Skip to main content

Influence of artificial intelligence in modern pharmaceutical formulation and drug development

Abstract

Background

Artificial intelligence (AI) revolutionized the formulation and development of modern pharmaceuticals. With the help of AI, researchers can now optimize drug design, develop formulations, and streamline clinical trials in a much accurate and efficient way. Drug development might be greatly expedited and time-consuming procedure; however, with the help of AI this are significantly reduced.

Main body of abstract

The main advantages of AI in pharmaceutical formulation are its capacity to analyse vast amounts of data and spot patterns and connections that human researchers would miss. Various tools and technologies, such as ANN, fuzzy logic, neuro-fuzzy logic, and genetic algorithm are used for analysing the date, of which ANN is popular and mostly used. AI enables the discovery of novel pharmacological targets and the creation of more potent medications. AI may also be used to improve medication formulations by forecasting the solubility, stability, and bioavailability of drug candidates, increasing the likelihood that clinical trials will be successful.

AI is also applied in designing clinical trials, reducing the time and cost of the process by identifying patient populations that are most likely to benefit from the treatment. Additionally, AI can monitor patients during clinical trials, detecting real-time adverse effects and adjusting dosages to improve patient outcomes.

Conclusion

AI is a potent pharmaceutical formulation and development tool, allowing researchers to analyse vast amounts of data, optimize drug formulations, and streamline clinical trials. As technology develops, experts anticipate that AI will increasingly show a crucial part in drug development, enabling faster, more efficient, and more effective treatments for various diseases.

Background

The phrase "Artificial Intelligence" (AI) is a general term that describes the least amount of human intervention possible when utilizing a computer to imitate intelligent behaviour. AI is the subfield of computer science that deals with programming to solve issues [1]. It has developed into a discipline that addresses problems in business, engineering, and healthcare. The creation of expert systems is one use for AI. An inference engine, an information base, and a user interface make up an expert system. AI possesses distinctive characteristics that enable it to reason and execute actions with the highest probability of accomplishing a specific objective. AI accomplishes this through a combination of algorithms that simulate even the most fundamental human intellectual capabilities. Nowadays, AI is gaining momentum across multiple sectors, and the pharmaceutical industry is at the forefront of this trend. In the past, formulators have favoured statistical methods, for example response surface approach, for analysing design space. However, optimization using this approach has the potential to be deceptive, especially when dealing with a complex formulation. Two methods that can tackle the issue at hand have been developed as a result of recent developments in mathematics and computer science. The first technique involves using neural networks to replicate how the human brain processes information. The second technique is genetic algorithms, which mimic biological systems' self-organizing and adaptive natures through an evolutionary method. The practical requests of AI in the pharmaceutical production house are highlighted in this paper, including drug research and development (R&D), drug repurposing, increasing pharmaceutical output, clinical testing, etc. These applications reduce the need for human labour and accelerate the drug development process [2]. Over the past few years, the pharmaceutical manufacturing company has significantly improved its data digitalization. To answer challenging clinical problems, acquiring, examining, and applying this knowledge is difficult due to this digitization. AI is used to address this since it can manage enormous volumes of data with increased automation. However, it does not endanger the physical existence of humans. AI uses hardware and software to analyse input data and learn from it in order to accomplish specified goals. This review explains that its services in the pharmaceutical sector are constantly growing. The rapid advancement of AI-guided automation, according to the McKinsey Global Institute, will fundamentally alter how society views labour. Every stage of the pharmaceutical product life cycle, including drug discovery, optimization, formulation development, characterization, quality testing, marketing, and post-marketing surveillance, can integrate AI to improve its efficacy [3].

Tools, technology, and networks

Artificial neural networks (ANN)

Machine learning (ML) is an essential subdivision of AI. A big part of ML is deep learning, which involves ANN. The ANN is made up of multilayer functional units that mimic how electrical impulses are transmitted in the human brain, i.e. it mimics a human brain. They are mainly biologically motivated systems. It takes input and learns directly from input data; primarily, neurons work mainly on the summation of all information and express an output [4].

The fundamental component of biological neural systems is the neuron. Neurons are electrochemical cells; they receive signals from one neuron and transmit signals to other neurons [4]. Like humans, the ANN system has a primary component known as a "perceptron" or node. Nodes are arranged into layers, and artificial neurons analyse input to create an output that is sent to the following perceptron. It is categorized into two states, supervised learning (SL) and unsupervised learning. In unsupervised learning, the network receives input data and identifies patterns or structures within the data thereby condensing the data into a more compact form [5].

In SL, the network is "taught" by receiving guidance during the learning process. In this SL, the network is given the relevant input and output data. The connection between the input and output data is established via the network. SL is considered the most popular and valuable network for formulation purposes [4, 5].

The arrangement of interconnected neurons in a neural network is called network architecture. There are various types of network architecture; among them, a multilayer perceptron (MLP) network is one of the best suitable examples.

X1, X2 & X3 are input variables, and Y is the output. The number of input, output, and hidden layers depends on the condition and the researcher's plan depicted in Fig. 1 [6].

Fig. 1
figure 1

Schematic diagram of artificial neural network

Fuzzy logic

Another tool of AI is fuzzy logic. People extensively use fuzzy logic in problem-solving. When ANN accompanies it, it cooperates to understand the formulation and optimization process [4]. Conventional sense relies on either 'true' or 'false.' So, this hypothesis falls between either in actual or wholly false part. Since the premise is true, the membership function in the true set is 1; otherwise, it is 0 in the false set [4].

Neuro-fuzzy logic

The main motto of fuzzy logic is to give in simple forms. Neural network modelling is necessary to form it. Neuro-fuzzy logic, by its name, is understood that neuro-fuzzy logic is composed of neural networks and fuzzy logic. It combines ANN's learning capacity with generality and fuzzy logic's ability to explore complex concepts. Neuro-fuzzy logic is tightly well suited to process data mining. It can develop good models from data and express the linguistic IF_THEN rules [4, 5].

Genetic algorithm

John Holland introduced genetic algorithms (GA) in the 1970s [5]. Like ANN, GA is also a biologically motivated system. Natural selection, the primary tenet of its genetic variation, imitates the fundamental principles of evolution over a generation. We create a genetic algorithm to select the best and most effective solution. They provide a 'search' strategy that is excellent for optimisation. An iterative procedure would be used to advance the trial population. As part of this procedure, we form a starting population and evaluate each member's fitness [4]. The fittest solutions change into 'parents' of the subsequent generations. It becomes a more ideal answer by including some introduction of recombination and mutation, which produces a larger degree of new stuff in the population. The genetic algorithm needs a criterion of "fitness." It is variable from problem to problem [5].

The best solution is found by creating a number of potential solutions when an ANN and a genetic algorithm are combined. These candidates are selected based on their fitness according to predefined criteria. Generating a new population of solutions is accomplished by utilizing the most effective solution and incorporating crossover and mutation techniques. The process persists until it fulfils the desired requirements, at which juncture the repetition is terminated [4].

An optimization technique is needed to develop a pharmaceutical product to find the best combination of ingredients and techniques. Researchers have proven that the combination of ANN and GA possesses the requisite solutions for developing dosage forms.

Evolutionary computing

It is just a definition of a computational technique that uses heredity, recombination, mutation, and selection to solve a problem. One particular branch of evolutionary algorithms has been used in formulation research [5].

Life cycle of pharmaceutical products

AI can support decision-making, enable rational drug design, determine the best course of a patient’s treatment with personalized medications, accomplish the clinical data produced, and utilised that data to create new drugs in the future [7]. From the lab to the bedside, it is logical to assume that AI will contribute to creating pharmaceutical products. Eularis created the E-VAI analytical and decision-making AI platform, which employs ML algorithms and a user-friendly interface to create analytical roadmaps based on rivals, crucial stakeholders, and the market share currently held to forecast critical factors in pharmaceutical sales [8]. This boosts sluggish sales and gives marketing directors the ability to foresee where to make expenditures. It also helps them allocate resources for optimum market share growth. Figure 2 presents an overview of several AI uses in drug discovery and development.

Fig. 2
figure 2

Applications of AI in various pharmaceutical business subfields, including pharmaceutical product management and drug development

AI in drug discovery

The research and development of new drugs is a challenging, expensive, and lengthy task. On average, the R&D cycle spans around 10–15 years. Despite the significant financial investment made by the pharmaceutical industry, pursuing the next blockbuster drug persists. This R&D is since only one in every ten potential drug candidates completes phase I clinical trials and attains regulatory approval [9, 10]. The cost and time constraints associated with developing newer therapeutic compounds may be a contributing factor in the pharmaceutical industry's acceptance of AI [11].

