Appearance
This report is written by MaltSci based on the latest literature and research findings
What is the role of organoids in disease modeling?
Abstract
The advent of organoid technology has revolutionized biomedical research, particularly in disease modeling. Organoids are three-dimensional (3D) structures derived from stem cells that closely mimic the architecture and functionality of actual organs. This unique capability allows researchers to investigate complex biological processes and disease mechanisms in a more physiologically relevant context compared to traditional two-dimensional (2D) cell cultures. Organoids have emerged as invaluable tools in studying various diseases, including cancer, neurodegenerative disorders, and infectious diseases. They provide a platform for personalized medicine, where patient-derived models can tailor treatments based on individual genetic backgrounds. Additionally, organoids contribute to drug discovery and toxicity testing, offering insights critical for developing novel therapeutic strategies. Current research has demonstrated their utility in cancer research, where tumor-derived organoids facilitate the study of tumor biology and drug sensitivity. In neurodegenerative diseases, organoids enable the investigation of underlying mechanisms and potential therapies. Despite the promising advancements, challenges such as variability in organoid production and the need for improved physiological relevance remain. The integration of organoids with cutting-edge technologies, including microfluidics and artificial intelligence, is essential for enhancing their functionality and reproducibility. This review explores the concept of organoids, their diverse applications in disease modeling, advantages and limitations, recent advances in research, and future directions for their use in personalized medicine.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 The Concept of Organoids
- 2.1 Definition and Characteristics of Organoids
- 2.2 Comparison with Traditional Cell Culture Models
- 3 Applications of Organoids in Disease Modeling
- 3.1 Cancer Research
- 3.2 Neurodegenerative Diseases
- 3.3 Infectious Diseases
- 4 Advantages and Limitations of Organoid Technology
- 4.1 Advantages: Physiological Relevance and Versatility
- 4.2 Limitations: Variability and Ethical Concerns
- 5 Recent Advances in Organoid Research
- 5.1 Innovations in Organoid Culturing Techniques
- 5.2 Integration with Genomics and Drug Screening
- 6 Future Directions and Perspectives
- 6.1 Potential for Personalized Medicine
- 6.2 Standardization and Reproducibility in Organoid Research
- 7 Conclusion
1 Introduction
The advent of organoid technology has marked a significant turning point in biomedical research, particularly in the realm of disease modeling. Organoids, which are three-dimensional (3D) structures derived from stem cells, possess the remarkable ability to closely replicate the architecture and functionality of actual organs. This unique feature positions them as a transformative tool for studying complex biological processes and disease mechanisms, surpassing traditional two-dimensional (2D) cell cultures that often fail to provide a physiologically relevant environment. By enabling researchers to observe cellular interactions, tissue development, and responses to therapeutic agents in a controlled setting, organoids have emerged as invaluable assets in advancing our understanding of various diseases, including cancer, neurodegenerative disorders, and infectious diseases [1][2].
The significance of organoids in disease modeling cannot be overstated. As they bridge the gap between basic research and clinical applications, organoids offer a platform for personalized medicine, where patient-derived models can be utilized to tailor treatments based on individual genetic backgrounds and disease characteristics [3][4]. Furthermore, organoids have the potential to reduce reliance on animal models, thus addressing ethical concerns associated with traditional research methods [5]. Their applications extend beyond mere modeling; they are instrumental in drug discovery, toxicity testing, and regenerative medicine, providing insights that are critical for the development of novel therapeutic strategies [6][7].
Current research in organoid technology has demonstrated a wide range of applications across various fields. In cancer research, organoids derived from tumors have been used to study tumor biology, drug sensitivity, and the tumor microenvironment, thereby facilitating the development of targeted therapies [4][8]. Similarly, in the context of neurodegenerative diseases, neural organoids have provided a novel approach to investigate the underlying mechanisms of disorders such as Alzheimer’s and Parkinson’s disease [9][10]. Additionally, organoids have proven to be effective in modeling infectious diseases, offering insights into host-pathogen interactions and aiding in the development of antiviral therapies [3][11].
Despite the promising advancements in organoid technology, several challenges remain. Issues such as variability in organoid production, difficulties in recapitulating complex tissue architectures, and ethical considerations surrounding their use in research necessitate ongoing investigation and refinement [1][5]. Moreover, as organoid models become increasingly integrated with cutting-edge technologies such as microfluidics and artificial intelligence, there is a pressing need for standardized protocols to ensure reproducibility and reliability across studies [12][13].
This review will be organized into several key sections to comprehensively explore the role of organoids in disease modeling. We will begin with a detailed examination of the concept of organoids, including their definition, characteristics, and a comparison with traditional cell culture models. Following this, we will delve into the diverse applications of organoids in disease modeling, focusing specifically on cancer research, neurodegenerative diseases, and infectious diseases. We will also discuss the advantages and limitations of organoid technology, highlighting the physiological relevance and versatility of organoids, as well as the challenges associated with their use. Furthermore, we will present recent advances in organoid research, particularly innovations in culturing techniques and their integration with genomics and drug screening. Finally, we will outline future directions and perspectives for organoid technology, emphasizing its potential in personalized medicine and the importance of standardization in organoid research.
In conclusion, organoids represent a groundbreaking advancement in the field of biomedical research, offering unparalleled opportunities for understanding disease mechanisms and developing innovative therapeutic strategies. As research continues to evolve, the integration of organoid technology into mainstream clinical practice holds the promise of revolutionizing patient care and enhancing our ability to combat a wide array of diseases.
2 The Concept of Organoids
2.1 Definition and Characteristics of Organoids
Organoids are three-dimensional (3D) culture systems that closely mimic the architecture and functionality of real organs, making them invaluable tools in disease modeling. These structures are derived from various sources, including pluripotent stem cells and adult stem cells, and they replicate the cellular diversity, organization, and microenvironment of the tissues from which they are derived. The ability of organoids to recapitulate the physiological and pathological characteristics of human organs allows researchers to explore disease mechanisms and therapeutic responses in a more biologically relevant context than traditional two-dimensional (2D) cell cultures or animal models.
The role of organoids in disease modeling encompasses several key aspects. Firstly, organoids serve as platforms for investigating the pathophysiology of various diseases, including genetic disorders, infectious diseases, and cancers. For instance, organoid models have significantly advanced our understanding of genetic diseases by enabling the identification of novel pathogenic genes and elucidating disease mechanisms through the study of organoid responses to genetic alterations[2]. Similarly, in the context of infectious diseases, organoids have been utilized to simulate host-pathogen interactions, providing insights into viral pathogenesis and aiding in the development of antiviral strategies[14].
