View Article

  • Explore the Use of Artificial Neural Networks in Medicine, Aiming to Understand the Classification, Mechanism, Current Trends, and Applications of Neural Networks in The Pursuit of Smart Health.

  • 1,2,3 Amrutvahini Institute Of  Pharmacy, Sangamner 
    4 College of Pharmaceutical Sciences Loni (PIMS)
     

Abstract

The integration of Artificial Intelligence has undeniably revolutionized the medical and pharmaceutical industries. Artificial neural networks collaborate to minimize time, costs, and effort in research and development. Machine Learning algorithms and predictive models are strategically employed to optimize pharmaceutical operations. This showcases the immense potential of artificial intelligence in Science & Technology, which demands further exploration. This review unequivocally elucidates the operational dynamics of neural networks in the medical field and other pertinent domains, thereby illuminating promising avenues for future research.

Keywords

Neural Network, Artificial Neural Network, Types of Neural Network, Neural Networks Applications

Introduction

The term "neural network" is derived from its conceptual similarity to the human brain. Artificial Intelligence (AI) aims to create advanced computational systems that mimic the human brain's complex neural networks, allowing them to perform complex tasks effortlessly.

AI has significantly transformed the medical and pharmaceutical sectors by utilizing Artificial Neural Networks (ANNs) to assist in various research and development tasks. These networks facilitate innovation in many applications, saving time, reducing costs, and streamlining efforts. However, there are still many untapped opportunities for AI in the fields of science and technology. This review aims to provide insights into the functioning of neural networks in medicine and related fields, as well as highlight potential future research directions.

Neural networks were initially proposed by Alexander Bain and William James separately in 1873 and 1890. In 1943, McCulloch and Pitts developed threshold logic, a computational model based on mathematical algorithms for neural networks, outlining two research approaches: one focused on biological brain processes and the other on AI applications.

In the past decade, artificial intelligence has played an increasingly critical role in medicine. Artificial Neural Networks (ANNs) have revolutionized our world by utilizing complex computational algorithms to find cures for incurable and rare diseases, as well as to optimize and enhance the use of medications and medical procedures. Among the various analytical tools used in AI and machine learning, Artificial Neural Networks (ANNs) have emerged as the most prominent due to their wide range of applications. In addition, during the recent COVID-19 pandemic, AI-based technologies, particularly the Artificial Neural Network (ANN), contributed to potential treatment options.   

Fig. No. 1: Hierarchy of Neural Networks

Introduction to ANNs:

The term "neural network" originates from the brain, and it is the goal of AI to develop intricate computational systems that can rival the human brain's performance. A neural network consists of multiple interconnected nodes or neurons, intended to solve AI-related problems. The connections in artificial neural networks (ANNs) are quantified by weights, with positive weights signifying excitatory connections and negative weights representing inhibitory connections. These weights are utilized in a linear combination of inputs, and the output is then subjected to an activation function to limit its value within a specific range. ANNs find extensive applications in predictive modelling, adaptive control, and various other tasks that involve training on datasets, learning from experience, and deriving insights from complex data.

The concept of the neural network was first conceived by Alexander Bain and William James in the late 19th century. However, it was not until 1943 that a computational model based on mathematical principles was developed by McCulloch and Pitts. This development bifurcated the future research on neural networks into two streams: one focusing on the biological processes of the brain, and the other on applications in AI.[1]

Classification/Types of Neural Network:

Artificial Intelligence (AI), particularly Artificial Neural Networks (ANNs), have garnered significant attention in the medical field in recent years. AI has radically transformed healthcare by uncovering treatments for previously considered incurable diseases and optimizing medication applications using highly complex algorithms. Among the plethora of analysis tools in AI and machine learning, ANNs have been prominently highlighted for their paramount role in treatment strategies during the COVID-19 pandemic.

Neural networks are a foundational technique in machine learning, empowering computers to learn tasks from examples. This network comprises numerous interconnected processing units with each node linked to specific nodes before and after it, enabling seamless information exchange. The network processes information from the training dataset through the input layer, meticulously transforming it into a complex representation as it traverses all layers, culminating at the output layer. With its architecture comprising input, hidden, and output cells, it stands as an incredibly potent tool applicable to a diverse array of purposes.[2]

Fig. No.2: Classification of Neural Networks

AI Algorithms

The following is just a brief overview of some of the algorithms in artificial intelligence with their descriptions:

1. Artificial Neural Networks (ANNs)

Interconnected processing units processing information using a dynamic state response to external inputs.[3]

2. Logistic Regression

 A mechanism to describe the probability of a categorical outcome using a logistic function consisting of one dependent binary variable and one or more independent variables.[4]

3. Hidden Markov Models (HMMs)

 Models the evolution of observable events which may depend upon internal unobservable factors.[5]

4. Clustering Algorithms

Procedures to assign data points to clusters in such a way that items in the same cluster are as similar as possible and items in different clusters are as dissimilar as possible.[6]

5. Fuzzy C-Means

A form of clustering where data points may belong to more than one cluster.

6. K-Means

creates K partitions iteratively, assigning data points to each partition with similarity to a representative centroid. Sparse Autoencoders create deep network structures for feature learning and data clustering. Deep Learning Algorithms are the new version of ANNs that uses plentiful cheap computation for building larger and complex neural networks.[7]

7. Convolutional Neural Networks (CNNs)

 contain an input scanner at the beginning and are typically applied in image processing, but they can deal with other analog inputs such as audio.[8]

8. Recurrent Neural Networks (RNNs)     

Model the temporal dynamic behaviour, conditioning on previous time instants, by interconnecting lower and higher levels.[9]

9. Random Forests (RFs)    

This are collections of many individual decision trees working as an ensemble, and the model's prediction is the class that gets the most votes.[10]

10. Linear Discriminant Analysis (LDA)

Dimensionality reduction of n-dimensional samples is done keeping the class-discriminatory information intact.[11]

11. Support Vector Machines (SVMs)       

Optimum separating hyperplane with the maximum margin, where the nearest point is called.[12]

12. Bayesian Algorithms

These algorithms explicitly apply Bayes' Theorem to problems such as classification and regression.[13]

13. Regression Algorithms

Model the relationship between variables using iteratively-refining a model by minimizing a measure of error in its predictions.[14]

Mechanism of AI algorithms          

1. Data Collection

Gathering relevant data: labelled in the case of supervised, and unlabelled in unsupervised learning.[15]

2. Data Preprocessing

Clean and organize the data for analysis.

3. Model Selection

The selection of an appropriate algorithm depending on the type of problem and data characteristics. Typical models are decision trees, support vector machines, random forests, and artificial neural networks.[16]

4. Training

The model learns patterns or relationships from the training data, and its parameters are changed accordingly.[17]

5. Evaluation

 The model performance is mainly measured using, among other things, accuracy, specificity, and sensitivity. This normally includes cross-validation that guarantees generalization on other data not used in the training stage.[18]

6. Prediction

The application of the trained model to new data to make a prediction or classification.

7. Feedback Loop

It enables the incorporation of feedback from the model's predictions in order to enhance its accuracy over time; this is very important in reinforcement learning scenarios in which a model learns from the outcome of its actions.