The tools and technologies employed by AI are valuable in rapidly identifying hit and lead materials, validating drug targets, and optimizing drug structure design, potentially benefiting the healthcare industry by reducing the cost and timeline associated with discovering novel molecules. However, despite these advantages, AI must still overcome significant data hurdles, including the data's complexity, growth, diversity, and ambiguity [12, 13].

The chemical structure that would elicit the desired reaction at the target location may be predicted using a variety of in silico approaches. This structure can then be improved to meet a variety of criteria, such as potency, safety, solubility, permeability, and synthetic tractability. These methods also make it possible to plan the production of the compound and anticipate the molecule's physicochemical characteristics [13, 14].

By utilizing both structure- and ligand-based methods, along with all available data, it is feasible to hasten the elimination of non-lead compounds. Recently, researchers have employed the quantitative structure–activity relationship (QSAR) modelling device for screening potential pharmacologically active compounds from a pool of one million candidates. Moreover, the deep learning approach, an evolution of the earlier ML approach, can now handle the massive amount of data gathered throughout the drug discovery and development procedure [15, 16].

Using a computer model based on the QSAR, large quantities of compounds or certain physicochemical qualities, such as log P or log D, may be swiftly predicted. These models, however, are far from being able to forecast with any degree of accuracy complex biological traits like a compound's efficacy and undesirable side effects. Additionally, QSAR-based models have issues with limited exercise groups, erroneous investigational facts, and a need for more trial validations. To address these problems, researchers can employ newly developed AI methodologies, such as Deep Learning (DL) and pertinent modelling lessons, to assess the safety and effectiveness of pharmaceutical molecules through extensive data showing and study [17, 18].

DL models beat traditional ML techniques in 15 drugs candidate-related absorption, distribution, metabolism, excretion, and toxicity (ADMET) data sets regarding predictability. Drug metabolism sites are identified using artificial intelligence (AI) techniques like XenoSite, FAME, and SMARTCyp. By displaying molecule distributions and properties, the huge virtual chemical space suggests the existence of a molecular topographic map. Chemical space visualization's idea is to collect positional information on nearby molecules to hunt for bioactive compounds; thus, virtual screening (VS) helps choose appropriate molecules for future investigation. PubChem, ChemBank, DrugBank, and ChemDB are a few open-access chemical databases.

For the purpose of locating prospective novel drugs, AI-based QSAR approaches, such as decision trees, support vector machines, random forests, and linear discriminant analysis (LDA), have evolved from QSAR modelling tools [15, 19, 20].

We have included a list of a few AI technologies used throughout the drug development phase in Table 1 to help readers understand. Figure 3 summarizes the various AI models used during drug development methods. Physicochemical characteristics, bioactivity, toxicity, target proteins, drug interactions, drug-protein binding interactions, and de novo synthesis of certain organic synthetic compounds are all predicted by these models [21].

Table 1 The AI techniques/tools used in the drug discovery process
Fig. 3
figure 3

Different applications of AI in drug discovery

AI in drug development

An acceptable dosage form with the essential delivery qualities must then include a unique medicinal component. In this case, AI can take the place of the conventional approach of trial and error [22]. With the use of QSPR, a variety of computational techniques may resolve issues in the formulation design area, such as instability issues, dissolving, porosity, and many more [23]. Decision-support technologies use rule-based algorithms to choose the kind, nature, and amount of the excipients depending on the physicochemical properties of the drug. They also use a feedback loop to keep an eye on and occasionally tweak the entire process [24].

Piroxicam direct-filling hard gelatin capsules were designed using a hybrid method that combines expert systems (ES) and ANN in order to achieve the necessary dissolving profile. Based on the input parameters, the Model Expert System (MES) delivers judgements and recommendations for formulation development. Contrarily, ANN make use of backpropagation learning to link the formulation parameters to the desired outcome, enabling trouble-free formulation creation. The control module collaboratively manages this process [22].

Using a variety of mathematical methods, including computational fluid dynamics (CFD), discrete element modelling (DEM), and the finite element method (FEM), researchers have investigated the effects of the powder's flow property on the die-filling and tablet compression processes [25, 26]. CFD may also be used to examine how tablet shape affects the profile of the tablet's disintegration [27]. Integrating these mathematical models with AI may have a huge positive impact on the rapid manufacturing of pharmaceutical products. Technologies incorporating AI have evolved into versatile tools that find wide application in various stages of drug development. These stages include identifying and validating drug targets, designing new drugs, repurposing existing drugs, enhancing R&D efficiency, aggregating and analysing biomedicine data, and making informed decisions regarding patient enrolment in clinical trials [17, 28, 29]. These prospective applications of AI offer the chance to mitigate bias and human interference while addressing the inefficiencies and uncertainties resulting from traditional drug development approaches [30].

Drug repurposing [31], pharmacological features [32], protein characteristics and efficacy [33], drug combination, drug-target interaction [34], and prediction of potential synthetic methods for drug-like molecules [35] are other uses of AI in the pharmaceutical industry. In addition, the identification of associations between drugs and illnesses and the development of novel biomarkers and therapeutic targets allow for the identification of new pathways and targets utilizing omics analysis [36, 37].

AI in drug formulation

Pharmaceutical sciences have seen various formulations, for example solid dispersions, extrudates, pellets, nanoparticles, and liposomes, arise in addition to standard dosage forms. The name "formulation techniques" is given to these techniques because they empower the development of formulations or incorporate functionality into common dosage forms such as tablets. AI applications in formulation techniques are even more worthwhile to investigate in order to create next-generation drug products with desired efficacy and health outcomes because these methods can successfully address a variety of API issues, such as low solubility, stability, bioavailability, and production capability [6].

Controlled-release tablet formulation

Researchers utilize pharmacokinetic simulations and ANN to develop controlled-release formulations [5]. The ANN model learns sophisticated and specialized abilities from the input and output data units with the use of Chem software. In order to anticipate the best tablet formulations based on two ideal in vitro dissolution-time profiles and two desirable in vivo release profiles, researchers use a sophisticated ANN model. Dissolution is the rate-limiting step in the in vivo absorption of the drug since it is linearly proportional to the amount of the drug taken in vivo. In vitro release patterns are often detected using the difference factors (f1), and similarity factor (f2) [38].

Immediate-release tablets formulation

To boost tablet strength, Turkoglu developed a direct compression tablet formulation utilizing hydrochlorothiazide [39]. In a different study, Kesavan and Peck developed a model of a caffeine tablet formulation to describe the diluting agent and binder content in each formulation, processing variables (type of granulator, method of adding binder), and granule and tablet properties (disintegration time, hardness, and friability). These two analyses demonstrated that neural networks performed better than traditional statistical methods. Kesavan and Peck's findings have so been re-evaluated by academics employing a variety of genetic algorithms and neural networks [40]. This presentation illustrated how the relative relevance of the output attributes and the restrictions placed on the several tiers of components and processing factors determined the ideal formulation [41]. Researchers used neuro-fuzzy computing to analyse the same data and frequently created helpful rules that highlighted the most important aspects of any item [5].

Hard gelatin capsule shell formulation

Developing hard gelatin capsule formulations involves using executive tools like ANN and expert systems (ES). ANN stimulate human mental processes, such as generalization, learning, prediction, and abstraction from domain knowledge. With ANNs, the data and statistics collected during investigative work may be transformed into knowledge very quickly, enabling the manufacturer to generate few domain-specific strategies for forthcoming occurrences or forecast the theoretical preparation’s characteristics [22]. By extending the Expert Network and conducting analysis, Wendy I. Wilson in 2005, created a capsule shell manufacturing of Biopharmaceutical Classification System II drugs, such as carbamazepine, ketoprofen, naproxen, and ibuprofen. Capsugel's expert system, for the formulation of powders in hard gelatin capsules, was used all over the world despite the drawback of just providing a proposed composition. During the initial test, researchers discovered that the system exhibited low prediction accuracy and a significant error rate. Researchers retrained the ANN using a new dataset, resulting in models with an R2 of less than 70%. Lastly, for the model drugs, the smart hybrid system predicted the quantity of drug soluble around 5%. By using only 10% of the newly generated data for cross-validation, the researchers showed that the system was capable of creating a formulation that satisfied its performance requirements. Researchers presented the system's ability to analyse several BCS class II drugs by considering wettability and intrinsic dissolving properties [42].