Secondly, organoids facilitate drug discovery and therapeutic testing. Patient-derived organoids (PDOs) are particularly useful in this regard, as they retain the genetic and phenotypic characteristics of the donor's tumor or tissue. This allows for the evaluation of drug efficacy and toxicity in a context that closely resembles the patient's actual disease state[4]. Furthermore, organoids can be employed to screen for novel therapeutic agents, assess drug combinations, and study drug resistance mechanisms, thereby accelerating the translation of preclinical findings to clinical applications[1].
Additionally, organoids contribute to personalized medicine by allowing for the development of tailored therapeutic strategies based on individual patient profiles. By integrating multi-omics approaches and bioengineering techniques, researchers can create organoid models that reflect the unique genetic and molecular landscape of a patient's disease, enabling the identification of the most effective treatment options[6].
Despite their advantages, organoids also face challenges, including limitations in vascularization, immune integration, and the complexity of recapitulating certain tissue architectures. Ongoing research aims to address these challenges through interdisciplinary innovations, such as the integration of microfluidics and artificial intelligence, which can enhance organoid functionality and scalability[1].
In summary, organoids represent a powerful tool in disease modeling, bridging the gap between traditional research methods and the complexities of human disease. Their ability to mimic the in vivo environment makes them crucial for advancing our understanding of disease mechanisms, enhancing drug discovery, and paving the way for personalized medicine approaches. As the field of organoid technology continues to evolve, it holds great promise for reshaping biomedical research and improving clinical outcomes.
2.2 Comparison with Traditional Cell Culture Models
Organoids represent a significant advancement in the field of biomedical research, particularly in disease modeling. They are three-dimensional (3D) cellular structures that mimic the architecture and functionality of actual organs, derived from stem cells, including embryonic stem cells, adult stem cells, or induced pluripotent stem cells. This innovative technology bridges the gap between traditional two-dimensional (2D) cell cultures and in vivo models, allowing for a more physiologically relevant representation of human diseases.
The role of organoids in disease modeling is multifaceted. Firstly, they provide a platform to study the mechanisms underlying various diseases, including genetic disorders, infectious diseases, and cancer. Organoids can be derived from patient tissues, which allows for personalized medicine approaches where researchers can investigate disease phenotypes and therapeutic responses specific to individual patients. For instance, liver organoids have been utilized to model liver diseases such as hereditary liver disorders, viral hepatitis, and nonalcoholic fatty liver disease, enabling a deeper understanding of disease mechanisms and potential treatments (Liu et al. 2023) [15].
Moreover, organoids are particularly valuable in infectious disease research. They have been employed to study host-pathogen interactions, providing insights into how viruses and bacteria infect human tissues. For example, lung organoids have been instrumental in investigating the pathogenesis of respiratory infections, including those caused by SARS-CoV-2, allowing researchers to explore viral entry mechanisms and test antiviral drugs (Li et al. 2025) [16]. This capability is crucial, especially given the complexities involved in traditional animal models that may not accurately reflect human disease conditions.
In comparison to traditional 2D cell culture models, organoids offer several advantages. Traditional cultures often lack the structural complexity and cellular heterogeneity of actual tissues, which can lead to misleading results regarding drug responses and disease mechanisms. In contrast, organoids maintain the spatial organization and functional properties of the original tissues, thereby preserving parental gene expression and mutation characteristics over extended culture periods (Yang et al. 2023) [17]. This fidelity enhances the translational validity of research findings, making organoids a more reliable model for drug discovery and therapeutic development.
Furthermore, organoids facilitate high-throughput screening and precision medicine applications, as they can be generated from various tissue types and customized to reflect specific genetic backgrounds. This adaptability allows for extensive research into the effects of genetic variations on disease progression and treatment efficacy (Azar et al. 2021) [18].
Despite their numerous advantages, organoids are not without limitations. Challenges such as the difficulty in replicating complex tissue architectures, the need for improved vascularization, and the variability in organoid generation efficiency remain significant hurdles (Rauth et al. 2021) [19]. Nonetheless, ongoing advancements in bioengineering and multi-omics integration continue to enhance the utility of organoids in disease modeling, offering promising avenues for future research and clinical applications.
In summary, organoids serve as a transformative tool in disease modeling, providing a more accurate and relevant platform for understanding disease mechanisms, testing therapeutics, and advancing personalized medicine. Their development marks a significant shift in how researchers can study human diseases, ultimately leading to improved outcomes in clinical settings.
3 Applications of Organoids in Disease Modeling
3.1 Cancer Research
Organoids, which are three-dimensional (3D) cultures derived from pluripotent or adult stem cells, have emerged as revolutionary tools in disease modeling, particularly in cancer research. They meticulously mimic human organ architecture and function, thereby offering a physiologically relevant model to address critical questions in oncology. Patient-derived organoids (PDOs) closely resemble the biology of tissues and tumors, enabling ex vivo modeling of human diseases and dissecting key features of tumor biology, such as anatomical diversity and inter- and intra-tumoral heterogeneity [20].
In the context of cancer research, organoids serve several significant roles. Firstly, they facilitate the development of personalized treatment plans by allowing drug sensitivity tests on organoids derived from tumor tissues of patients. For example, in colorectal cancer, organoids developed from patient tumor tissues have been utilized to inform personalized chemotherapy regimens, demonstrating their potential in guiding clinical decision-making [21]. Furthermore, organoids are increasingly recognized for their utility in drug discovery and testing, as they can replicate the complex interactions within the tumor microenvironment, thereby enhancing the relevance of findings to clinical practice [22].
Additionally, organoids provide insights into tumor heterogeneity and spatial organization, which are critical for understanding cancer progression and therapeutic resistance. By interrogating non-tumor stromal cells within the organoid cultures, researchers can gain a deeper understanding of the tumor microenvironment and its impact on cancer biology [22]. This capability is essential for the development of targeted therapies and for identifying novel therapeutic targets [23].
Moreover, organoids have been applied in the study of various cancer types, including ovarian, endometrial, and colorectal cancers. Their ability to reflect the primary tissue's biology and pathology allows for effective modeling of carcinogenesis and drug screening [4]. For instance, they have been instrumental in understanding high-grade serous ovarian cancer and its cells-of-origin, thereby aiding in the development of precise treatment options [4].