Artificial Neural Networks and Their Implementation in the Medicinal and Pharmaceutical Domains:

Artificial neural networks (ANNs) have extensive applications in the medicinal and pharmaceutical industries alongside their widespread use in engineering, computer science, and technology. In drug discovery, ANNs empower the use of various AI and ML architectures for drug screening, design, in-line quality control measurements, and clinical trials. Deep convolutional neural networks are specifically employed in drug development to predict the bioactivity of small compounds, utilizing local convolutional filters to obtain input data on the structural target. It is crucial for sparse ML algorithms, due to their local nature, to exercise caution when dealing with biochemical interactions, whether it involves seeking them out or evaluating them.[19]

Fig. No.3: Application of Artificial Neural Networks

1.3 D Pharmacophores and Neural Networks for Hit-Hopping Beyond the Patent Space of Drug Discovery

This is a very important issue in the area of drug discoveries: the search for hits with new scaffolds, also called scaffold hopping. In this respect, machine learning techniques, especially artificial neural networks (ANN), have been efficient in focusing the research on preferred regions of chemical space. A study was recently published reporting on the approach that uses a three-dimensional pharmacophore descriptor in combination with ANN in the virtual search for NAMs of the metabotropic glutamate receptor 5 (mGluR5) in the treatment of pain, anxiety, and Parkinson's disease. Since there was no available data for the binding of mGluR5 NAMs, a ligand-based virtual screening strategy described the enhancing effect of ANN in virtual screening and scaffold hopping.[20]

Supervised feed-forward networks to generate a mGluR5 focused library; the SOMs for the selection of structurally diverse compounds for bioactivity testing on mGluR5. The supervised networks showed high predictive accuracy in discriminating mGluR5 allosteric antagonists from molecules for other targets and could distinguish mGluR5 from mGluR1 antagonists with lower predictive accuracy. Network ensembles showed higher prediction accuracy than the individual network classifiers.

Commonly used descriptors include,

Hydrophobic descriptors-these descriptors identify hydrophobic regions crucial for binding NAMs to the allosteric sites.[21]

Hydrogen bond donors and acceptors-these are represented as vector indicating possible hydrogen bond formation locations and influence receptors conformation.

Aromatic rings-properties of aromatic rings in 3D space helps to define binding affinity and specificity of potential NAMs.               

2. Neural Network Prediction of Octanol-Water Partition Coefficients

Lipophilicity plays a significant role in the process of drug absorption and its distribution and, thus, is a vital aspect of drug design. Lipophilicity is also described by the partition coefficient in 1-octanol-water, logP, and these values are implemented in use QSAR methods for prediction.[22] Among all, the following ways are in use for predicting logP: hydrophobic fragment values, relationships between the Physico-chemical parameters and logP, and molecular descriptors combined by means of algorithms.[23] Traditional linear methods have often fragmented molecules into larger fragments, while newer methods focus on atomic contributions which then require correction terms due to non-linearity between structure and logP.[21]

A non-linear approach based on neural networks for higher accuracy of logP prediction has been reported to be better than the traditional models. The erroneous structures were filtered out, and the remaining compounds resulted in three sets of 12,729 in total, using a set of organic molecules from the PHYSPROP database. Training for neural networks was conducted using the atomic5 descriptors with 130 input neurons, 12 hidden neurons, and one output neuron. Test sets were used to check generalization and the best model was saved for external validation.[24, 25]

3.Cytotoxicity Prediction Classification by Neural Networks

Cytotoxic compounds can have their discovery costs and resources reduced because such compounds can be eliminated very early in the process. Hence, it is imperative to develop an ANN-based method using atomic fragmental descriptors in the classification of compounds showing in vitro human cytotoxicity.[26] The fragmental descriptors were obtained using Atomic7 linear logP calculation methods. Cytotoxicity data were gathered using in-house screening efforts where 30,000 drug-like molecules were screened for cytotoxic behaviour under pre-defined criteria for the human cell line of interest.[27]

A three-layer feed-forward neural network was set up using the Stuttgart Neural Network Simulator ?(SNNS). There were 164 input neurons,  13 hidden neurons, and 1 output neuron to predict cytotoxicity in human fibroblast cells. For all those compounds that were non-toxic the assigned viability was 0.1. A value of 0.9 was assigned to the toxic compounds. The training was set up based on inputting the Atomic7 descriptors and the output neuron using the scaled experimental viability values.[28]

4. Virtual screening of cytochrome P450 3A4 inhibitors using neural networks

Of the drugs coming under cytochrome P450 3A4, nearly 50% passes through its main isoenzyme 3A4 of cytochrome P450. Its inhibitors lead to drug-drug interactions. Because this enzyme is crucial for so many drugs in the market, the detection of potential 3A4 inhibitors at an early stage is necessary. Several methods of high throughput screening (HTS) have been developed for screening 3A4 inhibitors. Although HTD is more effective in determining potential substrates, it is sometimes incapable of discriminating between substrates and inhibitors. A three-dimensional (3D) QSAR study necessitates good quality IC50 information, which.[29]

To address this issue, a fast neural network model was designed for 3A4 inhibitory activity prediction using 2D structural data. Inhibitory and Gene test of a non-inhibitory were retrieved from Human P450 Metabolism Database tested by the company. Data contained 145 inhibitors and 145 non-inhibitors, which were then split into training and test sets.[22] Under the same method of fingerprinting, 992 numbers of input neurons were created, and the network was structured with 31 hidden neurons and one output neuron. It gave a classification accuracy of 97% and 95% for inhibitors and non-inhibitors, correspondingly. Therefore, it further indicated better prediction regarding those compounds that previously never took any single consideration.[30, 31]

5. Pattern Recognition and Analytical Data Modelling

(Neural networks (NNs) are extremely well suited to pattern recognition in complex or noisy data and hence very effective tools for estimating non-linear connections. No doubt that this is highly important when it comes to research and data analysis, key in identifying the signals appearing as peak-shapes in analytical data such (spectral) areas of study.[32]

For instance, artificial neural networks (ANNs) can differentiate an unknown sample by its complete spectrum and deconvolute overlapping known spectra. Traditional methods consider only one single peak in isolation, whereas the spectrum is used as a whole by ANNs for identification. In contrast, conventional multiple linear regression (MLR) approaches are very labor-intensive iterative procedures requiring a spectrum decomposition and polynomial regression for each peak fitting that can be time-consuming)[33]

In a specific application, ANNs were expertly combined with diffuse reflectance infrared (IR) spectral analysis and X-ray diffraction to swiftly and highly sensitively develop a method for the qualitative and quantitative control of ranitidine hydrochloride, an antihistaminic drug that exists in two polymorphic forms.[34, 35]

6. Modeling the Response Surface

Recent researches have proven clearly the ability of Artificial Neural Networks to model response surface in high performance liquid chromatography (HPLC) optimization. Including retention mapping, i.e. detailed knowledge on how solutes behave chromatographically in relation to the mobile phase components ANNs help a lot in estimating the capacity factors of solutes, which is very advantageous for optimization and it used to be quite an elaborate work requiring large number of experimental separations.[36] Again, several studies have been conducted on this subject and it has uniformly resulted in conclusion that ANNs are able to predict capacity factors more accurately than multilinear stepwise regression models.[37]