Solid dispersions (SD)

One or more APIs dispersed in a solid matrix describe solid dispersions [43, 44]. They are currently a practical and affordable approach for enhancing solubility and bioavailability [45]. They have been extensively employed in academics and industry to overcome concerns with API poor solubility. Many AI-based SD studies have used ANNs to optimize the formulations [46,47,48]. Researchers utilized ANNs to enhance the floating and drug release characteristics of SD of Nimodipine prepared with PEG and effervescent mixtures [46]. ANNs were employed to elucidate the relationship between variables such as API concentration, the molar mass of PEG, and temperature in a SD formulated with PVP [49]. Researchers recently developed a model using ML approaches to expect the stability of SD. They employed twenty molecular descriptors to compare eight ML methods. Among these methods, the RF model exhibited the highest estimate precision and provided insights into every input. The top five contributing parameters among the twenty descriptors they picked were the drug loading ratio, relative ambient humidity, storage temperature, preparation temperature, and molecular weight of polymers [50].

Emulsions, microemulsions, and nanoemulsions

Emulsions are biphasic systems with water and oil phases spread over each other and stabilized by an emulsifier [51]. The utilization of micro- and nanoemulsions has the potential to provide a variety of advantages, including increased API bioavailability, superb optical clarity, and improved long-term stability [52,53,54,55]. Researchers have published studies on these systems that utilize AI approaches. Kumar et al. regulated the fatty alcohol content with the use of ANNs to produce a steady o/w emulsion. Particle size, zeta potential, conductance, and viscosity were among the emulsion product properties that the ANNs could accurately predict. They also made it possible to quantify the relative significance of the inputs [56]. Gasperlin et al. successfully predicted the structures of microemulsions by creating two ANNs that can determine the kind of microemulsion from the desired composition or a differential scanning calorimetry (DSC) curve, respectively [57]. Additionally, Agatonovic-Kustrin et al. developed a stable microemulsion formulation for the oral administration of rifampicin and isoniazid using ANN model data for treating the ongoing stage of TB [58]. Amani et al. used ANNs to study potential influences on nanoemulsion particle size and discovered that the final particle size's most important factor was the total energy provided during preparation [59]. In addition, Seyed et al. looked into the component concentrations of nanoemulsion to catch the most stabilized structure with minimum cytotoxicity. They found that emulsifier concentration, which was shown to be the primary determinant of nanoemulsion stability, had no effect on cytotoxicity [60].

Self-emulsifying drug delivery systems (SEDDS)

Drugs, oils, surfactants, and occasionally cosolvents are combined in isotropic ways to create SEDDS [61]. SEDDS offer several advantages due to their physical stability, ease of production, and ability to address concerns regarding low drug bioavailability [62]. SEDDS can effectively tackle various API concerns, including enzymatic degradation, gut wall efflux, solubilization, and bioavailability [63]. Fatouros et al. utilized AI techniques such as neuro-fuzzy networks to create a dynamic lipolysis model that simulates medication absorption and predicts the IVIVC. Without requiring complex settings, the model showed significant prediction skills, indicating its potential for application in forecasting the in vivo behaviour of formulations made of lipids [64]. Utilizing ANNs coupled with I-optimal design, Parikh and Sawant optimized the crucial elements that determine the droplet size of SEDDS. When compared to the quadratic model based on I-optimal design, the ANN-coupled replicas showed the comparative contributions of every factor and were more accurate [65]. Li et al. used multiple linear regression (MLR) and ANN approaches to create quantitative structure-property relationship (QSPR) models that relate the molecular structures of the surfactant, co-surfactant, oil, and drug used in SEDDS with the drug solubility. The researchers found that key factors influencing drug solubility were the ratios of surfactant and oil, as well as the dipole moment and energy of the highest occupied molecular orbital [66].

Other formulation techniques

In addition to these formulation techniques, researchers have applied AI methods to beads and pellets [67,68,69,70,71], microparticles and nanoparticles [72,73,74,75,76,77,78,79,80,81,82,83,84,85], microspheres and nanospheres [86,87,88], liposomes and nanoliposomes [89], colloidal systems [90], micelles [91, 92], and liquid, solid dosage forms [93].

Scaling up AI across the pharmaceutical value chain

AI and ML are operating revolutions through several manufacturing company. Nevertheless, industries reliant on research, such as pharmaceuticals, are witnessing rapid advancements in these technologies. To speed up the delivery of life-saving medications, AI is re-designing the value chain and extracting insights from diverse data sets to make it highly interoperable [94].

Drug discovery

AI can aid labour and resource-intensive manual drug discovery. Biopharmaceutical companies have the opportunity to employ AI models to identify and validate various methods, leveraging the acquired knowledge to improve their predictive capabilities. AI could have a significant impact on generative modelling for molecule design and protein engineering in the field of molecular design. Although manual data entry remains essential for clinical trials, the utilization of AI-powered data flows allows for the integration of trial data from diverse sources, enabling the delivery of standardized digital data that seamlessly and automatically transfers to the relevant systems downstream [94].

Manufacturing

AI in biopharma manufacturing might stream crucial data, anticipate process bottlenecks, evaluate current quality control issues, and recommend necessary improvements without the need for time-consuming manual involvement [94].

It could lower operationally expenses and manual oversight in manufacturing operations by permitting tighter control with:

  • Quality control

  • AI in robotic process automation (RPA)

  • AI mock-ups to augment produce & output

  • AI-coupled prognostic maintenance to lessen appliance interruption.

Pharmaceutical market of AI

Pharmaceutical companies are turning to AI to reduce financial costs and failure risks. The AI market witnessed growth from US$200 million in 2015 to US$700 million in 2018, and it is expected to reach $5 billion by 2024 [95]. Experts predict that the pharmaceutical and medical industries will experience a 40% growth rate from 2017 to 2024 due to the impact of AI. Numerous pharmaceutical firms have invested in AI and are continuing to do so. They have also worked with AI firms to build crucial healthcare solutions. An example is the partnership between DeepMind Technologies, a Google company, and the Royal Free London NHS Foundation Trust for treating critical renal damage. Figure 4 lists critical pharmaceutical businesses and AI players [12].

Fig. 4
figure 4

Leading pharmaceutical firms and their connections to AI companies working in oncology, cardiovascular illness, and CNS disorders. [Pharmaceutical firm’s brand name and logo are subject to individual copyright]

Application of AI

Poly-pharmacology

In disease-related molecular networking, poly-pharmacology is the strategic creation of a therapeutic molecule with the innate capacity to interact with numerous targets or pathways (put, "one disease-multiple target"). One can use this method to create a more effective and less hazardous therapy than the currently available one. Several well-known databases that offer details on biochemical pathways, binding strengths, pharmacological goals, and physiological effects are PubChem, CheMBL, Drug Bank, and Binding DB [96, 97]. The AI system can use this data to probe and discover potential poly-pharmacological drugs selectively. One can use the range of available applications, improved computer capacity, and developments in AI technology to change the drug development process.

Quality control and quality assurance

Developing a pharmaceutical formulation within the given timeframe while ensuring quality necessitates a meticulous and scientifically-driven approach to navigate the intricate process successfully. Gathering information involves capturing data about the characteristics of drug compounds and excipients, their interactions, unit operations, and equipment. Various knowledge applications are utilized, such as heuristics, decision trees, correlation, and first-principal models. This information and knowledge inform the decision-making process for production, selection of excipients, and determination of equipment size [98]. Consequently, AI and its networks, technologies, and tools ensure higher product quality, less waste, and increased profit for manufacturing company. Quality-by-Design (QbD) method ensures the improved quality of the generated goods. This approach makes it easy to comprehend the crucial elements in the pharmaceutical production process that might have an impact on the final product's quality.