The integration of organoid technology with advanced techniques such as microfluidics, genetic editing, and artificial intelligence has further enhanced their applications in disease modeling. These advancements allow for better control over the microenvironment and functional maturation of organoids, thus facilitating more accurate modeling of disease [1]. Furthermore, organoids can also be co-cultured with immune cells, enabling the exploration of tumor-immune interactions and the development of immune-based therapies [24].
Despite their numerous advantages, challenges remain in the standardization of organoid culture methods and in achieving reliable representations of tumor heterogeneity [25]. Nonetheless, the promise of organoids in advancing our understanding of cancer biology and improving therapeutic strategies positions them as a transformative tool in precision medicine and cancer research [26].
In summary, organoids play a crucial role in disease modeling by accurately recapitulating the complexity of human tumors, providing a platform for personalized medicine, facilitating drug discovery, and enabling the study of tumor microenvironments and heterogeneity. Their applications in cancer research continue to evolve, highlighting their significance in bridging the gap between basic research and clinical practice.
3.2 Neurodegenerative Diseases
Organoids have emerged as a revolutionary tool in the modeling of neurodegenerative diseases, providing a platform that closely mimics the human brain's structure and function. These three-dimensional (3D) self-organizing structures derived from pluripotent stem cells enable researchers to investigate the complex pathophysiological mechanisms underlying various neurological disorders.
One of the primary advantages of organoids is their ability to replicate key features of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and Huntington's disease. For instance, brain organoids can exhibit amyloid plaque and neurofibrillary tangle-like structures characteristic of AD, thus facilitating the study of disease progression and the evaluation of potential therapeutic agents [27]. Moreover, organoids derived from human-induced pluripotent stem cells (hiPSCs) allow for patient-specific modeling, which can provide insights into the genetic and environmental factors contributing to neurodegeneration [28].
Organoids have also been utilized to explore the cellular interactions and microenvironmental factors that play critical roles in neurodegeneration. They can model the interactions between neurons and glial cells, which are essential for understanding the neuroinflammatory processes involved in diseases like AD [29]. Additionally, organoids can be engineered to include vascular structures and other non-neuronal cell types, thereby enhancing their physiological relevance and enabling more comprehensive studies of neurodevelopmental and neurodegenerative disorders [30].
The ability of organoids to recapitulate developmental processes and disease phenotypes makes them invaluable for drug discovery and testing. By providing a more accurate representation of human disease states, organoids facilitate the identification of novel therapeutic targets and the assessment of drug efficacy in a controlled environment [31]. This is particularly significant given the limitations of traditional 2D cultures and animal models, which often fail to accurately mimic human disease [32].
In summary, organoids serve as a powerful platform for modeling neurodegenerative diseases, allowing researchers to dissect the underlying mechanisms of these complex disorders, explore patient-specific variations, and develop targeted therapeutic strategies. Their capacity to mimic human brain development and pathology positions them at the forefront of neurological research and personalized medicine.
3.3 Infectious Diseases
Organoids, as three-dimensional (3D) multicellular structures derived from stem cells, have emerged as pivotal tools in the modeling of infectious diseases. Their ability to closely mimic the architecture and function of human organs enables researchers to study pathogen-host interactions in a more physiologically relevant context compared to traditional two-dimensional (2D) cell cultures or animal models. This advanced modeling approach provides significant insights into the mechanisms of infectious diseases, enhancing our understanding of their pathogenesis and potential therapeutic strategies.
One of the primary advantages of organoids is their capacity to replicate the complex tissue microenvironment. For instance, organoids derived from various human tissues, such as the lungs, intestines, and brain, have been instrumental in studying infections caused by a variety of pathogens, including viruses, bacteria, and protozoa. Research has shown that organoids can accurately simulate host responses to infections, allowing for the investigation of pathogen-induced lesions and immune responses in a controlled setting [3][11][33].
The COVID-19 pandemic significantly accelerated the adoption of organoid technology in infectious disease research. Organoids have been utilized to model the interactions between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human cells, leading to a deeper understanding of viral pathogenesis and host immune responses [14][34]. These models have proven crucial in drug screening and vaccine development, offering a platform for testing therapeutic agents in a setting that closely resembles human physiology [3][35].
Moreover, organoids facilitate the study of complex diseases by allowing for the exploration of co-infections and the effects of the microbiome on pathogen behavior. This capability is particularly important in understanding how different pathogens may interact within the same host environment, which is a critical aspect of infectious disease dynamics [13][36].
Despite their many advantages, organoid models also face challenges, including limitations in modeling chronic infections and the need for improved integration of immune components to fully emulate the human immune response [3][37]. Nevertheless, ongoing advancements in organoid technology, such as the development of vascularized organoids and integration with immune cells, hold promise for overcoming these limitations and enhancing the utility of organoids in infectious disease research [14][35].
In summary, organoids serve as a transformative platform in the modeling of infectious diseases, bridging the gap between in vitro studies and clinical applications. Their ability to replicate human organ systems allows for detailed investigation into pathogen-host interactions, disease mechanisms, and therapeutic responses, making them an invaluable resource in the fight against infectious diseases. As research progresses, organoids are expected to play an increasingly central role in the development of effective treatments and preventive measures against emerging infectious threats.
4 Advantages and Limitations of Organoid Technology
4.1 Advantages: Physiological Relevance and Versatility
Organoids have emerged as a transformative technology in biomedical research, particularly in the realm of disease modeling. They are three-dimensional (3D) in vitro structures that mimic the architecture and functionality of actual human organs, providing a more physiologically relevant environment compared to traditional two-dimensional cell cultures and animal models. This advancement allows for a more accurate representation of human diseases and the biological processes underlying them.
The physiological relevance of organoids is one of their most significant advantages. They preserve the molecular, structural, and functional characteristics of the tissues from which they are derived, thereby facilitating studies that are more biologically relevant. For instance, organoids can be derived from various cell types, including human primary progenitor cells, pluripotent stem cells, or tumor-derived cells, and can be co-cultured with immune or microbial cells to better replicate the tissue niche. This complexity allows researchers to study the interactions between different cell types and the microenvironment, which is crucial for understanding disease mechanisms and testing therapeutic strategies [38].
Moreover, organoids provide a versatile platform for modeling a wide range of diseases, including genetic disorders, cancers, infectious diseases, and chronic conditions. They can be used for drug screening, gene editing therapies, and even organ transplantation strategies. For example, lung organoids have been utilized to model diseases such as cystic fibrosis and chronic obstructive pulmonary disease, allowing for the evaluation of therapeutic interventions in a context that closely resembles human physiology [38][39]. Similarly, brain organoids have been employed to model neurodegenerative diseases, offering insights into the pathophysiology of conditions like Alzheimer's and Parkinson's disease [40].