7. Relationship between structure and retention

Structure-retention relationship (SRR) methodology was used which is designed to predict chromatography behavior on the basis of structural information about solutes. For example, predicting retention times for the changes in mobile phase pH and composition, as well as solute molecular parameters are related to ANNs.[38] That is, a neural network model accurately related the liquid chromatographic retention of a series of chemically diverse diuretics to their structural descriptors such that new and untested molecules could be assigned predicted retention times. In addition, we introduced a new and effective method to de-convolute overlapping peaks of chromatograms by cross correlating parameters that describe peak shapes with the constituent area thereby achieving remarkably high accuracy and speed over traditional methods.[39]

8. Applications in the Development of Pharmaceutical Products

Development of pharmaceutical products involves solution of complex multivariate optimization problems. The functional relationships among formulation and process variables cannot be easily modelled using conventional techniques. ANNs will find a good fit for such tasks since they can learn and identify patterns in input-output data pairs. After training, ANNs can predict results for new data sets; hence, they are valuable in optimizing pharmaceutical formulations.[40]

In contrast, the Response Surface Methodology, which mainly utilizes second-order polynomial equations, is often deficient in estimation power for predicting the best formulations. ANN models, on the other hand, have been proven to fit and predict better candidates in developing solid dosage forms. They consider many formulation variables and compression factors affecting tablet properties, such as dissolution. In another application of ANNs, it made possible the development of microemulsion-based drug delivery systems with minimal paperchase behaviour using a few inputs alone.[41]

9. QSPR and QSAR Methodologies

The SPR modelling method is based on the basic principle that a compound's molecular structure is very critical to its behaviour. The working of this methodology involves calculating a varied range of physicochemical descriptors, which may include hydrophobicity, topology, electronic properties, steric effects, and others that can be empirically or through modern computational techniques generated.[30]

At the initial periods of QSPR studies, a number of structural descriptors have been calculated with great care in order to precisely describe the chemical structure mathematically. The proper challenge, however, lies in the identification of the best subset of descriptors that would robustly encode the property of interest. The conventional methods of testing of all combinations can be so time-consuming.[24] To attain much better efficiency, genetic algorithms have been very effective optimization systems, using selection and recombination processes to create new sample points with improved fitness. While an effective subset of descriptors is identified, these can then be cleverly mapped to the property of interest using nonlinear computational neural networks.

For instance, it has been shown that back-propagation ANNs could be successfully trained with various descriptors for modelling the structure-activity relationships of capsaicin analogues, where very high correlation was obtained between experimental and predicted results. In one such interesting study, the ANN model correctly classified 34 out of 41 inactive compounds and 58 out of 60 active compounds among 101 capsaicin analogues.)

In conclusion, neural networks are exceedingly versatile and efficient, rendering them an invaluable tool across a multitude of domains, especially in analytical chemistry and pharmaceutical development.

10. Applications in Drug Absorption and Transfer

It has been shown that a more sophisticated neural network model can accurately predict the HIA% from a drug compound's molecular structure. A three-layer feed-forward neural network was effectively used for prediction with six descriptors in an extensive study of 86 drug and drug-like compounds.[42]

In addition, an accurate prediction of the degree of drug transfer into breast milk using a four-layer GNN model will be presented, whose results will be compared with the previous models. In the present study, in-depth research has been carried out on 60 drug compounds and experimentally derived M/P values taken from the literature.[43]

It has also proven its worth in neural network technology, especially in molecular sequence data, ranging from gene identification and protein structure predictions to sequence classification. Such methodologies are very important in understanding the relationship between protein structure and function because unstructured similarities may indicate evolutionary relationships not detectable through sequence analysis.[44]

For example, one back-propagation ANN recognized patterns in protein side-chain contact maps and yielded a remarkable 84.5% accuracy in differentiating the original from the randomized patterns. Even more recently, ANNs trained using genetic algorithms have enjoyed success in the alignment of RNA and DNA sequences and in the determination of RNA folding and secondary structure, firmly establishing the efficacy of this method within biological sequence analysis.[45]

11. Pharmacokinetics and Drug Design

An understanding of pharmacokinetics and pharmacodynamics is indispensable in determining proper drug dosage and selection. ANNs can give a model-independent approach to the analysis of PK-PD data that allows the exact prediction of PD profiles for multiple PK-PD relationships without recourse to detailed structural information. This flexibility gives ANNs a very distinct advantage over traditional model-dependent methods.

Very clearly, structure-based methods have emerged in drug design as superior to traditional methods and save enormous amounts of time and resources. Computational chemistry aspires to quantitative models that predict compound activities based on parameters such as hydrogen bonds, hydrophobic surface area, interaction energies, and desolation.

Two major approaches to finding molecules that fit active sites are:  Using libraries of molecular fragments. In this case, because very large numbers of combinations of fragments can be made, very large numbers of distinct molecules can be produced quickly; the limitations imposed by database size and conformational flexibility are sidestepped.[46]

Conclusion: The quantitative structure-property relationship integrated with genetic algorithms and neural networks has immensely extended molecular modelling, drug design, and biological analysis into areas such as easier and more accurate predictions of chemical behaviours and interactions.[20]

Artificial neural network for Different Physiological Disorders:

Artificial intelligence has made significant strides in the field of medical diagnosis and treatment, revolutionizing the management of a wide range of human ailments including severe cardiovascular conditions, central nervous system disorders, renal disorders, urogenital difficulties, and respiratory illnesses. Predictive modelling using various AI and machine learning tools not only accurately diagnose diseases and recommend treatment options but also forecast potential treatment outcomes, along with the impact of prescribed medication and potential adverse effects once the drug is available to the public.[47] This section aims to debunk exaggerated claims about the functions of different AI algorithms in treating diseases. It endeavours to demystify the mechanisms of various AI tools, such as artificial neural networks (ANNs) and convolutional neural networks (CNNs), to achieve desired outcomes for curing ailments and pathological conditions, thereby vastly improving overall health conditions. The goal is to make intricate AI algorithms more accessible to the general public and ensure transparency in their functioning.[48, 49]

Fig. No. 4: AI in the diagnosis, detection and treatment of human diseases

Use of ANN in UTI

A UTI treatment dataset was created from the information collected from the different patients.  This dataset consisted of data from the regular examination and the final examination report for 59 urinary infection patients. This paper models conclusive diagnosis results by developing three classification models: DT, SVM, RF, and ANN models using this data. The models were then evaluated using measures of accuracy, specificity, and sensitivity. AI in medicine also found its application in clinical diagnostics and treatment recommendations, showing successful implementation in disease classification, suggestions of treatment, and determination of the right drug dosage. Several AI methods such as SVM, ANN, DT, KNN, and RF all are useful methods in providing a strong tool for a physician to analyse and involve clinically complex data in discrete medical domains at the same time.[50, 51]

For similar reasons, AI and big data analytics in CVD are allowing clinicians to gather an even more refined set of predictions on the patient. In the research by Dawes TJW, the AI predicted windows in time when a person with heart disease can die. The software of AI within the group was executed from the scans of cardiac MRI and blood tests conducted over 256 patients, measuring 30,000 points on the heart structures in every beat. This, integrated with eight years of health records of the patients, AI predicted leading abnormal conditions that lead to death and became capable of predicting—with right prediction—detecting the one-year survival with an accuracy rate of 80% as against 60% by clinicians. Besides, deep learning has taken one step further to the scrutiny of cardiac imaging, helping in the assessment of the evaluation of coronary angiography, echocardiography, and electrocardiograms (ECG). In some not-so-distant future, AI's capabilities will likely be denser in the accuracy of the identification of coronary atherosclerotic plaques, with automated assessment of echocardiographic images for improved classification and staging of structural diseases, including valvular disease.[52, 53]

Use of ANN in Respiratory Track Disorders

ML may substantially improve the analysis of PFT scores. ML can identify complex, multidimensional patterns in PFT variations that help identify disease subtypes and hence allow personalization of diagnosis and treatment. Furthermore, it might increase the standardization of interpretation. One AI model was more accurate in identifying correct diagnostic categories than pulmonologists, although clinicians' performance was likely underestimated by the limited amount of clinical information provided to them.[54]

Imaging data is well analysed by convolutional neural networks, specialized ML techniques. They can support the diagnosis of respiratory diseases through chest X-rays or CT scans. For instance, in a specific study, it was found that an AI algorithm designed for the prioritization of chest X-rays significantly reduced the average time it took to review the scans with critical findings from 11.2 to 2.7 days.