Product development

The pharmaceutical sector focuses on accelerating pharmaceutical product development, cutting production costs, and enhancing process design for confirming an active medication. Various expert systems can serve as valuable tools for prompt decision-making in the rapid development of pharmaceutical products. An example of a decision-support tool is the rule-based expert system, which represents domain knowledge as a collection of rules structured as IF-THEN statements. These rules utilize input data to assist in addressing specific problems. Each rule consists of two components: the IF part, which establishes an assumption (such as medication insolubility), and the THEN part, which defines the corresponding action to be taken (such as using a soluble filler to address the solubility issue). This type of system, known as a production system or an expert system, represents the fundamental form of artificial intelligence. It functions based on pre-established rules that are provided as knowledge inputs to guide its operations. The choice of excipients for preparing tablets and capsules can be made using these expert systems. Such a system's regulations detail the system's mechanical, chemical, and physical characteristics and the requirements for the finished item. The system's inference engine uses this information to forecast the kind and number of excipients needed to achieve the requirements. This formula is used to create the product, which is then examined to see if it complies with the necessary standards. This information is then fed into the system, which aids with formula optimization (Fig. 5) [24].

Fig. 5
figure 5

Formulation of tablets and capsules uses rule-based systems

The creation of Logica's product formulation expert system (PFES), anticipated to direct the manufacturing technique using an order of duties, was motivated by the need for quick manufacture of generic formulations. PFES provides a framework for new formulation system development. The specification object, which encompasses the current understanding of the formulation problem, and the formulation object, which represents the current composition of the formulation, are taken into account. This knowledge is subsequently applied to facilitate the development of the formulation. The design of later iterations of PFES involves three levels: physical, task, and control. The control level conducts the studies, while the physical level receives domain information and can be accessed from the task level through a query interface. Expert systems in formulation development have several benefits, such as ensuring a consistent process for regulatory compliance, aiding in novice and professional training, reducing product development time and cost, and freeing up experts to focus on innovation [99].

Developing innovative drug delivery methods with an eye on optimizing efficacy and avoiding adverse effects has generally taken up most of the last ten years. AI has the potential to address certain challenges associated with controlled and conventional drug delivery systems, including issues such as systemic toxicity, narrow therapeutic ranges, and dose adjustments during long-term therapy. To illustrate how AI is utilized in medicines, an example can be given where microchips are employed to manage medication administration, lower systemic toxicity, narrow the therapeutic window, and reduce adverse effects. AI has made significant advancements in enhancing the efficacy, safety, and adherence of patients by innovating implanted drug delivery devices capable of regulating drug release timing and concentration. This strategy of using AI in managing chronic diseases like diabetes, which require prompt treatment and continuous monitoring, is very advantageous. Neural networks, fuzzy logic, integrators, and differentiators have all been used to develop control systems. It is crucial to monitor glucose levels in diabetic patients and provide insulin regularly to treat the condition. As mentioned earlier, the utilization of microchips can assist in the effective management of regular blood glucose level monitoring and insulin administration. The combination of glucose sensors, mathematical models, and control algorithms holds promise for facilitating this objective. Conventional therapeutic methods that have been utilized over time face certain limitations, which can be overcome by integrating information technology, wireless communication, and ANNs into standard therapeutic procedures. As an illustration, wireless connections can be utilized to transmit orders from outside sources to units. The data from these communications are gathered and tracked to control the drug's distribution [100, 101].

Pharmaceutical manufacturing

Continual production is a valuable strategy for reducing lot-to-lot variances. The FDA recommends a non-stop processing strategy to limit the variation of final products and patient outcomes. One can use various process analytical technology (PAT) tools, efficient and cost-effective to control uninterrupted production. Through automated ML, using AI in conjunction with PAT can improve the process overall and help regulate the production process [102].

Companies that produce biopharmaceutical products can benefit from data science by integrating logistics into their processing operations. Ensuring the proper upkeep of these variables is considered a crucial regulatory consideration due to the utilization of engineered living cells in the production of biopharmaceuticals. Managing and monitoring multiple factors are necessary to maintain the purity and consistency of the manufactured product. As a result, most of the world's leading chemical businesses frequently employ big data to improve vaccine production output and keep an eye on product quality [34].

Drug synergism and antagonism prediction

When treating someone over time, studying how different drugs interact is essential. This can reduce the dose needed and prevent harmful side effects from taking multiple medications together. SnuGen utilises the master regulator inference algorithm (MARINA) to forecast synergism and antagonism. The MARINA method was established to have the ability to predict synergism with an approximate accuracy of 56%. This method clarifies the "Mater Regulator" genes which may be applied to selecting beneficial descriptors for ML techniques. One can use network-based Laplacian regularized least square methods to predict synergism and antagonistic drug interactions. As was covered in the preceding section, various AI techniques combined with medication therapy can be advantageous in multiple ways. For proper prediction, a level of confidence percentage in the range of 0.7–0.9 has been attained, comparable to the performance of the most automatic prediction system. The prediction scores obtained using the various ML techniques do not significantly differ. While ANNs, Random Forest, and SVM all have benefits and drawbacks, choosing the correct input parameters is the main issue when utilizing AI for combo therapy. One must use the parameters that determine the quality of the prediction model when developing prediction algorithms [18].

Nanorobots for drug delivery

Integrated circuits, sensors, a power source, and a secure data backup constitute the primary components of nanorobots, which are maintained by computational technologies like AI [101]. They are designed with programming to prevent collisions, detect targets, locate and engage with them, and subsequently eliminate them from the body. Advancements in nano/microrobot technology empower them to navigate to specific locations within the body by leveraging physiological indicators like pH. This progress enhances their efficacy and reduces the occurrence of systemic side effects [103]. When creating implantable nanorobots for the controlled administration of drugs and genes, factors such as dosage customization, sustained release, and regulated release must be carefully considered. The automation of drug release necessitates the utilization of AI tools, such as neural networks (NNs), fuzzy logic, and integrators [104]. Microchip implants serve dual purposes, enabling both scheduled release of substances and precise localization within the body.

Nanomedicine

Nanomedicines merge nanotechnology and drugs to diagnose, treat, and monitor complex ailments like HIV, cancer, malaria, asthma, and various inflammatory conditions. Due to their enhanced treatment efficacy, the utilization of nanoparticle-modified drug delivery systems has experienced substantial growth in both the therapeutic and diagnostic domains [105, 106]. Combining AI with nanotechnology could solve numerous issues in product development [107, 108]. Through computational analysis, a nanosuspension of methotrexate was developed by examining the energy generated during the interaction between drug molecules and closely monitoring any conditions that could lead to formulation aggregation. Coarse-grained simulation and chemical computation can be employed to evaluate the interactions between drugs and dendrimers, as well as to analyse the encapsulation of drugs within the dendrimer structure [23].

Furthermore, researchers can investigate the impact of surface chemistry on the internalization of nanoparticles into cells using tools such as LAMMPS and GROMACS 4. The utilization of AI facilitated the development of silicasomes, which are a combination of multifunctional mesoporous silica nanoparticles loaded with irinotecan and the tumour-penetrating peptide iRGD. The inclusion of iRGD enhances the transcytosis of silicasomes, resulting in improved treatment outcomes and increased overall survival. As a result, there has been a significant three- to fourfold increase in the absorption of silicasomes [106].

Predicting the mode-of-action of compounds using AI

The prospect of an AI platform that can predict drugs' on- and off-target effects as well as in vivo safety profiles prior to synthesizing has medicinal chemists, in particular, thrilled. The existence of such platforms reduces the amount of time, money, and attrition rates needed to create new medications. Among these platforms are DeepTox, which predicts the toxicity of new drugs, and PrOCTOR, which assesses the probability of toxicity during clinical trials [109, 110]. If a comprehensive and precise dataset containing information on the toxicity and therapeutic characteristics of a wide range of drugs becomes available, the industry has the potential to enhance the predictive accuracy of these platforms by sharing and exchanging data.

As a substitute for chemo-proteomics, SPiDER, a novel AI tool, was recently created [111] to promote natural products for drug development. As a proof-of-concept, SPiDER was utilized to predict the molecular target of lapachone, a natural naphthoquinone with promising antitumour properties currently in clinical development. The platform expected that 5-lipoxygenase (5-LO) would be modulated allosterically and reversibly by lapachone. Through the use of a 5-LO functional test, the prediction is verified. An alternative AI methodology, known as read-across structure-activity relationships (RASAR) [112], has demonstrated the ability to effectively forecast the toxicity of unfamiliar compounds. By leveraging a vast chemical library and establishing connections between molecular structures and hazardous traits, RASAR proves valuable in this prediction process.

Conclusion and future prospect

In conclusion, the function of AI in pharmaceutical formulation and development is rapidly expanding, bringing a host of benefits to the industry. AI has already demonstrated its ability to analyse large data sets, optimize drug formulations, and streamline clinical trials. By doing so, the duration and expenses associated with drug development have been diminished, concurrently heightening the precision and efficacy of the complete process.

