View Article

Abstract

The traditional drug development pipeline is long, expensive, and heavily reliant on animal models that often fail to accurately predict human responses, leading to high attrition rates in clinical trials. In response to this challenge, the field of pharmaceutical research has seen a transformative shift toward the use of artificial organs and organoids as preclinical testing platforms. These biologically relevant in vitro models, engineered from stem cells or tissue scaffolds, mimic the structural and functional complexity of human organs, offering amore predictive and ethical alternative to animal testing. Organoids are three-dimensional, self-organizing miniaturized organs that replicate key aspects of organ physiology and pathology. Derived from pluripotent or adult stem cells, they have been successfully developed for organs such as the liver, kidney, intestine, brain, and lungs. These models allow for patient-specific disease mode ling, drug screening, and toxicity studies in a personalized manner, thus opening the door to precision medicine. Artificial organs, often bio fabricated using 3D bioprinting and tissue engineering technologies, aim to replicate entire organ systems and can be integrated with microfluidic platforms ("organs-on-chips") to simulate systemic pharmacokinetics and pharmacodynamics. These technologies significantly enhance the predictive accuracy of drug efficacy and safety, allowing researchers to assess drug metabolism, absorption, and organ-specific toxicity more effectively. Furthermore, they reduce ethical concerns associated with animal testing and align with the 3Rs principle (Replacement, Reduction, and Refinement). Regulatory bodies like the FDA and EMA have also begun acknowledging the potential of these systems in supplementing or even replacing animal data in preclinical stages. Despite their immense promise, challenges remain in standardizing organoid cultures, ensuring reproducibility, and scaling up for high-throughput screening. Integration of biosensors, artificial intelligence, and multi-organ systems (body-on-a-chip models) is expected to overcome these limitations and drive future advancements. In conclusion, artificial organs and organoids represent a paradigm shift in preclinical drug testing, offering physiologically relevant, ethically sustainable, and cost-effective models that can bridge the gap between in vitro studies and human clinical trials. As these technologies continue to mature, they hold the potential to revolutionize drug discovery, toxicity testing, and personalized medicine.

Keywords

Organoids, Artificial organs, Organ-on-chip, Body-on-chip systems, Microfluidics, Patient-derived stem cells, Personalized medicine, Drug screening, Toxicity testing, High-throughput screening, Biosensors.

Introduction

The development of new pharmaceutical compounds is a complex, costly, and time-consuming process, with high attrition rates during clinical trials often attributed to the limited predictive power of traditional preclinical models.  Historically, animal models and 2D cell cultures have served as the cornerstone of preclinical testing; however, they frequently fail to replicate the intricacies of human physiology, metabolism, and disease pathology. This discrepancy has led to an increasing demand for more human-relevant and ethically sustainable testing platforms. In response to these challenges, the field of biomedical engineering and pharmaceutical sciences has witnessed a revolutionary shift toward the use of artificial organs and organoids as alternative preclinical models. These systems offer three-dimensional, functional representations of human tissues and organs, enabling researchers to study drug absorption, distribution, metabolism, excretion (ADME), and toxicity in a more physiologically relevant context. Organoids, derived from pluripotent or adult stem cells, self-organize into miniature versions of organs such as the liver, brain, kidney, and intestine. Artificial organs, often bio fabricated using tissue engineering and 3D bioprinting technologies, aim to replicate entire organ systems and are increasingly integrated with organ-on-chip platforms to simulate systemic responses. Furthermore, these innovative models support the paradigm of personalized medicine, allowing researchers to generate patient-specific organoids for individualized drug testing and disease modelling. Coupled with advancements in biosensing, microfluidics, and machine learning, these platforms provide a powerful toolkit for enhancing the predictive accuracy of drug responses while reducing the need for animal experimentation, in line with the 3Rs principle (Replacement, Reduction, Refinement). This review explores the current landscape, applications, technological advancements, and challenges associated with artificial organs and organoids in preclinical drug testing, and also highlights the emerging role of virtual sensory technologies such as the VR Taste Simulator Lollipop in bridging the gap between biological response and patient experience.

"This Part of The Review Focuses On..."

1. Introduction to the Problem

  • Overview of traditional drug development and limitations
  • High failure rates in clinical trials due to poor predictability
  • Ethical concerns with animal testing

2. Emergence of Artificial Organs and Organoids

  • Definition and distinction between artificial organs and organoids
  • Historical background and scientific breakthroughs
  • Technologies enabling their development (e.g., stem cells, 3D bioprinting, tissue engineering)
  • Emerging Innovations: Integration of VR and Taste Simulators in Drug Testing

3. Types and Applications of Organoids

  • Liver organoids: for hepatotoxicity and metabolism studies
  • Kidney organoids: nephrotoxicity screening
  • Brain organoids: neurotoxicity and CNS drug studies
  • Intestinal organoids: drug absorption and gut microbiome interaction
  • Lung organoids: respiratory drug testing and COVID-19 research
  • Integration with VR & Sensory Simulation Platforms

4. Artificial Organs and Organs-on-Chips

  • Concept of organ-on-chip and body-on-chip systems
  • Microfluidic platforms for simulating systemic responses
  • Use in pharmacokinetics (PK) and pharmacodynamics (PD) studies
  • Integration with biosensors for real-time monitoring

5. Role in Personalized Medicine

  • Use of patient-derived stem cells for disease mode ling
  • Predicting patient-specific drug responses
  • High potential in rare disease drug discovery

6. Regulatory and Ethical Perspectives

  • FDA and EMA's stance on non-animal models
  • Alignment with 3Rs (Replacement, Reduction, Refinement)
  • Global regulatory initiatives supporting organoid research

7. Advantages Over Traditional Models

  • Human-specific physiological relevance
  • Reduction in animal use
  • Cost and time savings in early drug screening
  • More accurate toxicity and efficacy prediction

8. Challenges and Limitations

  • Standardization and reproducibility issues
  • Limited vascularization and immune components
  • Difficulty in scaling up for high-throughput testing
  • Variability between batches and protocols

9. Future Directions and Innovations

  • Integration with artificial intelligence (AI) and machine learning
  • Advancements in multi-organ systems (body-on-a-chip)
  • Improved vascularization and immune system mode ling
  • High-content screening and automation for drug pipelines
  • Integration with VR & Sensory Simulation Platforms

10. Conclusion

  • Summary of benefits and current limitations
  • Future promise in transforming drug development
  • Potential to bridge preclinical and clinical data gaps

An expanded overview of the above aspects will now be explored.