While there is huge potential for the use of ML to improve treatment methodologies for respiratory diseases, it is still in its early stages of research. Notable among these applications include tracking lung movements for the delivery of accurate radiotherapy to lung cancers. Current techniques are beset with challenges due to system latency, and real-time retraining of neural networks is shown to improve the accuracy of the delivery of treatments.[55]

Impacts of ANN on covid-19

First reported in Wuhan, China, in December 2019, the COVID-19 pandemic has drastically affected more than 200 countries, causing millions of cases with considerable associated deaths by April 2020. This pandemic has vigorously pursued unprecedented research and corporate efforts worldwide in establishing new ways of addressing the transmission of this viral infection and medical treatment. Technology, and more so artificial intelligence, has no doubt contributed considerably to the global efforts.[56]

AI technologies, ranging from facial recognition cameras to monitor people with travel histories to robots delivering essentials and drones that disinfect public areas and broadcast public health messages, were promptly and effectively deployed during the early outbreak in China. AI has had a very major role in drug discovery, where researchers have started using it to identify new therapeutic molecules. AI techniques are also being effectively applied in medical imaging to identify COVID-19 infections based on X-rays and CT scans.[57]

Tracking software, including monitoring bracelets, has been developed for those in quarantine violating its protocols. AI-enhanced thermal cameras installed on smartphones have contributed to the detection of fever at the same time. Taiwan was able to link national health insurance data with immigration data in order to identify potential COVID-19 patients based on travel and symptom history.

AI finds applications in the identification, tracking, and prediction of outbreaks, assistance in diagnostic practices, processing of healthcare claims, and use of supercomputers in the very important drug and vaccine development process.[55]

Technology Applications of ANNs

Radiological Imaging Diagnosis

  1. Advancements in AI technology have allowed the extraction of radiological features for accurate and timely diagnosis of COVID-19.
  2. Several Convolutional Neural Network models have been found to facilitate early detection by substantially increasing image datasets that would identify COVID-19.
  3. It should be recalled that COVID-Net, a specially developed deep CNN, has been designed with the purpose to detect COVID-19 in CT images and X-rays.
  4. Noting the disease tracking through abnormal respiratory pattern classifiers for large-scale screening.
  5. At the same time, the infection rate needs to be rightly projected through an efficient SIR model, and predict the trajectory of outbreaks by the SEIR model.
  6. In predictive health outcome assessment, the Boost classifier provides a direct clinical test for mortality risk assessment, underlining the importance of the same.
  7. Furthermore, machine learning-based CT radiomics models have been applied to attain high accuracy in predicting hospital stays for COVID-19 patients.
  8. In computational biology and medicine, AI has been playing a huge role in identifying baricitinib as a drug predicted to impede the ability of the virus to infect lung cells.
  9. Moreover, CASP makes a prediction for protein properties from genetic sequences with high accuracy while utilizing deep neural networks in protein structure predictions.
  10. Drug discovery has seen integrated AI-based pipelines manage to generate new drug compounds, which is a breakthrough in that particular field.[58, 59]

Some areas in which ANN has, Impact, made some relevant contributions are as listed below:

1. ANN-based Drug Repurposing

AI accelerated the process of finding drugs already approved and existing that could be potential therapeutic agents against COVID-19. Deep learning, knowledge graphs, and data mining techniques were utilized for ranking-order approved drugs for further investigation. This would usually reduce a number of years off time taken for drug discovery.[60]

2. ANN-Combination Therapy Design

This would aid in designing optimal drug combinations within a very short period, which is quite critical during pandemics where timely interventions are required. With the identification of appropriate drug regimens, even in the dearth of large amounts of data on mechanisms of diseases, platforms such as Project IDentif.AI will enable fast therapeutic development.[61]

3. Patient Matching for Clinical Trial

AI optimizes patient matching to the clinical trials and tailors the drug combinations according to their efficacy and safety profiles. This is a very critical component of increasing clinical successes and fast-tracking the testing process for possible treatments.[62]

4. Established Clinical Successes

AI-driven platforms, as technologies, have already been utilized with much success in oncology and infectious diseases and have the potential to be applied in the development of treatments for COVID-19. Successful application of these technologies further reiterates their validity in a timely manner on urgent health crises.[42, 63]

5. Cost and Time Efficiency

The urgent pressure to address COVID-19 has enabled rapid testing of AI-based drug repurposing strategies that are fast and inexpensive compared to traditional drug development pipelines. This has the potential to cut a large amount of time and resources in bringing effective treatments to market.

That is to say, AI has transformed the COVID-19 drug discovery and development landscape by facilitating rapid identification of promising drug candidates and combination therapies[64].

Medical Diagnosis Applications

AI algorithms, particularly ANNs, are being developed with much promise for medical applications. For instance, in the diagnosis of UTI, an ANN model made with a very few inputs like pollakiuria and suprapubic pain symptoms had accuracy as high as 98.3%, thus showing its power compared to the conventional diagnostic techniques that need a long list of clinical data.[65]

On brain diseases, AI techniques have been applied to several complex analyses of medical data for differential diagnosis and planning of treatment in Alzheimer's and schizophrenia. According to a systematic review published by Marzullo &Calimero, 2020, AI algorithms—chiefly, ANN—have been applied to several clinical applications such as diagnosis and surgical assistance.[66]

Use of machine learning in diagnosis

Machine learning forms a prominent subset of artificial intelligence and has superior relevance in the identification of categories and predicting unknown conditions through data. During the last years, several applications made a cost-effective analysis and processing of enormous brain data for the detection of conditions including but not limited to Alzheimer's, dementia, schizophrenia, multiple sclerosis, cancer, other infectious, and degenerative diseases. Recently, the subfield in AI—deep learning—has radically altered most neurosurgical tasks. DL algorithms are very good at computer vision, outperforming more traditional methods on numerous benchmarks of image analysis tasks. DL self-learns useful representations and features directly from raw data; there is no necessity to compute and select relevant attributes as in conventional models of machine learning.[67]

Various types of data are used for brain care, including gene sequences, EHR, EEG and MER data. The EHR systems contain a huge amount of medical data that represents real-world diseases and care processes, potentially providing richer data than in the traditional design and conduction of randomized clinical trials. EEG measures weak electromagnetic signals of the neuronal activity and, thus, the recording of both slow and rapid brain dynamics with a time resolution in the millisecond range. This capability allows researchers to study activities within a frequency range of neurons and thereby gain insight into the complex functionality of the brain. MER adds further to surgical accuracy in areas below the cortex that the Deep Brain Stimulation (DBS) leads are placed to ensure that these are placed correctly within the structures to which the leading are targeted.[68]

Challenges and Future Directions

Though these are very promising results, there are also a number of challenges with respect to the implementation of AI into healthcare: large and high-quality datasets are required for training, and integration of AI systems into existing clinical workflows is needed. Since AI is still developing, research is in progress regarding its application in fields like respiratory medicine, wherein there lie future perspectives for lung cancer screening and the analysis of pulmonary function test scores[41]. But still, it is inconceivably tricky to have full automation of diagnostic processes wherein algorithms have to identify a wide range of pathologies with high accuracy. The reference is avoided herein for the sake of brevity[69].