The prospects for AI in pharmaceutical formulation and development are up-and-coming. As AI continues to evolve and improve, it is predictable to show an even more significant function in drug development, aiding researchers in identifying new drug targets, drug interactions, and patient populations most likely to benefit from treatment.

Furthermore, as AI systems become more advanced, they can simulate biological systems more accurately, allowing researchers to develop more personalized and effective treatments. This, in turn, will lead to more efficient and targeted drug development and personalized medicine.

So, the role of AI in pharmaceutical formulation and development has already proven to be transformative and is expected to continue to revolutionize the industry in the coming years. By leveraging the power of AI, researchers can unlock new insights into complex diseases, create more effective treatments, and improve patient outcomes on a global scale.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author, upon reasonable request.

Abbreviations

AI:

Artificial intelligence

ML:

Machine learning

R&D:

Research and development

ANN:

Artificial neural network

CNN:

Convolutional neural network

SL:

Supervised learning

DL:

Deep learning

GA:

Genetic algorithms

QSAR:

Quantitative structural activity relationship

LDA:

Linear discriminant analysis

NLP:

Natural language processing

MES:

Model expert systems

CFD:

Computational fluids dynamics

DEM:

Discreet element modelling

FEM:

Finite element modelling

API:

Active pharmaceutical ingredients

SD:

Solid dispersions

SEDDS:

Self-emulsifying drug delivery system

MLR:

Multiple linear regression

PFES:

Product formulation expert systems

RASAR:

Read across structure activity relationship

References

  1. Singh D, Singh R, Gehlot A et al (2022) An imperative role of digitalization in monitoring cattle health for sustainability. Electronics 11:2702. https://doi.org/10.3390/electronics11172702

    Article  Google Scholar 

  2. Kalyane D, Sanap G, Paul D, et al (2020) Artificial intelligence in the pharmaceutical sector: current scene and future prospect. In: The future of pharmaceutical product development and research, pp 73–107. Elsevier

  3. Paul D, Sanap G, Shenoy S et al (2021) Artificial intelligence in drug discovery and development. Drug Discov Today 26:80–93. https://doi.org/10.1016/j.drudis.2020.10.010

    Article  CAS  PubMed  Google Scholar 

  4. Gao M, Liu S, Chen J et al (2021) Potential of Raman spectroscopy in facilitating pharmaceutical formulations development—an AI perspective. Int J Pharm 597:120334. https://doi.org/10.1016/j.ijpharm.2021.120334

    Article  CAS  PubMed  Google Scholar 

  5. Landin M, Rowe RC (2013) Artificial neural networks technology to model, understand, and optimize drug formulations. In: Formulation tools for pharmaceutical development, pp 7–37. Elsevier

  6. Al Kuwaiti A, Nazer K, Al-Reedy A et al (2023) a review of the role of artificial intelligence in healthcare. J Pers Med 13:951. https://doi.org/10.3390/jpm13060951

    Article  PubMed  PubMed Central  Google Scholar 

  7. Duch W, Swaminathan K, Meller J (2007) Artificial intelligence approaches for rational drug design and discovery. Curr Pharm Des 13:1497–1508. https://doi.org/10.2174/138161207780765954

    Article  CAS  PubMed  Google Scholar 

  8. Blasiak A, Khong J, Kee T (2020) CURATE.AI: optimizing personalized medicine with artificial intelligence. SLAS Technol 25:95–105. https://doi.org/10.1177/2472630319890316

    Article  CAS  PubMed  Google Scholar 

  9. Baronzio G, Parmar G, Baronzio M (2015) Overview of methods for overcoming hindrance to drug delivery to tumors, with special attention to tumor interstitial fluid. Front Oncol. https://doi.org/10.3389/fonc.2015.00165

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mishra SS, Behera PK, Kar B, Ray RC (2018) Advances in probiotics, prebiotics and nutraceuticals. Innov Technol Fermented Food Beverage Ind. https://doi.org/10.1007/978-3-319-74820-7_7

    Article  Google Scholar 

  11. Waring MJ, Arrowsmith J, Leach AR et al (2015) An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat Rev Drug Discov 14:475–486. https://doi.org/10.1038/nrd4609

    Article  CAS  PubMed  Google Scholar 

  12. Martin L, Hutchens M, Hawkins C, Radnov A (2017) How much do clinical trials cost? Nat Rev Drug Discov 16:381–382. https://doi.org/10.1038/nrd.2017.70

    Article  CAS  PubMed  Google Scholar 

  13. Mak K-K, Pichika MR (2019) Artificial intelligence in drug development: present status and future prospects. Drug Discov Today 24:773–780. https://doi.org/10.1016/j.drudis.2018.11.014

    Article  PubMed  Google Scholar 

  14. Sellwood MA, Ahmed M, Segler MH, Brown N (2018) Artificial intelligence in drug discovery. Future Med Chem 10:2025–2028. https://doi.org/10.4155/fmc-2018-0212

    Article  CAS  PubMed  Google Scholar 

  15. Petitjean M, Camproux A (2016) In silico medicinal chemistry: computational methods to support drug design. ChemMedChem 11:1480–1481. https://doi.org/10.1002/cmdc.201600165

    Article  CAS  Google Scholar 

  16. Zhang L, Tan J, Han D, Zhu H (2017) From machine learning to deep learning: progress in machine intelligence for rational drug discovery. Drug Discov Today 22:1680–1685. https://doi.org/10.1016/j.drudis.2017.08.010

    Article  PubMed  Google Scholar 

  17. Zhang Y, Zhang G, Shang Q (2017) Computer-aided clinical trial recruitment based on domain-specific language translation: a case study of retinopathy of prematurity. J Healthc Eng 2017:1–9. https://doi.org/10.1155/2017/7862672

    Article  CAS  Google Scholar 

  18. Zhu H (2020) Big data and artificial intelligence modeling for drug discovery. Annu Rev Pharmacol Toxicol 60:573–589. https://doi.org/10.1146/annurev-pharmtox-010919-023324

    Article  CAS  PubMed  Google Scholar 

  19. Ciallella HL, Zhu H (2019) Advancing computational toxicology in the big data era by artificial intelligence: data-driven and mechanism-driven modeling for chemical toxicity. Chem Res Toxicol 32:536–547. https://doi.org/10.1021/acs.chemrestox.8b00393

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jain N, Gupta S, Sapre N, Sapre NS (2015) In silico de novo design of novel NNRTIs: a bio-molecular modelling approach. RSC Adv 5:14814–14827. https://doi.org/10.1039/C4RA15478A

    Article  CAS  Google Scholar 

  21. Wang Y, Guo Y, Kuang Q et al (2015) A comparative study of family-specific protein–ligand complex affinity prediction based on random forest approach. J Comput Aided Mol Des 29:349–360. https://doi.org/10.1007/s10822-014-9827-y

    Article  CAS  PubMed  Google Scholar 

  22. Vyas M, Thakur S, Riyaz B et al (2018) Artificial intelligence: the beginning of a new era in pharmacy profession. Asian J Pharm 12:72–76. https://doi.org/10.22377/ajp.v12i02.2317

  23. Mintong G, Gunjan K, Wendy W et al (2002) A prototype intelligent hybrid system for hard gelatin capsule formulation development. Pharm Tech 26:44–60

  24. Mehta CH, Narayan R, Nayak UY (2019) Computational modeling for formulation design. Drug Discov Today 24:781–788. https://doi.org/10.1016/j.drudis.2018.11.018

    Article  CAS  PubMed  Google Scholar 

  25. Zhao C, Jain A, Hailemariam L et al (2006) Toward intelligent decision support for pharmaceutical product development. J Pharm Innov 1:23–35. https://doi.org/10.1007/BF02784878

    Article  Google Scholar 

  26. Rantanen J, Khinast J (2015) The future of pharmaceutical manufacturing sciences. J Pharm Sci 104:3612–3638. https://doi.org/10.1002/jps.24594

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ketterhagen WR, Am Ende MT, Hancock BC (2009) Process modeling in the pharmaceutical industry using the discrete element method. J Pharm Sci 98:442–470. https://doi.org/10.1002/jps.21466

    Article  CAS  PubMed  Google Scholar 

  28. Chen W, Desai D, Good D et al (2016) Mathematical model-based accelerated development of extended-release metformin hydrochloride tablet formulation. AAPS PharmSciTech 17:1007–1013. https://doi.org/10.1208/s12249-015-0423-9