  1. Introduction to the Problem

1.1 Overview of Traditional Drug Development and Limitations

The traditional drug development pipeline is a complex, time-consuming, and resource-intensive process. On average, it takes 10–15 years and over $2.5 billion to bring a new drug to market, with the vast majority of drug candidates failing during clinical trials.  The process includes several stages: discovery, preclinical testing (typically involving cell lines and animal models), and clinical trials in humans (Phase I–III). Each stage presents its own challenges in terms of scalability, translation, and accuracy. A major bottleneck lies in the preclinical stage, where candidate drugs are tested for safety and efficacy in vitro and in vivo (primarily using animal models). These models, though widely used, often fail to mimic human physiological complexity. They lack human-specific features such as genetic diversity, human metabolism, and immune responses—making them suboptimal predictors of clinical outcomes.

1.2 High Failure Rates in Clinical Trials Due to Poor Predictability

The attrition rate in drug development is alarmingly high. Studies show that over 90% of drug candidates that pass preclinical testing fail during human trials—mainly due to inefficacy or unexpected toxicity.  This high failure rate is largely attributed to the poor translational value of animal models, which do not fully replicate human organ function, disease progression, or pharmacokinetics.

For example:

  • Cardiotoxic drugs that appear safe in animals may cause lethal arrhythmias in humans.
  • Hepatotoxicity, a leading cause of drug withdrawals post-marketing, often goes undetected in animal testing.

These failures are not only costly but also delay potentially life-saving therapies from reaching patients.

1.3 Ethical Concerns with Animal Testing

Beyond scientific limitations, traditional drug testing raises significant ethical concerns. The widespread use of animals—often involving pain, distress, or death—has sparked criticism from both the public and scientific communities. Animal rights groups and ethicists advocate for more humane and sustainable research practices. Moreover, global regulatory and ethical frameworks such as the 3Rs principle (Replacement, Reduction, and Refinement) promote alternatives that minimize animal use wherever possible. Regulatory agencies like the FDA, EMA, and OECD are increasingly encouraging the adoption of human-relevant, non-animal models for drug testing.

High-Level Perspective:

As biotechnology and systems biology evolve, so must our research paradigms. The shift toward human-centric, organ-mimicking technologies isn’t merely a scientific innovation—it’s a necessary evolution of modern medicine to be more predictive, ethical, and efficient.

2. Emergence of Artificial Organs and Organoids

2.1 Definition and Distinction Between Artificial Organs and Organoids

While both artificial organs and organoids are engineered to replicate aspects of human organ function, they differ significantly in their origin, design, and purpose:

  • Organoids are three-dimensional (3D), miniature, self-organizing tissues derived from pluripotent or adult stem cells. They mimic the microanatomy and physiological functions of real human organs. Organoids grow in vitro in a scaffold or matrix, and they can model organ development, disease states, and drug responses.
  • They are typically used for research, drug screening, and personalized medicine, not transplantation.
  • Artificial organs, on the other hand, are engineered constructs (often combining synthetic biomaterials and living cells) designed to replicate or replace the function of entire organs. These include bioartificial livers, artificial kidneys, artificial hearts, and more.
  • Some artificial organs are implantable or extracorporeal (outside the body), and many are integrated with microfluidics and sensors for systemic simulation in vitro.
  1. While organoids serve as functional in vitro models, artificial organs are aimed at both in vitro testing and clinical applications such as organ transplantation or support.

2.2 Historical Background and Scientific Breakthroughs

The development of artificial organs and organoids represents a convergence of developmental biology, regenerative medicine, and bioengineering. Key historical landmarks include:

  • 1980s–1990s: Advancements in tissue culture and stem cell biology laid the foundation for organoid development.
  • 1998: Derivation of human embryonic stem cells (h ESC) enabled in vitro studies of human development.
  • 2006: Discovery of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka revolutionized regenerative medicine, allowing patient-derived cells to be reprogrammed into any tissue type.
  • 2009–2013: The first organoids of the intestine, liver, and brain were developed, showcasing self-organizing 3D structures from stem cells.
  • 2010s onward: Rapid emergence of organ-on-chip and multi-organ platforms, enabling simulation of organ interactions and systemic drug responses.
  • Recent years: Integration of 3D bioprinting, CRISPR gene editing, and AI-based mode ling accelerated both fields, making the models more precise and functional.

2.3 Technologies Enabling Their Development

Several cutting-edge technologies have enabled the rise of these sophisticated in vitro systems:

  • Stem Cell Technology:

The use of pluripotent stem cells (ESCs and iPSCs) allows researchers to derive patient-specific organoids that reflect genetic and phenotypic features of individuals. This enables personalized drug testing and disease modeling.

  • 3D Bioprinting:

This technology allows for the layer-by-layer construction of tissues or organ-like structures using bio-inks composed of living cells and biomaterials. It is especially significant in the creation of artificial organs and vascularized tissues.

2.4 Emerging Innovations: Integration of VR and Taste Simulators in Drug Testing

The future of drug development and testing is increasingly multisensory, interactive, and digitized.  As artificial organs and organoids bring unprecedented biological relevance to in vitro models, Virtual Reality (VR) and digital sensory technologies like taste simulators are beginning to play niche but promising roles, particularly in:

i. Patient Experience Simulation in Clinical Trials

VR and taste-simulation devices are being explored to simulate real-world patient responses to oral medications, especially in terms of:

  • Palatability Testing:

In paediatric and geriatric populations, taste is a crucial factor in medication adherence. A VR taste simulator lollipop, for example, uses electrical stimulation and temperature control to mimic sweet, sour, salty, and bitter sensations without real substances.  When coupled with VR environments, it creates immersive simulations of how a patient perceives taste, which is vital for oral drug formulation.

  • Digital Twins & Behavioural Feedback: VR platforms can be used to simulate how patients interact with medicines, observe behavioural responses, and tailor drug delivery systems. This is especially useful for neurodegenerative diseases or autism, where sensory perception plays a key role in treatment response.

ii. Training and Simulation with Organoids and Artificial Organs

In preclinical research and testing, VR is used to visualize 3D models of organoids and artificial organs, helping researchers to:

  • Understand spatial cell growth, organoid morphology, and interaction with drugs.
  • Manipulate experimental parameters in a virtual lab environment.
  • Provide real-time interactive learning tools for scientists to model pharmacokinetic responses using virtual organ-on-chip simulations.