Briefly, AI algorithms are revolutionizing industries in general and healthcare in particular by giving tools which improve diagnosis and reduce turnaround time. Working on these algorithms and making subsequent modification is bound to give further medical technologies and improvement to the lot of patients[70].

CONCLUSION:

Undoubtedly, Artificial Intelligence has had a profound influence on the global healthcare sector. It has made significant contributions to the pharmaceutical and medicinal field, being utilized in various areas such as drug discovery, formulation development, disease diagnosis and detection, and clinical research. Explainable Artificial Intelligence has demonstrated that numerous AI algorithms have effectively impacted the healthcare system and people's lives. However, the underlying mechanism of these AI techniques has not been easily understood by individuals utilizing these machine-mediated facilities for healthcare support.

REFERENCES

  1. Haglin, J.M., et al., Artificial neural networks in medicine. 2019. 9: p. 1-6.
  2. Krizhevsky, A., I. Sutskever, and G.E.J.C.o.t.A. Hinton, ImageNet classification with deep convolutional neural networks. 2017. 60(6): p. 84-90.
  3. Daponte, P. and D.J.M. Grimaldi, Artificial neural networks in measurements. 1998. 23(2): p. 93-115.
  4. Vittinghoff, E., et al., Logistic regression. 2012: p. 139-202.
  5. Yu, S.-Z., Hidden Semi-Markov models: theory, algorithms and applications. 2015: Morgan Kaufmann.
  6. Michaud, P.J.F.G.C.S., Clustering techniques. 1997. 13(2-3): p. 135-147.
  7. Kim, K., M.E. Aminanto, and H.C. Tanuwidjaja, Network intrusion detection using deep learning: a feature learning approach. 2018: Springer.
  8. Pinaya, W.H.L., et al., Convolutional neural networks, in Machine learning. 2020, Elsevier. p. 173-191.
  9. Salehinejad, H., et al., Recent advances in recurrent neural networks. 2017.
  10. Fawagreh, K., et al., Random forests: from early developments to recent advancements. 2014. 2(1): p. 602-609.
  11. Chamkalani, A., et al., Pattern recognition insight into drilling optimization of shaly formations. 2017. 156: p. 322-339.
  12. Gualtieri, J. and S. Chettri. Support vector machines for classification of hyperspectral data. in IGARSS 2000. IEEE 2000 International Geoscience and Remote Sensing Symposium. Taking the Pulse of the Planet: The Role of Remote Sensing in Managing the Environment. Proceedings (Cat. No. 00CH37120). 2000. IEEE.
  13. Denison, D.G., et al., Bayesian methods for nonlinear classification and regression. Vol. 386. 2002: John Wiley & Sons.
  14. Ayhan, H.M., S.J.C. K?r, and I. Engineering, Ml-driven approaches to enhance inventory planning: Inoculant weight application in casting processes. 2024: p. 110280.
  15. Roh, Y., et al., A survey on data collection for machine learning: a big data-ai integration perspective. 2019. 33(4): p. 1328-1347.
  16. Rodriguez-Galiano, V., et al., Machine learning predictive models for mineral prospectivity: An evaluation of neural networks, random forest, regression trees and support vector machines. 2015. 71: p. 804-818.
  17. Lai, G., et al. Modeling long-and short-term temporal patterns with deep neural networks. in The 41st international ACM SIGIR conference on research & development in information retrieval. 2018.
  18. Raschka, S.J.a.p.a., Model evaluation, model selection, and algorithm selection in machine learning. 2018.
  19. Baskin, I.I., D. Winkler, and I.V.J.E.o.o.d.d. Tetko, A renaissance of neural networks in drug discovery. 2016. 11(8): p. 785-795.
  20. Blaschke, T., et al., REINVENT 2.0: an AI tool for de novo drug design. 2020. 60(12): p. 5918-5922.
  21. Van de Waterbeemd, H., Hydrophobicity of Organic Compounds: How to Calculate it by Personal Computers. 1986: CompuDrug International.
  22. Molnár, L., G.M.J.B. Keseru, and M.C. Letters, A neural network based virtual screening of cytochrome P450 3A4 inhibitors. 2002. 12(3): p. 419-422.
  23. Wegner, J.K., A.J.J.o.C.I. Zell, and C. Sciences, Prediction of aqueous solubility and partition coefficient optimized by a genetic algorithm based descriptor selection method. 2003. 43(3): p. 1077-1084.
  24. Duprat, A.F., et al., Toward a principled methodology for neural network design and performance evaluation in QSAR. Application to the prediction of logP. 1998. 38(4): p. 586-594.
  25. Tetko, I.V., et al., Prediction of n-octanol/water partition coefficients from PHYSPROP database using artificial neural networks and E-state indices. 2001. 41(5): p. 1407-1421.
  26. Sun, H., et al., Predictive models for estimating cytotoxicity on the basis of chemical structures. 2020. 28(10): p. 115422.
  27. Karim, A., et al., Efficient toxicity prediction via simple features using shallow neural networks and decision trees. 2019. 4(1): p. 1874-1888.
  28. Zeiler, M.D., G.W. Taylor, and R. Fergus. Adaptive deconvolutional networks for mid and high level feature learning. in 2011 international conference on computer vision. 2011. IEEE.
  29. Keser?, G.M.J.J.o.c.-a.m.d., A virtual high throughput screen for high affinity cytochrome P450cam substrates. Implications for in silico prediction of drug metabolism. 2001. 15: p. 649-657.
  30. Ekins, S., et al., Three-and four-dimensional quantitative structure activity relationship analyses of cytochrome P-450 3A4 inhibitors. 1999. 290(1): p. 429-438.
  31. Ekins, S., et al., Three-dimensional-quantitative structure activity relationship analysis of cytochrome P-450 3A4 substrates. 1999. 291(1): p. 424-433.
  32. Bishop, C.M., Neural networks for pattern recognition. 1995: Oxford university press.
  33. Fidler, S., M. Boben, and A. Leonardis. Similarity-based cross-layered hierarchical representation for object categorization. in 2008 IEEE Conference on Computer Vision and Pattern Recognition. 2008. IEEE.
  34. He, K., et al. Deep residual learning for image recognition. in Proceedings of the IEEE conference on computer vision and pattern recognition. 2016.