Despite these advantages, organoid technology does have limitations. One significant challenge is the difficulty in recapitulating the full complexity of in vivo pathologies. While organoids can mimic many aspects of human tissues, they may not fully represent the intricate cellular interactions and environmental factors present in a living organism. Furthermore, scaling production and achieving consistent quality across different batches of organoids remain hurdles that need to be addressed for broader clinical application [2][41].
Another limitation is the current inability to fully integrate the immune system within organoid models, which can affect the study of diseases that are heavily influenced by immune responses. As organoid technology continues to evolve, addressing these challenges will be crucial for maximizing their utility in personalized medicine and therapeutic development [40][42].
In summary, organoids represent a significant advancement in disease modeling due to their physiological relevance and versatility. They offer a promising platform for understanding disease mechanisms and testing new therapies, while also presenting challenges that researchers must navigate to fully harness their potential in biomedical research.
4.2 Limitations: Variability and Ethical Concerns
Organoids play a crucial role in disease modeling, serving as innovative three-dimensional (3D) in vitro systems that replicate the architecture and function of human organs. This technology has emerged as a significant advancement in biomedical research, providing a more physiologically relevant platform compared to traditional two-dimensional cell cultures and animal models. The advantages of organoids in disease modeling are manifold, including their ability to closely mimic native tissue environments, their genetic fidelity, and their potential for personalized medicine.
One of the primary advantages of organoids is their capacity to recapitulate the complex tissue architecture and cellular interactions of the organs from which they are derived. This feature allows researchers to study disease mechanisms in a more biologically relevant context, enhancing the translational validity of findings. For instance, organoids derived from patient tissues can provide insights into individual disease pathology and therapeutic responses, thereby advancing personalized medicine approaches[2][42].
Moreover, organoids can be utilized for drug screening, offering a platform for evaluating therapeutic efficacy and safety in a system that closely resembles human physiology. This capability is particularly valuable in cancer research, infectious diseases, and genetic disorders, where organoids can model disease progression and therapeutic responses more accurately than traditional models[41][43].
However, despite these advantages, organoid technology is not without its limitations. Variability in organoid formation and maintenance can lead to inconsistencies in experimental outcomes. Factors such as the source of cells, culture conditions, and the complexity of the microenvironment can influence the behavior and characteristics of organoids, resulting in variability that complicates data interpretation and reproducibility[5][44].
Ethical concerns also arise in the context of organoid research, particularly regarding the sourcing of human tissues and the implications of using stem cells. The derivation of organoids from pluripotent stem cells or patient biopsies necessitates careful ethical considerations, including informed consent and the potential for unforeseen consequences in regenerative medicine applications[42][45].
In summary, while organoids represent a promising frontier in disease modeling, offering significant advantages in terms of physiological relevance and personalized medicine, challenges related to variability and ethical considerations must be addressed to fully harness their potential in biomedical research.
5 Recent Advances in Organoid Research
5.1 Innovations in Organoid Culturing Techniques
Organoids serve as pivotal tools in disease modeling, offering a more physiologically relevant alternative to traditional two-dimensional (2D) cell cultures and animal models. These three-dimensional (3D) structures are derived from stem cells and can recapitulate the architecture, cellular diversity, and functionality of actual organs. This capacity enables organoids to bridge the gap between in vitro and in vivo studies, facilitating more accurate disease modeling and therapeutic testing.
Recent advancements in organoid technology have significantly enhanced their utility in various biomedical applications. For instance, organoids have been integrated with innovative techniques such as microfluidics, genetic editing, bioprinting, and artificial intelligence. These integrations improve microenvironmental control, functional maturation, and scalability of organoid cultures, thereby enhancing their applicability in disease modeling and drug development [1].
In the context of genetic disorders, organoids have proven instrumental in identifying novel pathogenic genes and elucidating disease mechanisms. They enable researchers to conduct drug screening and gene-editing therapies, thus accelerating the translation of basic research into clinical applications [2]. Furthermore, organoids derived from specific patient tissues exhibit drug responses that closely mimic those observed in the patients themselves, thereby providing valuable insights into personalized medicine [46].
In infectious disease research, organoids have emerged as powerful models for studying host-pathogen interactions. They facilitate the investigation of viral and bacterial infections by replicating the tissue architecture and function of the organs affected by these pathogens. This capability allows for the exploration of complex disease mechanisms and the evaluation of therapeutic strategies [3]. The application of organoids in infectious disease modeling has gained particular prominence in light of recent global health challenges, such as the COVID-19 pandemic, where organoid models have been utilized to study the SARS-CoV-2 virus [14].
Innovations in culturing techniques have further refined the capabilities of organoids. The development of organoid-on-a-chip systems exemplifies this progress, allowing for the simulation of organ-level interactions in a controlled microenvironment. These platforms enhance the fidelity of disease modeling by incorporating mechanical and biochemical cues that mimic in vivo conditions [12]. Moreover, advancements in bioengineering have led to the creation of multi-organ systems that can study systemic diseases and drug interactions across different tissues, thereby providing a holistic view of disease pathology [13].
Despite these advancements, challenges remain in standardizing organoid production and improving their vascularization and immune integration, which are crucial for accurately mimicking the in vivo environment [1]. Future strategies aimed at interdisciplinary innovation will be essential to harness the full potential of organoids in reshaping biomedicine and enhancing their role in disease modeling [2].
In summary, organoids are revolutionizing disease modeling through their ability to accurately reflect human organ physiology and pathology. Innovations in culturing techniques and interdisciplinary approaches are paving the way for their expanded application in precision medicine, drug discovery, and regenerative therapies. As the field continues to evolve, organoids are expected to play an increasingly central role in biomedical research and clinical applications.
5.2 Integration with Genomics and Drug Screening
Organoids have emerged as pivotal tools in disease modeling, particularly due to their ability to closely replicate the structural and functional characteristics of human tissues. These three-dimensional (3D) cellular models, derived from stem cells, provide a more physiologically relevant platform compared to traditional two-dimensional (2D) cell cultures and animal models. Their inherent genetic fidelity allows for biologically relevant research, especially in the context of genetic disorders, where organoids can be used to identify novel pathogenic genes and elucidate disease mechanisms [2].
