images are from a 3D scene) encoded into a latent representation. This representation which has a radiance field or what we refer to as the 3D scene and the camera path is created in a disentangled way. The results are interpreted as free parameters. The problem is optimized by and formulation of a reconstruction objective.
  • This simple training process is then scaled to trajectories, thousands of them creating a large number of views. The model samples the radiance fields totally from the previous distribution that the model has learned.
  • The scenes are thus synthesized by interpolation within the hidden space.
  • The scaling of 3D scenes generates many scenes that contain thousands of images. During training, there is no issue related to canonical orientation or mode collapse.
  • A novel de-noising optimization technique is used to find hidden representations that collaborate in modelling the camera poses and the radiance field to create multiple datasets with state-of-the-art performance in generating 3D scenes by building a setup that uses images and text.

To conclude, GAUDI has more capabilities and can also be used for sampling various images and video datasets. Furthermore, this will make a foray into AR (augmented reality) and VR (virtual reality). With GAUDI in hand, the sky is only the limit in the field of media creation. So, if you enjoy reading about the latest development in the field of AI and ML, then keep a tab on the blog section of the E2E Networks website.

Reference Links

https://www.researchgate.net/publication/362323995_GAUDI_A_Neural_Architect_for_Immersive_3D_Scene_Generation

https://www.technology.org/2022/07/31/gaudi-a-neural-architect-for-immersive-3d-scene-generation/ 

https://www.patentlyapple.com/2022/08/apple-has-unveiled-gaudi-a-neural-architect-for-immersive-3d-scene-generation.html

Build on the most powerful infrastructure cloud

A vector illustration of a tech city using latest cloud technologies & infrastructure