  35. Kingma, D.P.J.a.p.a., Auto-encoding variational bayes. 2013.
  36. Pilkington, J.L., et al., Comparison of response surface methodology (RSM) and artificial neural networks (ANN) towards efficient extraction of artemisinin from Artemisia annua. 2014. 58: p. 15-24.
  37. D’Archivio, A.A., M.A. Maggi, and F.J.A.c.a. Ruggieri, Multi-variable retention modelling in reversed-phase high-performance liquid chromatography based on the solvation method: A comparison between curvilinear and artificial neural network regression. 2011. 690(1): p. 35-46.
  38. Kyngäs, J. and J.J.Q.S.A.R. Valjakka, Evolutionary Neural Networks in Quantitative Structure?Activity Relationships of Dihydrofolate Reductase Inhibitors. 1996. 15(4): p. 296-301.
  39. Andrea, T. and H.J.J.o.m.c. Kalayeh, Applications of neural networks in quantitative structure-activity relationships of dihydrofolate reductase inhibitors. 1991. 34(9): p. 2824-2836.
  40. Hussain, A.S., X. Yu, and R.D.J.P.r. Johnson, Application of neural computing in pharmaceutical product development. 1991. 8: p. 1248-1252.
  41. Jariwala, N., et al., Intriguing of pharmaceutical product development processes with the help of artificial intelligence and deep/machine learning or artificial neural network. 2023: p. 104751.
  42. Agatonovic-Kustrin, S., R.J.J.o.p. Beresford, and b. analysis, Basic concepts of artificial neural network (ANN) modeling and its application in pharmaceutical research. 2000. 22(5): p. 717-727.
  43. Terzi?, V., et al. Passive absorption prediction of transdermal drug application with Artificial Neural Network. in CMBEBIH 2017: Proceedings of the International Conference on Medical and Biological Engineering 2017. 2017. Springer.
  44. Chen, Y., et al., The application of an artificial neural network and pharmacokinetic simulations in the design of controlled-release dosage forms. 1999. 59(1): p. 33-41.
  45. Puri, M., et al., Introduction to artificial neural network (ANN) as a predictive tool for drug design, discovery, delivery, and disposition: Basic concepts and modeling, in Artificial neural network for drug design, delivery and disposition. 2016, Elsevier. p. 3-13.
  46. Clark, D.E. and S.D.J.D.d.t. Pickett, Computational methods for the prediction of ‘drug-likeness’. 2000. 5(2): p. 49-58.
  47. Vashistha, R., et al., Artificial intelligence integration for neurodegenerative disorders, in Leveraging Biomedical and Healthcare Data. 2019, Elsevier. p. 77-89.
  48. Yan, Y., et al., The primary use of artificial intelligence in cardiovascular diseases: what kind of potential role does artificial intelligence play in future medicine? 2019. 16(8): p. 585.
  49. Romiti, S., et al., Artificial intelligence (AI) and cardiovascular diseases: an unexpected alliance. 2020. 2020(1): p. 4972346.
  50. Yao, L., et al., Application of artificial intelligence in renal disease. 2021. 4: p. 54-61.
  51. Liang, X., et al., Artificial intelligence-aided ultrasound in renal diseases: a systematic review. 2023. 13(6): p. 3988.
  52. Ozkan, I.A., et al., Diagnosis of urinary tract infection based on artificial intelligence methods. 2018. 166: p. 51-59.
  53. Go?dzikiewicz, N., D. Zwoli?ska, and D.J.J.o.C.M. Polak-Jonkisz, The use of artificial intelligence algorithms in the diagnosis of urinary tract infections—a literature review. 2022. 11(10): p. 2734.
  54. Bibi, H., et al., Prediction of emergency department visits for respiratory symptoms using an artificial neural network. 2002. 122(5): p. 1627-1632.
  55. Ijaz, A., et al., Towards using cough for respiratory disease diagnosis by leveraging Artificial Intelligence: A survey. 2022. 29: p. 100832.
  56. Ghanim, M.S., et al., ANN-Based traffic volume prediction models in response to COVID-19 imposed measures. 2022. 81: p. 103830.
  57. Naveed, H.M., et al., Artificial neural network (ANN)-based estimation of the influence of COVID-19 pandemic on dynamic and emerging financial markets. 2023. 190: p. 122470.
  58. Piraino, D.W., et al., Application of an artificial neural network in radiographic diagnosis. 1991. 4: p. 226-232.
  59. Ashizawa, K., et al., Artificial neural networks in chest radiography: application to the differential diagnosis of interstitial lung disease. 1999. 6(1): p. 2-9.
  60. Dhanalakshmi, M., et al., Artificial neural network-based study predicts GS-441524 as a potential inhibitor of SARS-CoV-2 activator protein furin: a polypharmacology approach. 2022. 194(10): p. 4511-4529.
  61. Demir, C.J.J.o.S.R.-A., Examination of the effect of ANN and NLPCA technique on prediction performance in patients with breast tumors. 2024(057): p. 133-143.
  62. Metz, J.M., et al., An Internet-based cancer clinical trials matching resource. 2005. 7(3): p. e24.
  63. Chakravarty, K., et al., Driving success in personalized medicine through AI-enabled computational modeling. 2021. 26(6): p. 1459-1465.
  64. Wi?cek, D., A. Burduk, and I.J.A.M.S. Kuric, The use of ANN in improving efficiency and ensuring the stability of the copper ore mining process. 2019. 24(1).
  65. Kaur, S., et al., Medical diagnostic systems using artificial intelligence (ai) algorithms: Principles and perspectives. 2020. 8: p. 228049-228069.
  66. Patel, J.L. and R.K.J.C.c.p. Goyal, Applications of artificial neural networks in medical science. 2007. 2(3): p. 217-226.
  67. Abadi, M., et al., Tensorflow: Large-scale machine learning on heterogeneous distributed systems. 2016.
  68. Mekov, E., M. Miravitlles, and R.J.E.r.o.r.m. Petkov, Artificial intelligence and machine learning in respiratory medicine. 2020. 14(6): p. 559-564.
  69. Noorain, et al., Artificial intelligence in drug formulation and development: applications and future prospects. 2023. 24(9): p. 622-634.
  70. Vatansever, S., et al., Artificial intelligence and machine learning?aided drug discovery in central nervous system diseases: State?of?the?arts and future directions. 2021. 41(3): p. 1427-1473.