The role of organoids in disease modeling extends to various applications, including drug discovery and personalized medicine. They facilitate the evaluation of drug efficacy and toxicity by mimicking the complex interactions within human tissues. For instance, patient-derived organoids have shown promise in predicting individual drug responses, thereby enhancing the precision of therapeutic strategies [47]. This capability is particularly significant in oncology, where organoids can correlate genetic mutations with sensitivity to targeted therapies, enabling tailored treatment plans [2].
Recent advancements in organoid technology have further integrated multi-omics approaches, which encompass genomics, transcriptomics, proteomics, and metabolomics. This integration allows for a comprehensive understanding of disease mechanisms at multiple biological levels, thus enhancing the predictive power of organoid models in drug screening [48]. The incorporation of technologies such as CRISPR-Cas9 for gene editing enables precise modeling of genetic disorders, facilitating the exploration of specific disease pathways and therapeutic targets [48].
Moreover, organoids are being utilized in innovative drug delivery systems and high-throughput screening platforms, which can validate the targeting efficiency and therapeutic efficacy of new compounds [1]. The use of organoid-on-chip technologies has also been explored, enabling real-time monitoring of drug interactions and effects in a controlled microenvironment [12].
Despite the advancements, challenges remain in the field of organoid research. Issues such as the scalability of organoid production, the reproducibility of results, and the complexity of fully recapitulating organ functions need to be addressed [48]. Nonetheless, organoids continue to hold great potential for revolutionizing disease modeling, drug discovery, and the development of personalized therapeutic approaches, thereby significantly impacting biomedical research and clinical applications [49].
6 Future Directions and Perspectives
6.1 Potential for Personalized Medicine
Organoids have emerged as a pivotal tool in disease modeling, providing innovative approaches that bridge the gap between traditional in vitro and in vivo systems. These three-dimensional (3D) cellular structures, derived from stem cells, mimic the architecture and functionality of human organs, thus offering a more physiologically relevant context for studying various diseases.
The role of organoids in disease modeling is multifaceted. They facilitate the exploration of complex disease mechanisms, enabling researchers to investigate genetic disorders, cancer, infectious diseases, and other pathologies with greater accuracy. For instance, organoids derived from patient-specific cells can replicate the genetic and phenotypic characteristics of individual diseases, allowing for detailed studies of disease progression and therapeutic responses [2]. This capability is particularly beneficial in understanding genetic disorders, where organoids can help identify novel pathogenic genes and elucidate underlying disease mechanisms [2].
Moreover, organoids serve as powerful platforms for drug screening and therapeutic innovation. They enable the assessment of drug efficacy and toxicity in a setting that closely resembles human physiology, thus improving the predictive power of preclinical studies. Recent advancements in organoid technology, including integration with microfluidics and artificial intelligence, have further enhanced their application in drug discovery by facilitating high-throughput screening and real-time monitoring of drug responses [1]. Such innovations have positioned organoids as a key element in the future of precision medicine, where treatments can be tailored to the unique genetic and phenotypic profiles of individual patients [48].
Looking ahead, the potential for personalized medicine through organoid technology is substantial. Organoids derived from individual patients can be utilized to model specific diseases, thereby allowing for the customization of therapeutic strategies based on a patient’s unique disease characteristics. This personalized approach not only enhances the efficacy of treatments but also minimizes adverse effects by ensuring that therapies are better aligned with the patient's biological context [42]. Furthermore, the integration of multi-omics technologies with organoid models holds promise for providing deeper insights into drug metabolism and therapeutic outcomes, thereby refining personalized treatment protocols [48].
However, challenges remain in fully realizing the potential of organoids in personalized medicine. Issues such as scalability, reproducibility, and the complexity of modeling multifactorial diseases need to be addressed. Additionally, enhancing the vascularization and immune integration of organoids is crucial for improving their physiological relevance [1]. Future research efforts are expected to focus on overcoming these limitations through interdisciplinary collaboration, leveraging advancements in bioengineering, computational biology, and clinical research [42].
In conclusion, organoids represent a transformative advancement in disease modeling and personalized medicine. Their ability to replicate human organ functionality and respond to therapeutic interventions positions them as invaluable tools in biomedical research. As technology continues to evolve, organoids are poised to play a critical role in the development of personalized therapies, ultimately improving patient outcomes in various disease contexts.
6.2 Standardization and Reproducibility in Organoid Research
Organoids play a crucial role in disease modeling due to their ability to closely mimic the architecture and functionality of human tissues. They are derived from stem cells and can replicate the cellular heterogeneity and genetic characteristics of the original organs, thus providing a more physiologically relevant system compared to traditional two-dimensional cell cultures or animal models. This characteristic makes organoids particularly valuable for studying various diseases, including genetic disorders, cancers, and infectious diseases, as they allow researchers to investigate disease mechanisms, test drug responses, and develop personalized medicine approaches.
The advancement of organoid technology has opened new avenues for interdisciplinary research, integrating microfluidics, genetic editing, and artificial intelligence to enhance the modeling of complex diseases. For instance, organoids can be engineered to simulate specific disease states, allowing for the identification of novel pathogenic genes and elucidation of disease mechanisms. This has been particularly significant in the context of genetic disorders, where organoid models have facilitated drug screening platforms and gene-editing therapies [2].
However, despite the promising applications of organoids in disease modeling, there are significant challenges that must be addressed to fully harness their potential. Standardization and reproducibility are critical issues that researchers face in organoid research. The lack of standardized protocols for organoid construction, culture, and experimentation can lead to variability in results, which hinders the translation of findings into clinical practice. Therefore, establishing uniform guidelines and standard operating procedures for organoid research is essential. This would not only enhance the reliability of organoid models but also facilitate the comparison of results across different studies [50].
Future directions in organoid research should focus on improving scalability and reproducibility. This includes developing comprehensive databases for organoid biobanks, which can provide a wide range of patient-derived organoids for research and therapeutic purposes. Additionally, advancements in bioengineering and microfluidic technologies could enhance the complexity of organoid models, allowing for better simulation of in vivo environments and more accurate modeling of disease processes [1].
Furthermore, the integration of multi-omics approaches with organoid technology can provide deeper insights into disease mechanisms and therapeutic responses. By combining transcriptomics, proteomics, and metabolomics, researchers can gain a more comprehensive understanding of how organoids respond to various treatments, paving the way for precision medicine [48].
In summary, organoids serve as powerful tools in disease modeling, offering unprecedented opportunities for advancing our understanding of complex diseases and developing novel therapeutic strategies. However, addressing the challenges of standardization and reproducibility will be vital for the successful translation of organoid research into clinical applications. The future of organoid technology lies in interdisciplinary innovations that enhance their capabilities and reliability in biomedical research [2][48][50].