Combining these platforms with AI-driven data from artificial organs enhances the realism and prediction power of digital drug trials.

iii. Multisensory Feedback in Drug Testing Environments

As part of patient-centric drug design, especially in chronic therapies like oral chemotherapy, HIV, or anti-tubercular treatments, technologies such as:

  • Haptic feedback VR tools
  • Electrostimulation-based taste lollipops
  • Digital scent and taste cartridges...are being considered in acceptability and tolerability studies, bridging the gap between biological response (from organoid models) and patient sensory perception.

iv. Role in Clinical Trial Prototyping

Before actual trials begin, VR simulations (sometimes called "in silico clinical trials") can model:

  • Drug intake experiences (including taste and swallowing mechanisms)
  • Patient feedback loops
  • Real-time interaction with drug delivery platforms, particularly for implants or artificial organs (e.g., insulin pumps, drug-eluting devices)

Taste simulator technologies can provide non-invasive, ethical, and safe alternatives to early-phase acceptability testing—especially relevant for paediatric formulation development, where organoid models can test efficacy and VR tools test user experience.

  • Tissue Engineering: Combines cells, scaffolds, and bioactive molecules to build or regenerate functional tissues. Used heavily in both organoids and artificial organ design, particularly for building frameworks that mimic extracellular matrices.
  • Microfluidics and Organ-on-Chip Systems: These platforms simulate blood flow, mechanical stress, and inter-organ communication in vitro. They are crucial for creating physiologically dynamic models and are often integrated with artificial organ systems.
  • CRISPR and Gene Editing: Used to model genetic diseases within organoids, or to insert disease-specific mutations for studying pathogenesis and drug response.

High-Level Insight

The emergence of artificial organs and organoids is not merely a technical achievement—it reflects a paradigm shift in biomedical research.  We are moving from reductionist models (cell lines and animals) to complex, human-relevant systems that can simulate individualized biology.  These tools stand at the intersection of precision medicine, ethical innovation, and technological convergence, enabling scientists to reimagine what’s possible in disease modelling and drug development.

3. Types and Applications of Organoids in Drug Testing

Organoids, owing to their organ-like architecture and function, serve as highly predictive, human-relevant platforms in preclinical and translational drug research.  Derived from stem cells, these miniaturized 3D models allow for a better understanding of organ-specific pharmacodynamics (PD), pharmacokinetics (PK), and toxicity.  Their applications span across several organ systems, each contributing uniquely to the drug development process.

3.1 Liver Organoids: Hepatotoxicity and Metabolism Studies

The liver is a central organ for drug metabolism. Liver organoids replicate the complex hepatic environment, including cytochrome P450 enzyme activity, allowing researchers to:

  • Evaluate drug-induced liver injury (DILI)
  • Study phase I and II metabolism
  • Assess long-term chronic toxicity and bioaccumulation

Integration with microfluidics and biosensors enhances real-time monitoring of metabolic by-products, while AI modeling can predict hepatotoxicity profiles across patient genotypes.

3.2 Kidney Organoids: Nephrotoxicity Screening

Kidney organoids emulate glomerular filtration and tubular reabsorption, enabling:

  • Early screening for nephrotoxic effects (e.g., aminoglycosides, NSAIDs)
  • Functional testing of renal drug clearance
  • Understanding of drug-induced renal fibrosis and inflammation

They serve as excellent models for diseases like polycystic kidney disease and diabetic nephropathy, allowing targeted therapeutic testing in vitro.

3.3 Brain Organoids: Neurotoxicity and CNS Drug Studies

Brain organoids have emerged as powerful models to study:

  • Blood-brain barrier (BBB) penetration
  • Neurotoxicity assessment of CNS-acting drugs (e.g., antipsychotics, antidepressants)
  • Drug screening for neurodegenerative diseases (Alzheimer’s, Parkinson’s)
  • Electrophysiological responses in conditions like epilepsy

They are also useful for evaluating psychedelics, anesthetics and psychotropics—where VR-based simulations of neurological and sensory responses, such as taste perception or behavioural response in immersive environments, can be correlated with biological data from brain organoids.

3.4 Intestinal Organoids: Drug Absorption and Gut Microbiome Interaction

The intestine plays a crucial role in oral drug bioavailability. Intestinal organoids replicate the structure of crypts and villi and are used to:

  • Study first-pass metabolism
  • Evaluate drug absorption and transport
  • Examine interactions with gut microbiota
  • Test oral formulations and probiotics

Here, VR taste simulators (like the lollipop device) become relevant for oral drug formulation testing. In early development stages, these tools help simulate flavour perception, swallowing mechanics, and patient reactions. Data from such simulations can be linked with biological uptake studies in intestinal organoids, providing a complete pharmacokinetic and user-experience profile for orally administered drugs.

3.5 Lung Organoids: Respiratory Drug Testing and COVID-19 Research

Lung organoids have been crucial in:

  • Testing inhalable drug delivery systems
  • Modelling diseases such as asthma, COPD, and COVID-19
  • Evaluating viral-host interactions and antiviral drug efficacy

When combined with aerosolized VR environments, researchers can simulate user experience with inhalers or nasal drug devices, while assessing mucosal absorption and immune responses through lung organoids.

3.6 Integration with VR & Sensory Simulation Platforms

Across all organoid types, the emerging use of VR and taste simulators offers a novel way to:

  • Test patient experience (taste, texture, method of administration)
  • Collect behavioural and psychological feedback pre-clinically
  • Integrate in vitro organ responses with sensory data, creating a digital-physical loop for formulation refinement

For example, in paediatric drug development, combining intestinal organoid absorption data with VR taste tests helps optimize both efficacy and palatability, leading to improved compliance and reduced trial dropout rates.