Reference

  1. Haglin, J.M., et al., Artificial neural networks in medicine. 2019. 9: p. 1-6.
  2. Krizhevsky, A., I. Sutskever, and G.E.J.C.o.t.A. Hinton, ImageNet classification with deep convolutional neural networks. 2017. 60(6): p. 84-90.
  3. Daponte, P. and D.J.M. Grimaldi, Artificial neural networks in measurements. 1998. 23(2): p. 93-115.
  4. Vittinghoff, E., et al., Logistic regression. 2012: p. 139-202.
  5. Yu, S.-Z., Hidden Semi-Markov models: theory, algorithms and applications. 2015: Morgan Kaufmann.
  6. Michaud, P.J.F.G.C.S., Clustering techniques. 1997. 13(2-3): p. 135-147.
  7. Kim, K., M.E. Aminanto, and H.C. Tanuwidjaja, Network intrusion detection using deep learning: a feature learning approach. 2018: Springer.
  8. Pinaya, W.H.L., et al., Convolutional neural networks, in Machine learning. 2020, Elsevier. p. 173-191.
  9. Salehinejad, H., et al., Recent advances in recurrent neural networks. 2017.
  10. Fawagreh, K., et al., Random forests: from early developments to recent advancements. 2014. 2(1): p. 602-609.
  11. Chamkalani, A., et al., Pattern recognition insight into drilling optimization of shaly formations. 2017. 156: p. 322-339.
  12. Gualtieri, J. and S. Chettri. Support vector machines for classification of hyperspectral data. in IGARSS 2000. IEEE 2000 International Geoscience and Remote Sensing Symposium. Taking the Pulse of the Planet: The Role of Remote Sensing in Managing the Environment. Proceedings (Cat. No. 00CH37120). 2000. IEEE.
  13. Denison, D.G., et al., Bayesian methods for nonlinear classification and regression. Vol. 386. 2002: John Wiley & Sons.
  14. Ayhan, H.M., S.J.C. K?r, and I. Engineering, Ml-driven approaches to enhance inventory planning: Inoculant weight application in casting processes. 2024: p. 110280.
  15. Roh, Y., et al., A survey on data collection for machine learning: a big data-ai integration perspective. 2019. 33(4): p. 1328-1347.
  16. Rodriguez-Galiano, V., et al., Machine learning predictive models for mineral prospectivity: An evaluation of neural networks, random forest, regression trees and support vector machines. 2015. 71: p. 804-818.
  17. Lai, G., et al. Modeling long-and short-term temporal patterns with deep neural networks. in The 41st international ACM SIGIR conference on research & development in information retrieval. 2018.
  18. Raschka, S.J.a.p.a., Model evaluation, model selection, and algorithm selection in machine learning. 2018.
  19. Baskin, I.I., D. Winkler, and I.V.J.E.o.o.d.d. Tetko, A renaissance of neural networks in drug discovery. 2016. 11(8): p. 785-795.
  20. Blaschke, T., et al., REINVENT 2.0: an AI tool for de novo drug design. 2020. 60(12): p. 5918-5922.
  21. Van de Waterbeemd, H., Hydrophobicity of Organic Compounds: How to Calculate it by Personal Computers. 1986: CompuDrug International.
  22. Molnár, L., G.M.J.B. Keseru, and M.C. Letters, A neural network based virtual screening of cytochrome P450 3A4 inhibitors. 2002. 12(3): p. 419-422.
  23. Wegner, J.K., A.J.J.o.C.I. Zell, and C. Sciences, Prediction of aqueous solubility and partition coefficient optimized by a genetic algorithm based descriptor selection method. 2003. 43(3): p. 1077-1084.
  24. Duprat, A.F., et al., Toward a principled methodology for neural network design and performance evaluation in QSAR. Application to the prediction of logP. 1998. 38(4): p. 586-594.
  25. Tetko, I.V., et al., Prediction of n-octanol/water partition coefficients from PHYSPROP database using artificial neural networks and E-state indices. 2001. 41(5): p. 1407-1421.
  26. Sun, H., et al., Predictive models for estimating cytotoxicity on the basis of chemical structures. 2020. 28(10): p. 115422.
  27. Karim, A., et al., Efficient toxicity prediction via simple features using shallow neural networks and decision trees. 2019. 4(1): p. 1874-1888.
  28. Zeiler, M.D., G.W. Taylor, and R. Fergus. Adaptive deconvolutional networks for mid and high level feature learning. in 2011 international conference on computer vision. 2011. IEEE.
  29. Keser?, G.M.J.J.o.c.-a.m.d., A virtual high throughput screen for high affinity cytochrome P450cam substrates. Implications for in silico prediction of drug metabolism. 2001. 15: p. 649-657.
  30. Ekins, S., et al., Three-and four-dimensional quantitative structure activity relationship analyses of cytochrome P-450 3A4 inhibitors. 1999. 290(1): p. 429-438.
  31. Ekins, S., et al., Three-dimensional-quantitative structure activity relationship analysis of cytochrome P-450 3A4 substrates. 1999. 291(1): p. 424-433.
  32. Bishop, C.M., Neural networks for pattern recognition. 1995: Oxford university press.
  33. Fidler, S., M. Boben, and A. Leonardis. Similarity-based cross-layered hierarchical representation for object categorization. in 2008 IEEE Conference on Computer Vision and Pattern Recognition. 2008. IEEE.
  34. He, K., et al. Deep residual learning for image recognition. in Proceedings of the IEEE conference on computer vision and pattern recognition. 2016.
  35. Kingma, D.P.J.a.p.a., Auto-encoding variational bayes. 2013.
  36. Pilkington, J.L., et al., Comparison of response surface methodology (RSM) and artificial neural networks (ANN) towards efficient extraction of artemisinin from Artemisia annua. 2014. 58: p. 15-24.
  37. D’Archivio, A.A., M.A. Maggi, and F.J.A.c.a. Ruggieri, Multi-variable retention modelling in reversed-phase high-performance liquid chromatography based on the solvation method: A comparison between curvilinear and artificial neural network regression. 2011. 690(1): p. 35-46.
  38. Kyngäs, J. and J.J.Q.S.A.R. Valjakka, Evolutionary Neural Networks in Quantitative Structure?Activity Relationships of Dihydrofolate Reductase Inhibitors. 1996. 15(4): p. 296-301.
  39. Andrea, T. and H.J.J.o.m.c. Kalayeh, Applications of neural networks in quantitative structure-activity relationships of dihydrofolate reductase inhibitors. 1991. 34(9): p. 2824-2836.
  40. Hussain, A.S., X. Yu, and R.D.J.P.r. Johnson, Application of neural computing in pharmaceutical product development. 1991. 8: p. 1248-1252.
  41. Jariwala, N., et al., Intriguing of pharmaceutical product development processes with the help of artificial intelligence and deep/machine learning or artificial neural network. 2023: p. 104751.
  42. Agatonovic-Kustrin, S., R.J.J.o.p. Beresford, and b. analysis, Basic concepts of artificial neural network (ANN) modeling and its application in pharmaceutical research. 2000. 22(5): p. 717-727.
  43. Terzi?, V., et al. Passive absorption prediction of transdermal drug application with Artificial Neural Network. in CMBEBIH 2017: Proceedings of the International Conference on Medical and Biological Engineering 2017. 2017. Springer.
  44. Chen, Y., et al., The application of an artificial neural network and pharmacokinetic simulations in the design of controlled-release dosage forms. 1999. 59(1): p. 33-41.
  45. Puri, M., et al., Introduction to artificial neural network (ANN) as a predictive tool for drug design, discovery, delivery, and disposition: Basic concepts and modeling, in Artificial neural network for drug design, delivery and disposition. 2016, Elsevier. p. 3-13.
  46. Clark, D.E. and S.D.J.D.d.t. Pickett, Computational methods for the prediction of ‘drug-likeness’. 2000. 5(2): p. 49-58.
  47. Vashistha, R., et al., Artificial intelligence integration for neurodegenerative disorders, in Leveraging Biomedical and Healthcare Data. 2019, Elsevier. p. 77-89.
  48. Yan, Y., et al., The primary use of artificial intelligence in cardiovascular diseases: what kind of potential role does artificial intelligence play in future medicine? 2019. 16(8): p. 585.
  49. Romiti, S., et al., Artificial intelligence (AI) and cardiovascular diseases: an unexpected alliance. 2020. 2020(1): p. 4972346.
  50. Yao, L., et al., Application of artificial intelligence in renal disease. 2021. 4: p. 54-61.
  51. Liang, X., et al., Artificial intelligence-aided ultrasound in renal diseases: a systematic review. 2023. 13(6): p. 3988.
  52. Ozkan, I.A., et al., Diagnosis of urinary tract infection based on artificial intelligence methods. 2018. 166: p. 51-59.
  53. Go?dzikiewicz, N., D. Zwoli?ska, and D.J.J.o.C.M. Polak-Jonkisz, The use of artificial intelligence algorithms in the diagnosis of urinary tract infections—a literature review. 2022. 11(10): p. 2734.
  54. Bibi, H., et al., Prediction of emergency department visits for respiratory symptoms using an artificial neural network. 2002. 122(5): p. 1627-1632.
  55. Ijaz, A., et al., Towards using cough for respiratory disease diagnosis by leveraging Artificial Intelligence: A survey. 2022. 29: p. 100832.
  56. Ghanim, M.S., et al., ANN-Based traffic volume prediction models in response to COVID-19 imposed measures. 2022. 81: p. 103830.
  57. Naveed, H.M., et al., Artificial neural network (ANN)-based estimation of the influence of COVID-19 pandemic on dynamic and emerging financial markets. 2023. 190: p. 122470.
  58. Piraino, D.W., et al., Application of an artificial neural network in radiographic diagnosis. 1991. 4: p. 226-232.
  59. Ashizawa, K., et al., Artificial neural networks in chest radiography: application to the differential diagnosis of interstitial lung disease. 1999. 6(1): p. 2-9.
  60. Dhanalakshmi, M., et al., Artificial neural network-based study predicts GS-441524 as a potential inhibitor of SARS-CoV-2 activator protein furin: a polypharmacology approach. 2022. 194(10): p. 4511-4529.
  61. Demir, C.J.J.o.S.R.-A., Examination of the effect of ANN and NLPCA technique on prediction performance in patients with breast tumors. 2024(057): p. 133-143.
  62. Metz, J.M., et al., An Internet-based cancer clinical trials matching resource. 2005. 7(3): p. e24.
  63. Chakravarty, K., et al., Driving success in personalized medicine through AI-enabled computational modeling. 2021. 26(6): p. 1459-1465.
  64. Wi?cek, D., A. Burduk, and I.J.A.M.S. Kuric, The use of ANN in improving efficiency and ensuring the stability of the copper ore mining process. 2019. 24(1).
  65. Kaur, S., et al., Medical diagnostic systems using artificial intelligence (ai) algorithms: Principles and perspectives. 2020. 8: p. 228049-228069.
  66. Patel, J.L. and R.K.J.C.c.p. Goyal, Applications of artificial neural networks in medical science. 2007. 2(3): p. 217-226.
  67. Abadi, M., et al., Tensorflow: Large-scale machine learning on heterogeneous distributed systems. 2016.
  68. Mekov, E., M. Miravitlles, and R.J.E.r.o.r.m. Petkov, Artificial intelligence and machine learning in respiratory medicine. 2020. 14(6): p. 559-564.
  69. Noorain, et al., Artificial intelligence in drug formulation and development: applications and future prospects. 2023. 24(9): p. 622-634.
  70. Vatansever, S., et al., Artificial intelligence and machine learning?aided drug discovery in central nervous system diseases: State?of?the?arts and future directions. 2021. 41(3): p. 1427-1473.