7 Conclusion
Organoids represent a transformative advancement in biomedical research, particularly in the modeling of diseases. They bridge the gap between traditional two-dimensional cell cultures and in vivo models, providing a more physiologically relevant context for studying complex biological processes. The ability of organoids to replicate the architecture and functionality of actual organs has significantly enhanced our understanding of various diseases, including cancer, neurodegenerative disorders, and infectious diseases. Current research highlights their utility in personalized medicine, where patient-derived organoids can guide tailored therapeutic strategies based on individual genetic profiles. However, challenges remain, including variability in organoid production, limitations in recapitulating complex tissue architectures, and ethical considerations surrounding their use. Future research should focus on standardizing protocols to ensure reproducibility, enhancing organoid functionality through integration with advanced technologies, and addressing the limitations in vascularization and immune integration. The continued evolution of organoid technology holds great promise for reshaping biomedical research and improving clinical outcomes, ultimately leading to more effective treatments for a wide array of diseases.
References
- [1] Ting Huang;Weitao Huang;Qiong Bian. Organoids as predictive platforms: advancing disease modeling, therapeutic innovation, and drug delivery systems.. Journal of controlled release : official journal of the Controlled Release Society(IF=11.5). 2025. PMID:40939865. DOI: 10.1016/j.jconrel.2025.114222.
- [2] Yuanhang Zhu;Nanshan Lin;Juan Li;Haoqian Zhang;Ping Zhang;Xin Cheng;Qian Yang;Ling Liu. Organoids in Genetic Disorders: from Disease Modeling to Translational Applications.. Stem cell reviews and reports(IF=4.2). 2025. PMID:40931310. DOI: 10.1007/s12015-025-10973-x.
- [3] Ji-O Ryu;Yu-Jeong Seong;Eunyoung Lee;Sang-Yun Lee;Dong Woo Lee. Applications and research trends in organoid based infectious disease models.. Scientific reports(IF=3.9). 2025. PMID:40645979. DOI: 10.1038/s41598-025-07816-7.
- [4] Iason Psilopatis;Alexandros G Sykaras;Georgios Mandrakis;Kleio Vrettou;Stamatios Theocharis. Patient-Derived Organoids: The Beginning of a New Era in Ovarian Cancer Disease Modeling and Drug Sensitivity Testing.. Biomedicines(IF=3.9). 2022. PMID:36672509. DOI: 10.3390/biomedicines11010001.
- [5] Ancuta Jurj;Sergiu Pasca;Cornelia Braicu;Ioana Rusu;Schuyler S Korban;Ioana Berindan-Neagoe. Focus on organoids: cooperation and interconnection with extracellular vesicles - Is this the future of in vitro modeling?. Seminars in cancer biology(IF=15.7). 2022. PMID:34896267. DOI: 10.1016/j.semcancer.2021.12.002.
- [6] Qigu Yao;Sheng Cheng;Qiaoling Pan;Jiong Yu;Guoqiang Cao;Lanjuan Li;Hongcui Cao. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine.. MedComm(IF=10.7). 2024. PMID:39309690. DOI: 10.1002/mco2.735.
- [7] Srushti Tambe;Rohit Kumar;Purnima Amin;Monalisa Mishra;Madhu Gupta;Kavitha Govarthanan;Ashwin Kumar Narasimhan;Piyush Kumar Gupta. Current aspects of organoid technology for biomaterial toxicity analysis.. Future medicinal chemistry(IF=3.4). 2023. PMID:37140141. DOI: 10.4155/fmc-2023-0043.
- [8] Juan Wang;Emilien Loeuillard;Gregory J Gores;Sumera I Ilyas. Cholangiocarcinoma: what are the most valuable therapeutic targets - cancer-associated fibroblasts, immune cells, or beyond T cells?. Expert opinion on therapeutic targets(IF=4.4). 2021. PMID:34806500. DOI: 10.1080/14728222.2021.2010046.
- [9] Rebecca J Levy;Sergiu P Paşca. What Have Organoids and Assembloids Taught Us About the Pathophysiology of Neuropsychiatric Disorders?. Biological psychiatry(IF=9.0). 2023. PMID:36739210. DOI: 10.1016/j.biopsych.2022.11.017.
- [10] Katrina Albert;Sara Kälvälä;Vili Hakosalo;Valtteri Syvänen;Patryk Krupa;Jonna Niskanen;Sanni Peltonen;Tuuli-Maria Sonninen;Šárka Lehtonen. Cellular Models of Alpha-Synuclein Aggregation: What Have We Learned and Implications for Future Study.. Biomedicines(IF=3.9). 2022. PMID:36289910. DOI: 10.3390/biomedicines10102649.
- [11] Lucas Felipe de Oliveira;Daniel Mendes Filho;Bruno Lemes Marques;Giovana Figueiredo Maciel;Ricardo Cambraia Parreira;José Rodrigues do Carmo Neto;Priscilla Elias Ferreira Da Silva;Rhanoica Oliveira Guerra;Marcos Vinicius da Silva;Helton da Costa Santiago;Alexander Birbrair;Alexandre H Kihara;Valdo José Dias da Silva;Talita Glaser;Rodrigo R Resende;Henning Ulrich. Organoids as a novel tool in modelling infectious diseases.. Seminars in cell & developmental biology(IF=6.0). 2023. PMID:36182613. DOI: 10.1016/j.semcdb.2022.09.003.
- [12] Yujia Yang;Jinhui Sheng;Fengqiu Sheng;Xiangyang Liang;Cuixia Ma;Ruixue Chen;Shengkun Wang;Xuesong Qian. Advances and applications of organoid-on-a-chip in disease modeling.. The Analyst(IF=3.3). 2025. PMID:41044936. DOI: 10.1039/d5an00514k.
- [13] T Thangam;Krupakar Parthasarathy;K Supraja;V Haribalaji;Vignesh Sounderrajan;Sudhanarayani S Rao;Sakthivel Jayaraj. Lung Organoids: Systematic Review of Recent Advancements and its Future Perspectives.. Tissue engineering and regenerative medicine(IF=4.1). 2024. PMID:38466362. DOI: 10.1007/s13770-024-00628-2.