High-Level Insight

The convergence of biological realism (organoids) and immersive feedback (VR and sensory simulation) represents a leap toward human- drug design. It transcends traditional efficacy-toxicity endpoints by integrating organ-level response, human behaviour, and real-time patient feedback, ultimately enhancing both scientific predictivity and clinical relevance. Here’s an expanded and analytical version of Section 4, integrating artificial organs, organ-on-chip platforms, and their significance in pharmacokinetics (PK), pharmacodynamics (PD), and real-time drug monitoring, with an emphasis on how this fits into the larger context of pharmaceutical innovation:

4. Artificial Organs and Organs-on-Chips

The evolution of artificial organs and organs-on-chips (OOCs) reflects a pivotal shift from static, reductionist models to dynamic, human-mimicking platforms that better simulate whole-body physiology in drug development.  These systems not only emulate organ-specific functions but can be interconnected to replicate systemic interactions, revolutionizing both preclinical testing and personalized medicine.

4.1 Concept of Organ-on-Chip and Body-on-Chip Systems

An organ-on-chip is a micro engineered, biomimetic device—typically the size of a USB stick that houses living human cells cultured in a 3D matrix, mimicking the structural, mechanical, and functional features of a specific organ (e.g., lung, heart, liver, or intestine).

  • These chips are microfluidic devices, containing tiny channels lined with human cells, allowing the continuous flow of nutrients, drugs, or even immune cells to simulate blood circulation and tissue dynamics.
  • A body-on-chip, or multi-organ system, interconnects several organ-chips (e.g., liver + gut + kidney) to simulate inter-organ communication, systemic drug exposure, and metabolite formation and clearance—essential for accurately modelling human drug responses.

Example: A liver–kidney chip can simulate the metabolic conversion of a prodrug in the liver and its renal excretion and toxicity in the kidney, offering predictive data on systemic drug behaviour.

4.2 Microfluidic Platforms for Simulating Systemic Responses

Microfluidic technology enables precise control of fluid flow and microenvironmental conditions within OOCs. These platforms replicate:

  • Shear stress from blood flow
  • Mechanical strain from muscle movement (e.g., in cardiac chips)
  • Oxygen gradients and nutrient perfusion

Such dynamic conditions allow realistic simulation of systemic drug responses, including:

  • Drug absorption and circulation
  • Cross-talk between tissues and immune cells
  • Time-dependent metabolism and clearance

These platforms are particularly useful for modelling rare diseases, cancer metastasis, and immune responses, which are difficult to recreate in traditional 2D cultures or animal models.

4.3 Use in Pharmacokinetics (PK) and Pharmacodynamics (PD) Studies

OOC systems are increasingly recognized as highly predictive tools in studying:

  • Pharmacokinetics (PK): absorption, distribution, metabolism, and excretion (ADME) in real time
  • Pharmacodynamics (PD): drug-receptor interactions, dose-response curves, and downstream effects

They enable detailed, patient-specific modelling of:

  • First-pass metabolism (gut-liver chip)
  • Tissue-specific drug delivery (e.g., in brain or lung chips)
  • Longitudinal tracking of therapeutic or toxic responses

These chips allow reduction of clinical trial failure rates, as they offer in vitro human-like responses that can better predict efficacy and toxicity compared to animal models.

4.4 Integration with Biosensors for Real-Time Monitoring

A major advancement in organ-on-chip systems is the integration of biosensors, which enable non-invasive, real-time monitoring of biological markers. These include:

  • Electrochemical sensors: detect ions, metabolites, or pH changes
  • Optical sensors: monitor oxygen levels, cell viability, or fluorescence-based assays
  • TEER sensors (Transepithelial Electrical Resistance): measure barrier integrity in intestinal or blood–brain barrier models

With biosensors, researchers can:

  • Track drug metabolism in real time
  • Measure toxic by-products or inflammatory markers
  • Adjust dosage dynamically in personalized OOC systems
  • Conduct continuous, automated monitoring over days or weeks

Future-Ready Add-on: When combined with AI algorithms, biosensor data can generate real-time models of patient-specific responses—paving the way for adaptive drug trials and digital twins.

Advanced Perspective and Connection to Review Theme

Artificial organs and OOC systems bridge the biological depth of organoids with mechanical precision and real-time feedback, making them ideal for predictive modelling, formulation testing, and even regulatory submissions.

Moreover, when coupled with sensory simulation platforms like the VR Taste Simulator Lollipop, these technologies offer comprehensive testing environments that integrate:

  • Biological response (from chips/organoids)
  • Sensory experience (from VR/taste tech)
  • Real-time monitoring and feedback (via biosensors)

Together, they form a powerful next-generation toolkit for preclinical and early clinical drug development, tailored toward personalized, efficient, and human-centred medicine.

5. Role in Personalized Medicine

The integration of patient-derived models into preclinical and translational research marks a turning point in the shift from “one-size-fits-all” pharmacotherapy toward precision medicine.  Technologies like organoids, organ-on-chip systems, and VR-based sensory simulators enable a personalized approach that factors in a patient’s genetic background, metabolic profile, and phenotypic variability, making it possible to design customized drug regimens.

5.1 Use of Patient-Derived Stem Cells for Disease Modelling

Patient-derived induced pluripotent stem cells (iPSCs) can be reprogrammed from somatic cells (e.g., skin fibroblasts, blood cells) and differentiated into multiple organ-specific lineages to generate organoids. This allows the creation of “disease-in-a-dish” models that:

  • Mimic individual-specific pathophysiology
  • Carry genetic mutations unique to the patient (e.g., cystic fibrosis, Parkinson's disease, familial cancer syndromes)
  • Enable targeted therapy testing and mechanistic disease exploration

Example: Cystic fibrosis patient-derived intestinal organoids are now used to evaluate drug efficacy for CFTR modulators, offering real-world evidence of clinical relevance before therapy initiation.

5.2 Predicting Patient-Specific Drug Responses

By generating organoids or artificial organs from a patient's cells, researchers can test multiple drugs or doses to assess:

  • Therapeutic efficacy (e.g., tumour regression in cancer organoids)
  • Toxicity thresholds (e.g., cardiotoxicity in heart-on-chip models)
  • Drug-drug interactions in polytherapy regimens

Organoid-Drug Matching: Several trials have demonstrated success in organoid-based drug screening predicting patient responses more accurately than genomic profiling alone—especially in colorectal, pancreatic, and prostate cancers. In future applications, AI-integrated organ-on-chip systems could map out patient-specific PK/PD models in silico, improving drug selection and dosing in real time.