    Article  CAS  PubMed  Google Scholar 

  29. Huang Z, Juarez JM, Li X (2017) Data mining for biomedicine and healthcare. J Healthc Eng 2017:1–2. https://doi.org/10.1155/2017/7107629

    Article  Google Scholar 

  30. Mamoshina P, Vieira A, Putin E, Zhavoronkov A (2016) Applications of deep learning in biomedicine. Mol Pharm 13:1445–1454. https://doi.org/10.1021/acs.molpharmaceut.5b00982

    Article  CAS  PubMed  Google Scholar 

  31. Seddon G, Lounnas V, McGuire R et al (2012) Drug design for ever, from hype to hope. J Comput Aided Mol Des 26:137–150. https://doi.org/10.1007/s10822-011-9519-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schneider G (2018) Automating drug discovery. Nat Rev Drug Discov 17:97–113. https://doi.org/10.1038/nrd.2017.232

    Article  CAS  PubMed  Google Scholar 

  33. Klopman G, Chakravarti SK, Zhu H et al (2004) ESP: a method to predict toxicity and pharmacological properties of chemicals using multiple MCASE databases. J Chem Inf Comput Sci 44:704–715. https://doi.org/10.1021/ci030298n

    Article  CAS  PubMed  Google Scholar 

  34. Menden MP, Iorio F, Garnett M et al (2013) Machine learning prediction of cancer cell sensitivity to drugs based on genomic and chemical properties. PLoS One 8:e61318. https://doi.org/10.1371/journal.pone.0061318

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Challa S, Potumarthi R (2013) Chemometrics-based process analytical technology (PAT) tools: applications and adaptation in pharmaceutical and biopharmaceutical industries. Appl Biochem Biotechnol 169:66–76. https://doi.org/10.1007/s12010-012-9950-y

    Article  CAS  PubMed  Google Scholar 

  36. Matthews H, Hanison J, Nirmalan N (2016) “Omics”-informed drug and biomarker discovery: opportunities, challenges and future perspectives. Proteomes 4:28. https://doi.org/10.3390/proteomes4030028

    Article  PubMed  PubMed Central  Google Scholar 

  37. Merk D, Friedrich L, Grisoni F, Schneider G (2018) De Novo design of bioactive small molecules by artificial intelligence. Mol Inform 37:1700153. https://doi.org/10.1002/minf.201700153

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hamet P, Tremblay J (2017) Artificial intelligence in medicine. Metabolism 69:S36–S40. https://doi.org/10.1016/j.metabol.2017.01.011

    Article  CAS  Google Scholar 

  39. Hughes J, Rees S, Kalindjian S, Philpott K (2011) Principles of early drug discovery. Br J Pharmacol 162:1239–1249. https://doi.org/10.1111/j.1476-5381.2010.01127.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Katsila T, Spyroulias GA, Patrinos GP, Matsoukas M-T (2016) Computational approaches in target identification and drug discovery. Comput Struct Biotechnol J 14:177–184. https://doi.org/10.1016/j.csbj.2016.04.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mohs RC, Greig NH (2017) Drug discovery and development: role of basic biological research. Alzheimers Dement Transl Res Clin Interv 3:651–657. https://doi.org/10.1016/j.trci.2017.10.005

    Article  Google Scholar 

  42. Emig D, Ivliev A, Pustovalova O et al (2013) Drug target prediction and repositioning using an integrated network-based approach. PLoS One 8:e60618. https://doi.org/10.1371/journal.pone.0060618

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Vora LK, Gholap AD, Jetha K et al (2023) Artificial intelligence in pharmaceutical technology and drug delivery design. Pharmaceutics 15:1916. https://doi.org/10.3390/pharmaceutics15071916

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen Y, McCall TW, Baichwal AR, Meyer MC (1999) The application of an artificial neural network and pharmacokinetic simulations in the design of controlled-release dosage forms. J Controlled Release 59:33–41. https://doi.org/10.1016/S0168-3659(98)00171-0

    Article  CAS  Google Scholar 

  45. Kesavan JG, Peck GE (1996) Pharmaceutical granulation and tablet formulation using neural networks. Pharm Dev Technol 1:391–404. https://doi.org/10.3109/10837459609031434

    Article  CAS  PubMed  Google Scholar 

  46. Colbourn E, Rowe R (1996) Modelling and optimization of a formulation using neural networks and genetic algorithms. Pharm Technol Eur 8:46–55

    Google Scholar 

  47. Wilson WI, Peng Y, Augsburger LL (2005) Generalization of a prototype intelligent hybrid system for hard gelatin capsule formulation development. AAPS PharmSciTech 6:E449–E457. https://doi.org/10.1208/pt060356

    Article  PubMed  PubMed Central  Google Scholar 

  48. Sekiguchi K, Obi N (1961) Studies on absorption of eutectic mixture. I. A comparison of the behavior of eutectic mixture of sulfathiazole and that of ordinary sulfathiazole in man. Chem Pharm Bull (Tokyo) 9:866–872. https://doi.org/10.1248/cpb.9.866

    Article  CAS  Google Scholar 

  49. Huang Y, Dai W-G (2014) Fundamental aspects of solid dispersion technology for poorly soluble drugs. Acta Pharm Sin B 4:18–25. https://doi.org/10.1016/j.apsb.2013.11.001

    Article  PubMed  Google Scholar 

  50. Serajuddin ATM (1999) Solid dispersion of poorly water-soluble drugs: early promises, subsequent problems, and recent breakthroughs. J Pharm Sci 88:1058–1066. https://doi.org/10.1021/js980403l

    Article  CAS  PubMed  Google Scholar 

  51. Barmpalexis P, Kachrimanis K, Georgarakis E (2011) Solid dispersions in the development of a nimodipine floating tablet formulation and optimization by artificial neural networks and genetic programming. Eur J Pharm Biopharm 77:122–131. https://doi.org/10.1016/j.ejpb.2010.09.017

    Article  CAS  PubMed  Google Scholar 

  52. Patel TB, Patel L, Patel TR, Suhagia B (2015) Artificial neural network as tool for quality by design in formulation development of solid dispersion of fenofibrate. Bull Pharm Res 5:20–27

    CAS  Google Scholar 

  53. Barmpalexis P, Koutsidis I, Karavas E et al (2013) Development of PVP/PEG mixtures as appropriate carriers for the preparation of drug solid dispersions by melt mixing technique and optimization of dissolution using artificial neural networks. Eur J Pharm Biopharm 85:1219–1231. https://doi.org/10.1016/j.ejpb.2013.03.013

    Article  CAS  PubMed  Google Scholar 

  54. Han R, Xiong H, Ye Z et al (2019) Predicting physical stability of solid dispersions by machine learning techniques. J Controlled Release 311–312:16–25. https://doi.org/10.1016/j.jconrel.2019.08.030

    Article  CAS  Google Scholar 

  55. Khan BA (2011) Basics of pharmaceutical emulsions: a review. Afr J Pharm Pharmacol. https://doi.org/10.5897/AJPP11.698

    Article  Google Scholar 

  56. Bagwe RP, Kanicky JR, Palla BJ et al (2001) Improved drug delivery using microemulsions: rationale, recent progress, and new horizons. Crit Rev Ther Drug Carrier Syst 18:77–140

    CAS  PubMed  Google Scholar 

  57. McClements DJ (2012) Nanoemulsions versus microemulsions: terminology, differences, and similarities. Soft Matter 8:1719–1729. https://doi.org/10.1039/C2SM06903B

    Article  CAS  Google Scholar 

  58. Sarker D (2005) Engineering of nanoemulsions for drug delivery. Curr Drug Deliv 2:297–310. https://doi.org/10.2174/156720105774370267

    Article  CAS  PubMed  Google Scholar 

  59. Solans C, Izquierdo P, Nolla J et al (2005) Nano-emulsions. Curr Opin Colloid Interface Sci 10:102–110. https://doi.org/10.1016/j.cocis.2005.06.004

    Article  CAS  Google Scholar 

  60. Tenjarla S (1999) Microemulsions: an overview and pharmaceutical applications. Crit Rev Ther Drug Carrier Syst 16:461–521

    Article  CAS  PubMed  Google Scholar 

  61. Kumar K, Panpalia G, Priyadarshini S (2011) Application of artificial neural networks in optimizing the fatty alcohol concentration in the formulation of an O/W emulsion. Acta Pharm 61:249–256. https://doi.org/10.2478/v10007-011-0013-7