- [14] Yijing Wang;Dingkun Peng;Meilin Li;Meng Yao;Tianlong Li;Su Li;Hua-Ji Qiu;Lian-Feng Li. Organoids: physiologically relevant ex vivo models for viral disease research.. Journal of virology(IF=3.8). 2025. PMID:40879383. DOI: 10.1128/jvi.01132-25.
- [15] Yue Liu;Jian-Ying Sheng;Chun-Fang Yang;Junjun Ding;Yun-Shen Chan. A decade of liver organoids: Advances in disease modeling.. Clinical and molecular hepatology(IF=16.9). 2023. PMID:36880210. DOI: 10.3350/cmh.2022.0428.
- [16] Jun Li;Wendong Yang;Xiaoli Liu;Keda Yang;Jialin Zhou;Xiaochun Yang. Research progress of lung organoids in infectious respiratory diseases.. European journal of pharmacology(IF=4.7). 2025. PMID:41016568. DOI: 10.1016/j.ejphar.2025.178201.
- [17] Siqi Yang;Haijie Hu;Hengchung Kung;Ruiqi Zou;Yushi Dai;Yafei Hu;Tiantian Wang;Tianrun Lv;Jun Yu;Fuyu Li. Organoids: The current status and biomedical applications.. MedComm(IF=10.7). 2023. PMID:37215622. DOI: 10.1002/mco2.274.
- [18] Joseph Azar;Hisham F Bahmad;Darine Daher;Maya M Moubarak;Ola Hadadeh;Alissar Monzer;Samar Al Bitar;Mohamed Jamal;Mohamed Al-Sayegh;Wassim Abou-Kheir. The Use of Stem Cell-Derived Organoids in Disease Modeling: An Update.. International journal of molecular sciences(IF=4.9). 2021. PMID:34299287. DOI: 10.3390/ijms22147667.
- [19] Sanchita Rauth;Saswati Karmakar;Surinder K Batra;Moorthy P Ponnusamy. Recent advances in organoid development and applications in disease modeling.. Biochimica et biophysica acta. Reviews on cancer(IF=8.3). 2021. PMID:33640383. DOI: 10.1016/j.bbcan.2021.188527.
- [20] Johannes Betge;Rene Jackstadt. From organoids to bedside: Advances in modeling, decoding and targeting of colorectal cancer.. International journal of cancer(IF=4.7). 2023. PMID:36121667. DOI: 10.1002/ijc.34297.
- [21] Chunhui Cai;Xinxin Han. Organoids in biomedicine: Bridging innovation, disease modeling, and regulatory transformation.. Cell transplantation(IF=3.2). 2025. PMID:41126771. DOI: 10.1177/09636897251376507.
- [22] Le Tong;Weiyingqi Cui;Boya Zhang;Pedro Fonseca;Qian Zhao;Ping Zhang;Beibei Xu;Qisi Zhang;Zhen Li;Brinton Seashore-Ludlow;Ying Yang;Longlong Si;Andreas Lundqvist. Patient-derived organoids in precision cancer medicine.. Med (New York, N.Y.)(IF=11.8). 2024. PMID:39341206. DOI: 10.1016/j.medj.2024.08.010.
- [23] Shanqiang Qu;Rongyang Xu;Guozhong Yi;Zhiyong Li;Huayang Zhang;Songtao Qi;Guanglong Huang. Patient-derived organoids in human cancer: a platform for fundamental research and precision medicine.. Molecular biomedicine(IF=10.1). 2024. PMID:38342791. DOI: 10.1186/s43556-023-00165-9.
- [24] Yuxuan Xiao;Yutao Li;Xilin Jing;Lin Weng;Xu Liu;Qingyun Liu;Kezhong Chen. Organoid models in oncology: advancing precision cancer therapy and vaccine development.. Cancer biology & medicine(IF=8.4). 2025. PMID:40708272. DOI: .
- [25] Changchun Zhou;Yuanbo Wu;Zeyu Wang;Yanli Liu;Jiaqi Yu;Weiping Wang;Sunrui Chen;Weihua Wu;Jidong Wang;Guowei Qian;Aina He. Standardization of organoid culture in cancer research.. Cancer medicine(IF=3.1). 2023. PMID:37081739. DOI: 10.1002/cam4.5943.
- [26] Mohammad Hadi Abbasian;Navid Sobhani;Mahsa Mollapour Sisakht;Alberto D'Angelo;Marianna Sirico;Raheleh Roudi. Patient-Derived Organoids: A Game-Changer in Personalized Cancer Medicine.. Stem cell reviews and reports(IF=4.2). 2025. PMID:39432173. DOI: 10.1007/s12015-024-10805-4.
- [27] Angelos Papaspyropoulos;Magdalini Tsolaki;Nicolas Foroglou;Anastasia A Pantazaki. Modeling and Targeting Alzheimer's Disease With Organoids.. Frontiers in pharmacology(IF=4.8). 2020. PMID:32300301. DOI: 10.3389/fphar.2020.00396.
- [28] Amirah Syamimi Jusop;Kalaiselvaan Thanaskody;Gee Jun Tye;Sylvia Annabel Dass;Wan Safwani Wan Kamarul Zaman;Fazlina Nordin. Development of brain organoid technology derived from iPSC for the neurodegenerative disease modelling: a glance through.. Frontiers in molecular neuroscience(IF=3.8). 2023. PMID:37602192. DOI: 10.3389/fnmol.2023.1173433.
- [29] Evanthia A Makrygianni;George P Chrousos. From Brain Organoids to Networking Assembloids: Implications for Neuroendocrinology and Stress Medicine.. Frontiers in physiology(IF=3.4). 2021. PMID:34177605. DOI: 10.3389/fphys.2021.621970.
- [30] Yoshiaki Tanaka;In-Hyun Park. Regional specification and complementation with non-neuroectodermal cells in human brain organoids.. Journal of molecular medicine (Berlin, Germany)(IF=4.2). 2021. PMID:33651139. DOI: 10.1007/s00109-021-02051-9.
- [31] Hanieh Jalali;Sana Rahimian;Nasim Shahsavarian;Rozhan Norouzi;Zahra Ahmadiyeh;Hossein Najafi;Hasti Golchin. The organoid modeling approach to understanding the mechanisms underlying neurodegeneration: A comprehensive review.. Life sciences(IF=5.1). 2024. PMID:39486620. DOI: 10.1016/j.lfs.2024.123198.
- [32] Karl Grenier;Jennifer Kao;Phedias Diamandis. Three-dimensional modeling of human neurodegeneration: brain organoids coming of age.. Molecular psychiatry(IF=10.1). 2020. PMID:31444473. DOI: 10.1038/s41380-019-0500-7.