5.3 High Potential in Rare Disease Drug Discovery

Rare diseases often lack robust models due to limited patient numbers and variability in clinical phenotypes. Patient-derived organoids fill this gap by:

  • Allowing high-content screening on rare mutations
  • Modelling disease mechanisms in ultra-rare metabolic, genetic, or neurodevelopmental disorders
  • Facilitating repurposing of FDA-approved drugs based on specific cellular responses

 Example: Brain organoids from Rett syndrome patients are used to test neuroactive compounds; similarly, liver organoids are being explored in rare inborn errors of metabolism.

Emerging Synergy with VR Sensory Simulation

While personalized medicine is biologically driven, sensory perception also plays a role—especially in treatment acceptance and adherence. The VR Taste Simulator Lollipop can be used to:

  • Simulate individual taste preferences for oral medications
  • Customize palatability profiles based on age, disease condition, or sensory sensitivity
  • Empower paediatric, geriatric, or chemo-affected patients to engage with their medication experience in a low-stress, virtual trial

When combined with biological data from personalized organoids, this approach leads to holistic personalization—accounting for both biological effectiveness and sensory user experience.

High-Level Insight

The convergence of patient-specific biological models (organoids, chips) and patient-specific sensory interfaces (VR taste simulators) represents a revolution in personalized drug development.

 It bridges the gap between genotype-to-phenotype variability and user-centric treatment design, making medicine not only more precise, but also more palatable, ethical, and engaging.

6. Regulatory and Ethical Perspectives

The rise of organoids, artificial organs, and organ-on-chip systems, alongside virtual sensory platforms like the VR Taste Simulator Lollipop, has catalysed a re-evaluation of traditional drug development frameworks. Regulatory authorities are progressively recognizing the scientific rigor and ethical superiority of these models compared to conventional animal-based testing.

6.1 FDA and EMA's Stance on Non-Animal Models

Both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have acknowledged the potential of organoid and organ-on-chip technologies to serve as complementary or alternative models to animal testing.

  • In 2022, the FDA Modernization Act 2.0 was signed into U.S. law, eliminating the legal requirement for animal testing before human clinical trials and allowing the use of “nonclinical test methods,” including organoids, OOCs, and computational models.
  • The EMA and European Commission support the integration of human-relevant models through programs like SEURAT-1 and EPAA (European Partnership for Alternatives to Animal Testing), promoting validated in vitro systems as part of safety evaluation.
  • Both agencies now support Qualification of Novel Methodologies (QONM) programs to encourage the validation and use of organ-on-chip models in regulatory submissions for pharmacokinetics and toxicology.

Implication: Pharmaceutical companies can now leverage these models earlier in the pipeline to generate regulatory-acceptable preclinical data—improving speed, accuracy, and ethical compliance.

6.2 Alignment with the 3Rs Principle (Replacement, Reduction, Refinement)

The organoid and OOC approaches are naturally aligned with the 3Rs framework, a cornerstone of biomedical ethics:

  • Replacement: These technologies directly replace the use of animals by using human-derived cells to model physiology and pathology.
  • Reduction: When organoids or artificial organs are used, fewer animal studies are needed—resulting in reduced animal use and more efficient research pipelines.
  • Refinement: These models reduce suffering by improving the predictive quality of preclinical testing, minimizing unnecessary trials or failed therapeutic interventions in humans.
  • Ethical Bonus: Virtual platforms like the VR Taste Simulator further align with 3Rs by enabling taste testing, formulation selection, and sensory feedback without the need for human ingestion or animal trials—especially valuable for paediatric and oncology drugs.

6.3 Global Regulatory Initiatives Supporting Organoid Research

Around the world, several multilateral initiatives and funding programs have been established to accelerate the development and regulatory acceptance of advanced human-relevant models:

  • OECD Guidelines: The Organisation for Economic Co-operation and Development (OECD) has issued test guidelines supporting in vitro toxicology assays, with organ-on-chip methods being evaluated for formal inclusion.
  • Japan’s PMDA (Pharmaceuticals and Medical Devices Agency): Actively funds research into micro physiological systems, supporting their integration into drug safety and efficacy assessments.
  • ICCVAM (Interagency Coordinating Committee on the Validation of Alternative Methods) in the U.S. promotes validation and harmonization of non-animal methods, including multi-organ-on-chip systems.
  • Global Harmonization Task Force (GHTF): Works toward international convergence on the use of human-relevant nonclinical models, paving the way for consistent approval processes worldwide. Interconnected Validation: As organoids and artificial organs become integrated with sensor-based and digital feedback systems, such as VR sensory platforms, regulators are beginning to consider multi-modal evidence—combining biological, behavioural, and digital markers for comprehensive evaluation.

High-Level Insight

Regulatory frameworks are catching up with innovation, opening the door to ethically robust, human-specific, and digitally integrated platforms for drug testing. As bioengineered models (organoids and chips) and human-interactive technologies (like the VR Taste Simulator) converge, regulators are building flexible pathways to validate and implement these tools in mainstream pharmaceutical research. The future will likely involve hybrid regulatory submissions—including organoid efficacy data, biosensor feedback, and even VR-based patient preference reports—as part of a more holistic and humane approach to drug development.

7. Advantages Over Traditional Models

The shift from traditional models—such as 2D cell cultures and animal testing—to human-relevant, bioengineered systems marks a paradigm shift in pharmaceutical science.  Technologies like organoids, artificial organs, organ-on-chips, and VR-based taste simulators offer transformative advantages in terms of accuracy, ethics, efficiency, and translational value.

7.1 Human-Specific Physiological Relevance

One of the most critical limitations of animal models is their poor translatability to human biology. Organoids and artificial organs are derived from human stem cells, allowing them to replicate:

  • Organ-specific architecture and microanatomy
  • Cell-cell and cell-matrix interactions
  • Human-specific gene expression and receptor profiles

Organoids simulate human pathophysiology (e.g., liver fibrosis, brain neuroinflammation), while organ-on-chips replicate tissue-tissue interfaces, enabling precise modelling of complex disease states and drug responses.

Impact: Human-relevant models improve prediction of pharmacokinetics/pharmacodynamics (PK/PD), thereby reducing late-stage clinical trial failures due to efficacy or toxicity mismatches.