    Article  CAS  PubMed  Google Scholar 

  62. Gašperlin M, Podlogar F, Šibanc R (2008) Evolutionary artificial neural networks as tools for predicting the internal structure of microemulsions. J Pharm Pharm Sci. https://doi.org/10.18433/J3F594

    Article  PubMed  Google Scholar 

  63. Agatonovic-Kustrin S, Glass BD, Wisch MH, Alany RG (2003) Prediction of a stable microemulsion formulation for the oral delivery of a combination of antitubercular drugs using ANN methodology. Pharm Res 20:1760–1765. https://doi.org/10.1023/b:pham.0000003372.56993.39

    Article  CAS  PubMed  Google Scholar 

  64. Amani A, York P, Chrystyn H et al (2008) Determination of factors controlling the particle size in nanoemulsions using artificial neural networks. Eur J Pharm Sci Off J Eur Fed Pharm Sci 35:42–51. https://doi.org/10.1016/j.ejps.2008.06.002

    Article  CAS  Google Scholar 

  65. Seyedhassantehrani N, Karimi R, Tavoosidana G, Amani A (2017) Concurrent study of stability and cytotoxicity of a novel nanoemulsion system - an artificial neural networks approach. Pharm Dev Technol 22:383–389. https://doi.org/10.1080/10837450.2016.1212878

    Article  CAS  PubMed  Google Scholar 

  66. Charman SA, Charman WN, Rogge MC et al (1992) Self-emulsifying drug delivery systems: formulation and biopharmaceutic evaluation of an investigational lipophilic compound. Pharm Res 9:87–93. https://doi.org/10.1023/a:1018987928936

    Article  CAS  PubMed  Google Scholar 

  67. Gursoy RN, Benita S (2004) Self-emulsifying drug delivery systems (SEDDS) for improved oral delivery of lipophilic drugs. Biomed Pharmacother Biomedecine Pharmacother 58:173–182. https://doi.org/10.1016/j.biopha.2004.02.001

    Article  CAS  Google Scholar 

  68. Singh B, Bandopadhyay S, Kapil R et al (2009) Self-emulsifying drug delivery systems (SEDDS): formulation development, characterization, and applications. Crit Rev Ther Drug Carrier Syst 26:427–521. https://doi.org/10.1615/critrevtherdrugcarriersyst.v26.i5.10

    Article  CAS  PubMed  Google Scholar 

  69. Fatouros DG, Nielsen FS, Douroumis D et al (2008) In vitro-in vivo correlations of self-emulsifying drug delivery systems combining the dynamic lipolysis model and neuro-fuzzy networks. Eur J Pharm Biopharm Off J Arbeitsgemeinschaft Pharm Verfahrenstechnik EV 69:887–898. https://doi.org/10.1016/j.ejpb.2008.01.022

    Article  CAS  Google Scholar 

  70. Parikh KJ, Sawant KK (2018) Comparative study for optimization of pharmaceutical self-emulsifying pre-concentrate by design of experiment and artificial neural network. AAPS PharmSciTech 19:3311–3321. https://doi.org/10.1208/s12249-018-1173-2

    Article  CAS  PubMed  Google Scholar 

  71. Li C-W, Yang S-Y, He R et al (2011) Development of quantitative structure-property relationship models for self-emulsifying drug delivery system of 2-aryl propionic acid NSAIDs. J Nanomater 2011:1–12. https://doi.org/10.1155/2011/206320

    Article  CAS  Google Scholar 

  72. Giang Thi Thu V, Nghia Thi P, Huyen Thi N et al (2020) Application of the artificial neural network to optimize the formulation of self-nanoemulsifying drug delivery system containing rosuvastatin. J Appl Pharm Sci. https://doi.org/10.7324/JAPS.2020.10901

    Article  Google Scholar 

  73. Huang Y, Yao Q, Zhu C et al (2015) Comparison of novel granulated pellet-containing tablets and traditional pellet-containing tablets by artificial neural networks. Pharm Dev Technol 20:670–675. https://doi.org/10.3109/10837450.2014.910809

    Article  CAS  PubMed  Google Scholar 

  74. Mendyk A, Kleinebudde P, Thommes M et al (2010) Analysis of pellet properties with use of artificial neural networks. Eur J Pharm Sci Off J Eur Fed Pharm Sci 41:421–429. https://doi.org/10.1016/j.ejps.2010.07.010

    Article  CAS  Google Scholar 

  75. Peh KK, Lim CP, Quek SS, Khoh KH (2000) Use of artificial neural networks to predict drug dissolution profiles and evaluation of network performance using similarity factor. Pharm Res 17:1384–1388. https://doi.org/10.1023/a:1007578321803

    Article  CAS  PubMed  Google Scholar 

  76. Sankalia MG, Mashru RC, Sankalia JM, Sutariya VB (2005) Papain entrapment in alginate beads for stability improvement and site-specific delivery: physicochemical characterization and factorial optimization using neural network modeling. AAPS PharmSciTech 6:E209-222. https://doi.org/10.1208/pt060231

    Article  PubMed  PubMed Central  Google Scholar 

  77. Vaithiyalingam S, Khan MA (2002) Optimization and characterization of controlled release multi-particulate beads formulated with a customized cellulose acetate butyrate dispersion. Int J Pharm 234:179–193. https://doi.org/10.1016/s0378-5173(01)00959-0

    Article  CAS  PubMed  Google Scholar 

  78. Asadi H, Rostamizadeh K, Salari D, Hamidi M (2011) Preparation of biodegradable nanoparticles of tri-block PLA-PEG-PLA copolymer and determination of factors controlling the particle size using artificial neural network. J Microencapsul 28:406–416. https://doi.org/10.3109/02652048.2011.576784

    Article  CAS  PubMed  Google Scholar 

  79. Baharifar H, Amani A (2017) Size, loading efficiency, and cytotoxicity of albumin-loaded chitosan nanoparticles: an artificial neural networks study. J Pharm Sci 106:411–417. https://doi.org/10.1016/j.xphs.2016.10.013

    Article  CAS  PubMed  Google Scholar 

  80. Hashad RA, Ishak RAH, Fahmy S et al (2016) Chitosan-tripolyphosphate nanoparticles: optimization of formulation parameters for improving process yield at a novel pH using artificial neural networks. Int J Biol Macromol 86:50–58. https://doi.org/10.1016/j.ijbiomac.2016.01.042

    Article  CAS  PubMed  Google Scholar 

  81. Shahsavari S, Rezaie Shirmard L, Amini M, Abedin Dokoosh F (2017) Application of artificial neural networks in the design and optimization of a nanoparticulate fingolimod delivery system based on biodegradable Poly(3-Hydroxybutyrate-Co-3-Hydroxyvalerate). J Pharm Sci 106:176–182. https://doi.org/10.1016/j.xphs.2016.07.026

    Article  CAS  PubMed  Google Scholar 

  82. Shalaby KS, Soliman ME, Casettari L et al (2014) Determination of factors controlling the particle size and entrapment efficiency of noscapine in PEG/PLA nanoparticles using artificial neural networks. Int J Nanomed 9:4953–4964. https://doi.org/10.2147/IJN.S68737

    Article  CAS  Google Scholar 

  83. Leonardi D, Salomón CJ, Lamas MC, Olivieri AC (2009) Development of novel formulations for Chagas’ disease: optimization of benznidazole chitosan microparticles based on artificial neural networks. Int J Pharm 367:140–147. https://doi.org/10.1016/j.ijpharm.2008.09.036

    Article  CAS  PubMed  Google Scholar 

  84. Li Y, Abbaspour MR, Grootendorst PV et al (2015) Optimization of controlled release nanoparticle formulation of verapamil hydrochloride using artificial neural networks with genetic algorithm and response surface methodology. Eur J Pharm Biopharm Off J Arbeitsgemeinschaft Pharm Verfahrenstechnik EV 94:170–179. https://doi.org/10.1016/j.ejpb.2015.04.028

    Article  CAS  Google Scholar 

  85. Metwally AA, Hathout RM (2015) Computer-assisted drug formulation design: novel approach in drug delivery. Mol Pharm 12:2800–2810. https://doi.org/10.1021/mp500740d