- [33] Jin Yan;Jean Monlong;Céline Cougoule;Sonia Lacroix-Lamandé;Agnès Wiedemann. Mapping the scientific output of organoids for animal and human modeling infectious diseases: a bibliometric assessment.. Veterinary research(IF=3.5). 2024. PMID:38926765. DOI: 10.1186/s13567-024-01333-7.
- [34] Shirley Pei Shan Chia;Sharleen Li Ying Kong;Jeremy Kah Sheng Pang;Boon-Seng Soh. 3D Human Organoids: The Next "Viral" Model for the Molecular Basis of Infectious Diseases.. Biomedicines(IF=3.9). 2022. PMID:35884846. DOI: 10.3390/biomedicines10071541.
- [35] Sijing Zhu;Dan Chen;Xinzhi Yang;Liuliu Yang;Yuling Han. Organoid Models to Study Human Infectious Diseases.. Cell proliferation(IF=5.6). 2025. PMID:39973397. DOI: 10.1111/cpr.70004.
- [36] Pasquale Marrazzo;Monica Cricca;Claudia Nastasi. Are the Organoid Models an Invaluable Contribution to ZIKA Virus Research?. Pathogens (Basel, Switzerland)(IF=3.3). 2021. PMID:34684182. DOI: 10.3390/pathogens10101233.
- [37] Linhan Sun;Yangmiao Duan;Qingzhan Lan;Yunshan Wang;Alice S T Wong;Yang Ni;Duanrui Liu. Organoids in host pathology: Unveiling microbial-induced therapeutic responses.. Microbiological research(IF=6.9). 2025. PMID:40834629. DOI: 10.1016/j.micres.2025.128313.
- [38] Veronika Bosáková;Marco De Zuani;Lucie Sládková;Zuzana Garlíková;Shyam Sushama Jose;Teresa Zelante;Marcela Hortová Kohoutková;Jan Frič. Lung Organoids-The Ultimate Tool to Dissect Pulmonary Diseases?. Frontiers in cell and developmental biology(IF=4.3). 2022. PMID:35912110. DOI: 10.3389/fcell.2022.899368.
- [39] Lorenzo Zanetto;Luca Bonadies;Raquel Moll-Diaz;Jeffrey Beekman;Maurizio Muraca;Michela Pozzobon;Eugenio Baraldi. Lung organoids: a new frontier in neonatology and paediatric respiratory medicine.. European respiratory review : an official journal of the European Respiratory Society(IF=10.4). 2025. PMID:40769535. DOI: 10.1183/16000617.0255-2024.
- [40] Saniyah Shaikh;Luqman Siddique;Hafsah T Khalifey;Rutaba Mahereen;Thaabit Raziq;Rushdan M Firdous;Aisha Siddique;Ismail M Shakir;Zara Ahmed;Arshiya Akbar;Eman A Alshehri;Raja Chinappan;Alaa Alzhrani;Tanveer Ahmed Mir;Ahmed Yaqinuddin. Brain organoid model systems of neurodegenerative diseases: recent progress and future prospects.. Frontiers in neuroscience(IF=3.2). 2025. PMID:40486729. DOI: 10.3389/fnins.2025.1604435.
- [41] Yisheng Huang;Zhijie Huang;Zhengming Tang;Yuanxin Chen;Mingshu Huang;Hongyu Liu;Weibo Huang;Qingsong Ye;Bo Jia. Research Progress, Challenges, and Breakthroughs of Organoids as Disease Models.. Frontiers in cell and developmental biology(IF=4.3). 2021. PMID:34869324. DOI: 10.3389/fcell.2021.740574.
- [42] Monique M A Verstegen;Rob P Coppes;Anne Beghin;Paolo De Coppi;Mattia F M Gerli;Nienke de Graeff;Qiuwei Pan;Yoshimasa Saito;Shaojun Shi;Amir A Zadpoor;Luc J W van der Laan. Clinical applications of human organoids.. Nature medicine(IF=50.0). 2025. PMID:39901045. DOI: 10.1038/s41591-024-03489-3.
- [43] Masayuki Fujii;Toshiro Sato. Somatic cell-derived organoids as prototypes of human epithelial tissues and diseases.. Nature materials(IF=38.5). 2021. PMID:32807924. DOI: 10.1038/s41563-020-0754-0.
- [44] Long Bai;Yan Wu;Guangfeng Li;Wencai Zhang;Hao Zhang;Jiacan Su. AI-enabled organoids: Construction, analysis, and application.. Bioactive materials(IF=20.3). 2024. PMID:37746662. DOI: 10.1016/j.bioactmat.2023.09.005.
- [45] Intan Rosalina Suhito;Christina Sunil;Andy Tay. Engineering human immune organoids for translational immunology.. Bioactive materials(IF=20.3). 2025. PMID:39474315. DOI: 10.1016/j.bioactmat.2024.10.010.
- [46] Jingyao Chen;Feifei Na. Organoid technology and applications in lung diseases: Models, mechanism research and therapy opportunities.. Frontiers in bioengineering and biotechnology(IF=4.8). 2022. PMID:36568297. DOI: 10.3389/fbioe.2022.1066869.
- [47] Lisa Liu;Lei Yu;Zhichao Li;Wujiao Li;WeiRen Huang. Patient-derived organoid (PDO) platforms to facilitate clinical decision making.. Journal of translational medicine(IF=7.5). 2021. PMID:33478472. DOI: 10.1186/s12967-020-02677-2.
- [48] Dilpreet Singh;Akshay Thakur; Rakesh;Akshay Kumar. Advancements in Organoid-Based Drug Discovery: Revolutionizing Precision Medicine and Pharmacology.. Drug development research(IF=4.2). 2025. PMID:40522262. DOI: 10.1002/ddr.70121.
- [49] Peter V Hauser. Advances in Organoid Research and Developmental Engineering.. Bioengineering (Basel, Switzerland)(IF=3.7). 2024. PMID:39768093. DOI: 10.3390/bioengineering11121275.
- [50] Ruixin Yang;Yao Qi;Xiaoyan Zhang;Hengjun Gao;Yingyan Yu. Living biobank: Standardization of organoid construction and challenges.. Chinese medical journal(IF=7.3). 2024. PMID:39663560. DOI: 10.1097/CM9.0000000000003414.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Organoids · Disease Modeling · Personalized Medicine · Drug Discovery · Neurodegenerative Diseases
© 2025 MaltSci