7.2 Reduction in Animal Use

Bioengineered systems contribute directly to the ethical advancement of research by reducing or replacing the need for animal testing:

  • Organoids allow high-throughput in vitro screening for hundreds of compounds without animal involvement.
  • Organ-on-chip systems can simulate multi-organ interactions, previously only modelled in living animals.
  • VR sensory simulators (like the Taste Simulator Lollipop) allow formulation acceptability studies without requiring live testing in animals or humans.

Together, these models adhere to and enhance the 3Rs principle (Replacement, Reduction, Refinement), reducing ethical dilemmas and aligning with modern regulatory standards.

7.3 Cost and Time Savings in Early Drug Screening

Traditional drug development is time-intensive (10–15 years) and costly (over $2.5 billion). Organoid and chip-based models offer:

  • Faster drug candidate screening (within weeks rather than months)
  • Parallel testing of drug libraries with reduced reagent and labour costs
  • Early identification of off-target effects, minimizing downstream trial failures

For instance, liver and kidney organoids can be used early in development to eliminate hepatotoxic or nephrotoxic candidates—saving millions in wasted development. Moreover, VR-based systems simulate sensory feedback early in formulation development, reducing reformulation cycles based on patient taste rejection.

7.4 More Accurate Toxicity and Efficacy Prediction

Predicting human-specific adverse effects has long been a challenge using animals due to species differences in metabolism, immunity, and organ function. Organoids and artificial organs offer:

  • Organotypic toxicity models (e.g., liver organoids for DILI, brain organoids for neurotoxicity)
  • Personalized drug screening using patient-derived iPSCs
  • Chronic toxicity evaluation in long-term culture platforms
  • Systemic interaction studies via body-on-chip systems

Data from these systems often correlate more closely with clinical outcomes than data from preclinical animal trials—improving confidence in drug safety and therapeutic index. In addition, VR sensory platforms contribute by assessing patient acceptability and adherence, especially important in paediatric, geriatric, or oncology contexts where taste or swallowing experience can directly influence therapeutic success.

High-Level Insight

The integration of organoids, organ-on-chips, and VR taste simulation technologies represents not just an upgrade in tools, but a fundamental transformation in philosophy—from testing drugs on proxies to understanding how real human tissues respond in dynamic, interactive environments.

This approach promises to:

  • Accelerate innovation
  • Lower drug development costs
  • Improve patient outcomes
  • Humanize pharmaceutical research

8. Challenges and Limitations

Despite the transformative potential of organoids, artificial organs, organ-on-chip systems, and VR sensory platforms, several limitations continue to hinder their widespread adoption in mainstream pharmaceutical research and clinical applications.  These challenges are largely centred on technical reproducibility, scalability, biological fidelity, and regulatory standardization.

8.1 Standardization and Reproducibility Issues

One of the foremost limitations is the lack of standardized protocols for the generation, maintenance, and analysis of organoids and organ-on-chip systems. Key challenges include:

  • Protocol-to-protocol variability even within the same lab
  • Use of undefined culture components (e.g., Matrigel) that vary between batches
  • Inconsistency in cell differentiation efficiency and organoid maturity
  • Diverse data analysis methods, leading to difficulties in comparing results across platforms or institutions

This impedes validation and regulatory acceptance, particularly for clinical decision-making or drug approval workflows.

8.2 Limited Vascularization and Immune Components

While organoids mimic structural and functional features of organs, they still lack key systemic features such as:

  • Vascular networks: which are essential for realistic modelling of oxygen/nutrient diffusion, drug perfusion, and metabolite clearance
  • Immune cells and inflammatory signalling: critical for studying immunotoxicity, cancer immunotherapy, and vaccine responses

Artificial organ platforms are beginning to integrate microfluidic perfusion and immune co-cultures, but these systems are still in early stages of complexity. This limitation is particularly important when predicting host-pathogen interactions, immune-modulating therapies, or multi-organ failure scenarios.

8.3 Difficulty in Scaling Up for High-Throughput Testing

Although organoids and OOCs offer detailed insights, scaling them for high-throughput drug screening remains a technical and logistical challenge:

  • Microfabrication of chips and precise cell seeding is labour-intensive and costly
  • Automation of complex protocols is still under development
  • Many platforms lack compatibility with standard robotic screening systems used in pharma

As a result, their use is often confined to late-stage validation or mechanistic studies, rather than early-phase compound screening where cost-efficiency is critical.

8.4 Variability Between Batches and Protocols

Biological models, especially those derived from iPSCs or primary cells, often show donor-specific variability due to:

  • Genetic background differences
  • Epigenetic memory and reprogramming artifacts
  • Lab-to-lab inconsistencies in media, timing, and differentiation cues

This batch-to-batch variability affects:

  • Reproducibility of drug efficacy or toxicity results
  • Predictive value of pharmacological outcomes
  • Data reliability in regulatory submissions or personalized therapy selection

The field is actively exploring biomanufacturing standards, AI-based quality control, and multi-omics integration to address this challenge.

Additional Challenges for VR Taste Simulation Platforms

Though distinct from biological models, VR taste simulation technologies like the Taste Simulator Lollipop face their own limitations:

  • Taste simulation accuracy is still limited to a few basic taste profiles (sweet, sour, salty, bitter, umami)
  • Electrical stimulation risks (e.g., sensory fatigue, oral discomfort) must be managed for safety
  • Lack of standard user experience metrics for taste fidelity and psychological feedback
  • High initial development and deployment costs, particularly for use in clinical trials

High-Level Insight

The current limitations don’t diminish the promise of these technologies—but rather highlight the need for interdisciplinary collaboration, technological refinement, and global standardization frameworks. Overcoming these challenges will be essential to fully transition from experimental novelty to regulatory-validated standard practice in pharmaceutical research and personalized medicine.

9. Future Directions and Innovations

The future of pharmaceutical research lies at the convergence of biology, engineering, and computational science.  Organoids, artificial organs, and VR sensory technologies are rapidly evolving from experimental models into integrated, intelligent, and interactive platforms capable of reshaping drug discovery, toxicity testing, and personalized medicine.  Key innovations will address current limitations while opening new possibilities for predictive, ethical, and patient-centred research.