Photo
Priyanka Gavande
Corresponding author

Amrutvahini Institute Of  Pharmacy, Sangamner

Photo
Pallavi Shelar
Co-author

Amrutvahini Institute Of  Pharmacy, Sangamner

Photo
Rutuja Borhade
Co-author

Amrutvahini Institute Of  Pharmacy, Sangamner

Photo
Mahesh Gavande
Co-author

College of Pharmaceutical Sciences Loni (PIMS)

Priyanka Gavande, Pallavi Shelar, Rutuja Borhade, Mahesh Gavande, Explore the Use of Artificial Neural Networks in Medicine, Aiming to Understand the Classification, Mechanism, Current Trends, and Applications of Neural Networks in The Pursuit of Smart Health., Int. J. of Pharm. Sci., 2025, Vol 3, Issue 10, 132-149. https://doi.org/10.5281/zenodo.17241829

More related articles
Development And Evaluation Luliconazole Embedded T...
K.M. Manjanna, G. Shiva Krishna, Mohammed Azharuddin, G. Govinda ...
Nanocarrier-Based Drug Delivery in Cholangiocarcin...
Pooja Paliwal , Vidhya Thorat , Madhuri Sonawane , Rohan Sawant ,...
A Comprehensive Review on Clerodendrum Serratum: P...
Anuradha Pailwan, Sakshi Mhetre, Manorama Khumbar, Shrirang Kharm...
No-Rinse Body Wash: Formulation, Evaluation, and Efficacy for Water-Limited Use...
Katrin Mae Ortega, Mikyla A. Candava, Krystle A. Abrau, Marcus Daryl C. Antonio, Hannah Noreen C. Ba...
Nanoparticle-Based Therapeutic Strategies for the Treatment of Malaria: A Compre...
Rohan Sawant , Vidhya Thorat , Pooja Paliwal, Madhuri Sonawane , Yashashri Deore , Dr. P. N. Sabale,...
Related Articles
Exploring Herbal Immunomodulators as Potential Therapeutic Agents for Long COVID...
Needa Sheikh, Vaidehi Nikure, Mohd. Shaklain, Ganesh Akone, Iftesham Ansari, Rubina Sheikh, Nujba Fa...
Chocolate-Based Drug Delivery Systems: A Palatable Platform for Novel Therapeuti...
Gaurav Pawar, Dr. Shivraj Jadhav, Mayur Bhamare, Dr. Sunil Mahajan, ...
NanoEmulsion-Infused Buccal Films: A Hybrid Drug Delivery Platform For Overcomin...
Drashti Patel, Dhwanit Darji, Jay Maske, Rakesh Melaja, Jaini Patel, ...
Development And Evaluation Luliconazole Embedded Transungual (Nail) Patches for ...
K.M. Manjanna, G. Shiva Krishna, Mohammed Azharuddin, G. Govinda Reddy, ...
More related articles
Development And Evaluation Luliconazole Embedded Transungual (Nail) Patches for ...
K.M. Manjanna, G. Shiva Krishna, Mohammed Azharuddin, G. Govinda Reddy, ...
Nanocarrier-Based Drug Delivery in Cholangiocarcinoma: The Role of Liposomes...
Pooja Paliwal , Vidhya Thorat , Madhuri Sonawane , Rohan Sawant , Yashashri Deore , Dr. P. N. Sable,...
A Comprehensive Review on Clerodendrum Serratum: Phytochemistry, Pharmacological...
Anuradha Pailwan, Sakshi Mhetre, Manorama Khumbar, Shrirang Kharmate, ...
Development And Evaluation Luliconazole Embedded Transungual (Nail) Patches for ...
K.M. Manjanna, G. Shiva Krishna, Mohammed Azharuddin, G. Govinda Reddy, ...
Nanocarrier-Based Drug Delivery in Cholangiocarcinoma: The Role of Liposomes...
Pooja Paliwal , Vidhya Thorat , Madhuri Sonawane , Rohan Sawant , Yashashri Deore , Dr. P. N. Sable,...
A Comprehensive Review on Clerodendrum Serratum: Phytochemistry, Pharmacological...
Anuradha Pailwan, Sakshi Mhetre, Manorama Khumbar, Shrirang Kharmate, ...