    Article  CAS  PubMed  Google Scholar 

  86. Rizkalla N, Hildgen P (2005) Artificial neural networks: comparison of two programs for modeling a process of nanoparticle preparation. Drug Dev Ind Pharm 31:1019–1033. https://doi.org/10.1080/03639040500306294

    Article  CAS  PubMed  Google Scholar 

  87. Rodríguez-Dorado R, Landín M, Altai A et al (2018) A novel method for the production of core-shell microparticles by inverse gelation optimized with artificial intelligent tools. Int J Pharm 538:97–104. https://doi.org/10.1016/j.ijpharm.2018.01.023

    Article  CAS  PubMed  Google Scholar 

  88. Youshia J, Ali ME, Lamprecht A (2017) Artificial neural network based particle size prediction of polymeric nanoparticles. Eur J Pharm Biopharm Off J Arbeitsgemeinschaft Pharm Verfahrenstechnik EV 119:333–342. https://doi.org/10.1016/j.ejpb.2017.06.030

    Article  CAS  Google Scholar 

  89. Hassanzadeh P, Atyabi F, Dinarvand R (2017) Application of modelling and nanotechnology-based approaches: the emergence of breakthroughs in theranostics of central nervous system disorders. Life Sci 182:93–103. https://doi.org/10.1016/j.lfs.2017.06.001

    Article  CAS  PubMed  Google Scholar 

  90. Le TC, Mulet X, Burden FR, Winkler DA (2013) Predicting the complex phase behavior of self-assembling drug delivery nanoparticles. Mol Pharm 10:1368–1377. https://doi.org/10.1021/mp3006402

    Article  CAS  PubMed  Google Scholar 

  91. Padhi R, Bhardhwaj JR (2009) An adaptive drug delivery design using neural networks for effective treatment of infectious diseases: a simulation study. Comput Methods Programs Biomed 94:207–222. https://doi.org/10.1016/j.cmpb.2008.12.010

    Article  PubMed  Google Scholar 

  92. Yüksel N, Türkoglu M, Baykara T (2000) Modelling of the solvent evaporation method for the preparation of controlled release acrylic microspheres using neural networks. J Microencapsul 17:541–551. https://doi.org/10.1080/026520400417603

    Article  PubMed  Google Scholar 

  93. Labouta HI, el-Khordagui LK, Molokhia AM, Ghaly GM (2009) Multivariate modeling of encapsulation and release of an ionizable drug from polymer microspheres. J Pharm Sci 98:4603–4615. https://doi.org/10.1002/jps.21753

    Article  CAS  PubMed  Google Scholar 

  94. Zaki MR, Varshosaz J, Fathi M (2015) Preparation of agar nanospheres: comparison of response surface and artificial neural network modeling by a genetic algorithm approach. Carbohydr Polym 122:314–320. https://doi.org/10.1016/j.carbpol.2014.12.031

    Article  CAS  PubMed  Google Scholar 

  95. Moussa HG, Husseini GA, Abel-Jabbar N, Ahmad SE (2017) Use of model predictive control and artificial neural networks to optimize the ultrasonic release of a model drug from liposomes. IEEE Trans Nanobiosci 16:149–156. https://doi.org/10.1109/TNB.2017.2661322

    Article  Google Scholar 

  96. Agatonovic-Kustrin S, Alany RG (2001) Role of genetic algorithms and artificial neural networks in predicting the phase behavior of colloidal delivery systems. Pharm Res 18:1049–1055. https://doi.org/10.1023/a:1010913017092

    Article  CAS  PubMed  Google Scholar 

  97. Husseini GA, Abdel-Jabbar NM, Mjalli FS, Pitt WG (2007) Modeling and sensitivity analysis of acoustic release of Doxorubicin from unstabilized pluronic P105 using an artificial neural network model. Technol Cancer Res Treat 6:49–56. https://doi.org/10.1177/153303460700600107

    Article  CAS  PubMed  Google Scholar 

  98. Siepmann J, Siepmann F (2008) Mathematical modeling of drug delivery. Int J Pharm 364:328–343. https://doi.org/10.1016/j.ijpharm.2008.09.004

    Article  CAS  PubMed  Google Scholar 

  99. Wang W, Ye Z, Gao H, Ouyang D (2021) Computational pharmaceutics—a new paradigm of drug delivery. J Controlled Release 338:119–136. https://doi.org/10.1016/j.jconrel.2021.08.030

    Article  CAS  Google Scholar 

  100. Husseini GA, Mjalli FS, Pitt WG, Abdel-Jabbar NM (2009) Using artificial neural networks and model predictive control to optimize acoustically assisted Doxorubicin release from polymeric micelles. Technol Cancer Res Treat 8:479–488. https://doi.org/10.1177/153303460900800609

    Article  CAS  PubMed  Google Scholar 

  101. Barmpalexis P, Karagianni A, Nikolakakis I, Kachrimanis K (2018) Artificial neural networks (ANNs) and partial least squares (PLS) regression in the quantitative analysis of cocrystal formulations by Raman and ATR-FTIR spectroscopy. J Pharm Biomed Anal 158:214–224. https://doi.org/10.1016/j.jpba.2018.06.004

    Article  CAS  PubMed  Google Scholar 

  102. Sjödin D, Parida V, Palmié M, Wincent J (2021) How AI capabilities enable business model innovation: scaling AI through co-evolutionary processes and feedback loops. J Bus Res 134:574–587

    Article  Google Scholar 

  103. Capecchi A, Awale M, Probst D, Reymond J-L (2019) PubChem and ChEMBL beyond Lipinski. Mol Inform 38:e1900016. https://doi.org/10.1002/minf.201900016

    Article  CAS  PubMed  Google Scholar 

  104. Wishart DS, Knox C, Guo AC et al (2006) DrugBank: a comprehensive resource for in silico drug discovery and exploration. Nucleic Acids Res 34:D668-672. https://doi.org/10.1093/nar/gkj067

    Article  CAS  PubMed  Google Scholar 

  105. Hassanzadeh P, Atyabi F, Dinarvand R (2019) The significance of artificial intelligence in drug delivery system design. Adv Drug Deliv Rev 151–152:169–190. https://doi.org/10.1016/j.addr.2019.05.001

    Article  CAS  PubMed  Google Scholar 

  106. Staples M, Daniel K, Cima MJ, Langer R (2006) Application of micro- and nano-electromechanical devices to drug delivery. Pharm Res 23:847–863. https://doi.org/10.1007/s11095-006-9906-4

    Article  CAS  PubMed  Google Scholar 

  107. Gams M, Horvat M, Ožek M et al (2014) Integrating artificial and human intelligence into tablet production process. AAPS PharmSciTech 15:1447–1453. https://doi.org/10.1208/s12249-014-0174-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Sacha GM, Varona P (2013) Artificial intelligence in nanotechnology. Nanotechnology 24:452002. https://doi.org/10.1088/0957-4484/24/45/452002

    Article  CAS  PubMed  Google Scholar 

  109. Tsigelny IF (2019) Artificial intelligence in drug combination therapy. Brief Bioinform 20:1434–1448. https://doi.org/10.1093/bib/bby004

    Article  CAS  PubMed  Google Scholar 

  110. Luo M, Feng Y, Wang T, Guan J (2018) Micro-/nanorobots at work in active drug delivery. Adv Funct Mater 28:1706100. https://doi.org/10.1002/adfm.201706100

    Article  CAS  Google Scholar 

  111. Fu J, Yan H (2012) Controlled drug release by a nanorobot. Nat Biotechnol 30:407–408. https://doi.org/10.1038/nbt.2206

    Article  CAS  PubMed  Google Scholar 

  112. Wilson B, Km G (2020) Artificial intelligence and related technologies enabled nanomedicine for advanced cancer treatment. Nanomedicine 15:433–435. https://doi.org/10.2217/nnm-2019-0366

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are grateful to Dr. Sanjit Kumar Roy for his technical support to carry out the research work.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

SM: conceptualization, methodology, visualization; SG and SG: writing—original draft preparation; PM: writing, illustrative diagrams; SC: reviewing and finalizing; KAA: writing, reviewing, editing, and supervision.

Corresponding author

Correspondence to Kazi Asraf Ali.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

The authors declare no conflict of interest.

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ali, K.A., Mohin, S., Mondal, P. et al. Influence of artificial intelligence in modern pharmaceutical formulation and drug development. Futur J Pharm Sci 10, 53 (2024). https://doi.org/10.1186/s43094-024-00625-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-024-00625-1

Keywords