9.1 Integration with Artificial Intelligence (AI) and Machine Learning (ML)

The integration of AI/ML with organoid and organ-on-chip systems will enable:

  • Predictive modelling of drug responses across patient populations
  • Automated image analysis for organoid morphology, viability, and differentiation quality
  • Multi-omics data interpretation from high-throughput organoid assays (e.g., transcriptomics, metabolomics)
  • Real-time monitoring and adaptive control of organ-on-chip systems via feedback algorithms

 AI will also enhance VR sensory simulations, allowing real-time adaptation of virtual environments based on user preferences, taste sensitivity, and physiological reactions—creating customized sensory experiences for drug compliance testing. Above design to helpful to understand the future perceptive.

9.2 Advancements in Multi-Organ Systems (Body-on-a-Chip)

Next-gen organ-on-chip platforms will move toward multi-organ integration on a single microfluidic device, allowing:

  • Simulation of drug absorption (gut), metabolism (liver), distribution (blood), and excretion (kidney)
  • Inter-organ communication modelling (e.g., gut-liver-brain axis in neurodegenerative diseases)
  • Disease modelling for systemic conditions such as sepsis, autoimmune disorders, or metabolic syndromes

These body-on-chip systems will serve as patient-specific avatars in preclinical pipelines—accelerating drug development while minimizing risks.

9.3 Improved Vascularization and Immune System Modelling

Key to increasing physiological realism will be:

  • Development of vascularized organoids using 3D bioprinting and endothelial co-culture systems
  • Incorporation of immune cells (e.g., macrophages, T cells, dendritic cells) for immunotoxicity and immunotherapy research
  • Engineering of lymphatic channels and inflammatory responses to model diseases like cancer, COVID-19, or IBD more accurately

These improvements will support precision immunology and vaccine development, and help evaluate biologic drugs and immunomodulators.

9.4 High-Content Screening and Automation for Drug Pipelines

To scale up and industrialize these models, future platforms will focus on:

  • Robotic automation of organoid culture and compound delivery
  • Integration with liquid handling systems, imaging tools, and AI-based analytics
  • High-content, multiparametric screening for efficacy, cytotoxicity, metabolism, and gene expression

These systems will bridge the gap between academic innovation and pharmaceutical industry application, enabling rapid and reproducible testing across hundreds of candidates simultaneously.

9.5 Integration with VR and Sensory Simulation Platforms

A pioneering frontier lies in merging biological realism with human experience through sensory simulation platforms:

  • VR Taste Simulators, like the electrically activated lollipop, will evolve to:
    • Simulate more complex and personalized taste profiles
    • Integrate haptic and olfactory feedback for full sensory immersion
    • Support patient-centric formulation testing in virtual clinical trials
  • Digital twin models could incorporate data from organoids, chips, and VR feedback to simulate real-world patient responses before a single dose is administered.

This synergy will revolutionize drug acceptability studies, appetite stimulation therapies, pediatric compliance research, and rehabilitation for taste disorders—bringing human behavior into the heart of pharmaceutical design.

Vision for the Future

The convergence of AI, bioengineering, and immersive tech marks the dawn of a new research ecosystem that is:

  • Human-specific rather than animal-dependent
  • Predictive rather than reactive
  • Personalized rather than generic
  • Multisensory and interactive, bridging molecular response with patient behaviour

These innovations will not only reduce costs and ethical concerns but also drastically improve clinical outcomes, paving the way for a truly intelligent, immersive, and individualized future of drug development.

10. CONCLUSION

The convergence of bioengineered organ systems, such as organoids, artificial organs, and organ-on-chip platforms, alongside emerging technologies like the VR Taste Simulator Lollipop, represents a paradigm shift in the landscape of pharmaceutical research.  These models offer a biologically relevant, ethically sustainable, and technologically innovative alternative to traditional animal-based and two-dimensional systems, which often fail to predict human responses accurately.

The key benefits of these systems include:

  • Enhanced physiological fidelity, as organoids and artificial organs replicate key structural and functional aspects of human tissues;
  • Reduction in animal use, aligning with global ethical and regulatory expectations;
  • Greater efficiency in early-phase drug discovery, with reduced costs and timelines;
  • Human-specific insights into toxicity, metabolism, and efficacy, leading to better candidate selection for clinical trials;
  • The incorporation of patient-derived cells, enabling personalized medicine approaches and rare disease modelling;
  • Sensory simulation technologies like VR-based taste platforms, which bring human behavioural responses into the drug design and testing process.

Despite these advances, significant challenges remain, including:

  • A lack of standardized protocols and batch-to-batch reproducibility;
  • Incomplete representation of systemic factors like vascularization, immune components, and organ cross-talk;
  • Limited scalability for high-throughput screening;
  • Technological and safety concerns around electrical taste stimulation in VR environments.

Looking ahead, the integration of these biological models with artificial intelligence, multi-organ systems, and VR platforms holds immense promise. Such innovation will enable a holistic view of drug behaviour—from molecular interaction in engineered tissues to patient perception and adherence in virtual simulations. Most importantly, these technologies have the potential to bridge the gap between preclinical and clinical phases, increasing translational success rates and facilitating the development of safer, more effective, and patient-friendly therapeutics.  As the field matures and regulatory frameworks evolve, these platforms are expected to become essential tools in the pharmaceutical pipeline, marking a transformative era of predictive, ethical, and personalized drug development.

REFERENCES

  1. Krushna Darade*, Omkar Purnale, Vijay Tambade, Abhishek Gade, Kishor K. Dudhal, Ajit Jadhav, Amol Supekar, Review on: VR Taste Simulator Lollipop in Pharmaceuticals, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 3, 789-800. https://doi.org/10.5281/zenodo.15000467
  2. Shivkanya Jadhav*, Amol Supekar, Kanchan Jawale, Aarti Garje, Vaishnavi Jaygude, Sanika Sarode, Novel Drug Delivery System: Exploring advancements in drug delivery systems such as Nanoparticles, Lyposome and Implantable devices, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 2, 1569-1581. https://doi.org/10.5281/zenodo.14889397
  3. Maji, D., & Banerjee, R. (2022). Organ-on-Chip Technologies in India: Challenges and Opportunities. Journal of Biosciences, 47(2), 55–64.
  4. Srivastava, A., & Shukla, Y. (2021). Human Liver Organoids as a Tool for Drug Screening. Indian Journal of Pharmacology, 53(3), 193–199.
  5. Chatterjee, S., & Banerjee, S. (2023). Microfluidics and Organs-on-Chips: Emerging Tools in Indian Biomedical Research. Current Science, 124(8), 954–962.
  6. Deshmukh, S., & Patil, A. (2020). Virtual Reality for Sensory and Behavioral Simulations in Indian Clinical Practice. Journal of Medical Engineering & Technology India, 44(2), 88–96.
  7. Verma, R., et al. (2022). 3D Bioprinting and Artificial Organs: Future of Regenerative Medicine in India. Indian Journal of Medical Research, 155(5), 560–568.
  8. Joshi, R., & Nair, P. (2023). A Study on the Feasibility of VR-based Taste Simulators in Pediatric Drug Design. Journal of Clinical Research in Pediatrics, 15(1), 22–30.
  9. Mandal, P., & Roy, S. (2021). Induced Pluripotent Stem Cells and Organoids: A New Frontier in Indian Personalized Medicine. Journal of Cell Science and Therapy, 12(4), 134–142.

Gupta, A., & Sharma, V. (2022). Application of Organoid Models in Indian Rare Disease Research. NIPER Journal of Pharmaceutical Sciences, 4(2), 85–91.

Reference

  1. Krushna Darade*, Omkar Purnale, Vijay Tambade, Abhishek Gade, Kishor K. Dudhal, Ajit Jadhav, Amol Supekar, Review on: VR Taste Simulator Lollipop in Pharmaceuticals, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 3, 789-800. https://doi.org/10.5281/zenodo.15000467
  2. Shivkanya Jadhav*, Amol Supekar, Kanchan Jawale, Aarti Garje, Vaishnavi Jaygude, Sanika Sarode, Novel Drug Delivery System: Exploring advancements in drug delivery systems such as Nanoparticles, Lyposome and Implantable devices, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 2, 1569-1581. https://doi.org/10.5281/zenodo.14889397
  3. Maji, D., & Banerjee, R. (2022). Organ-on-Chip Technologies in India: Challenges and Opportunities. Journal of Biosciences, 47(2), 55–64.
  4. Srivastava, A., & Shukla, Y. (2021). Human Liver Organoids as a Tool for Drug Screening. Indian Journal of Pharmacology, 53(3), 193–199.
  5. Chatterjee, S., & Banerjee, S. (2023). Microfluidics and Organs-on-Chips: Emerging Tools in Indian Biomedical Research. Current Science, 124(8), 954–962.
  6. Deshmukh, S., & Patil, A. (2020). Virtual Reality for Sensory and Behavioral Simulations in Indian Clinical Practice. Journal of Medical Engineering & Technology India, 44(2), 88–96.
  7. Verma, R., et al. (2022). 3D Bioprinting and Artificial Organs: Future of Regenerative Medicine in India. Indian Journal of Medical Research, 155(5), 560–568.
  8. Joshi, R., & Nair, P. (2023). A Study on the Feasibility of VR-based Taste Simulators in Pediatric Drug Design. Journal of Clinical Research in Pediatrics, 15(1), 22–30.
  9. Mandal, P., & Roy, S. (2021). Induced Pluripotent Stem Cells and Organoids: A New Frontier in Indian Personalized Medicine. Journal of Cell Science and Therapy, 12(4), 134–142.
  10. Gupta, A., & Sharma, V. (2022). Application of Organoid Models in Indian Rare Disease Research. NIPER Journal of Pharmaceutical Sciences, 4(2), 85–91.

Photo
Darade Krushna
Corresponding author

Department Of Pharmaceutical Science in Savitribai Phule University Pune.

Photo
Garje Manoj
Co-author

Department Of Pharmaceutical Science in Savitribai Phule University Pune.

Photo
Garje Aarti
Co-author

Department Of Pharmaceutical Science in Savitribai Phule University Pune.

Photo
Darade Shraddha
Co-author

Department Of Pharmaceutical Science in Savitribai Phule University Pune.

Darade Krushna*, Garje Manoj Sunil, Garje Aarti Sunil, Darade Shraddha Somnath, Artificial Organs and Organoids in Preclinical Drug Testing: Bridging Biology and Innovation, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 4, 2790-2809 https://doi.org/10.5281/zenodo.15269130

More related articles
Cancer Research: Progress, Challenges, and Future...
Kashinath Sakhare, Shubhangi Dhoble , Pandit Biradar, Kapil Pat...
Emerging of Artificial Intelligence and Technology...
Manimaran k, Naveen kumar J, R. V. Siva prakash, ...
Nanobots in Medicine ...
Iranna B. Gulagonda, Dhiraj D. Mane, Sanjay L. Dhavale, Vishal K....
Related Articles
Personalized Medicine Approaches in the Treatment Of Mental Health Disorder...
Mr. Swapnil D. Aade, Prof. Zameer M. Shah, Dr. Shivshankar D. Mhaske, Prof. Tejas J. Sharma, ...
Organoids As Promising Tools In Therapy...
Dr. Pittu Vishnu Priya, Keshineni Shravani, Pabbathi Sai Divya, AVSSS Gupta, ...
Advancements And Challenges In The Integration Of Artificial Intelligence With H...
Patil Jayesh Hilal, Patil Harish , Chordiya Harshita , Panpatil Ashishkumar, Deore Rashmi , Sarode S...
Review on Regulatory Affairs in the Era of Artificial Intelligence: Roles, Needs...
Sushant Shinde , Prachi Telavane, Shubham Prabhu, Mohit Jadhav , ...
Cancer Research: Progress, Challenges, and Future Directions ...
Kashinath Sakhare, Shubhangi Dhoble , Pandit Biradar, Kapil Patne , Swati Narwate , Tukaram Pilgur...
More related articles
Cancer Research: Progress, Challenges, and Future Directions ...
Kashinath Sakhare, Shubhangi Dhoble , Pandit Biradar, Kapil Patne , Swati Narwate , Tukaram Pilgur...
Nanobots in Medicine ...
Iranna B. Gulagonda, Dhiraj D. Mane, Sanjay L. Dhavale, Vishal K. Mote, Rahul A. Kedar, ...
Cancer Research: Progress, Challenges, and Future Directions ...
Kashinath Sakhare, Shubhangi Dhoble , Pandit Biradar, Kapil Patne , Swati Narwate , Tukaram Pilgur...
Nanobots in Medicine ...
Iranna B. Gulagonda, Dhiraj D. Mane, Sanjay L. Dhavale, Vishal K. Mote, Rahul A. Kedar, ...