🟥 Patient-Specific Organoids for Personalized Disease Modeling and Drug Screening Patient-specific organoid models are revolutionizing the landscape of personalized medicine, providing a powerful platform for modeling individual diseases and testing drug responses. Because these 3D mini-organs are derived from induced pluripotent stem cells (iPSCs) or directly from patient biopsy samples, they replicate the structural and functional characteristics of real tissues, allowing researchers to study disease mechanisms in a highly personalized environment. Unlike traditional 2D cultured cell models or animal models, patient-specific organoid models retain the genetic, molecular, and cellular diversity of each patient, making them an ideal tool for understanding disease progression and treatment resistance. Currently, researchers have successfully developed organoids for a variety of tissues, including the intestine, liver, lung, kidney, brain, and tumors, providing disease-relevant organoid models for diseases such as cystic fibrosis, cancer, neurodegenerative diseases, and infectious diseases. In drug discovery and screening, patient-derived organoid models can also perform high-throughput testing of therapeutic compounds to help determine which drugs or combinations are most effective for an individual. This approach not only accelerates the development of targeted therapies, but also reduces unnecessary exposure to ineffective or toxic drugs, improving patient safety and treatment outcomes. In addition, some universities and biotech companies are building biobanks of organoids representing different populations, which are important for enhancing our understanding of genetic variation and different drug responses. Combined with technologies such as CRISPR gene editing and single-cell sequencing, patient-specific organoids are becoming an important tool for precision medicine, supporting the development of more effective personalized therapies. Looking ahead, as the field develops, patient-specific organoid models will play a central role in customizing treatments, predicting outcomes, and changing the way we study and treat human diseases at the individual level. References [1] Yi Zhao et al., Cell Reports Medicine 2024 (https://lnkd.in/e_VeckQS) [2] Zilong Zhou et al., Frontiers in Oncology 2021 (https://lnkd.in/efgtAFGz) #Organoids #StemCells #PersonalizedMedicine #DrugScreening #DiseaseModeling #PrecisionMedicine #CRISPR #BiotechInnovation #RegenerativeMedicine #3DBiology #NextGenTherapeutics #CSTEAMBiotech
The Importance of Organoids in Scientific Research
Explore top LinkedIn content from expert professionals.
Summary
Organoids are tiny, three-dimensional tissue cultures grown from stem cells that replicate the structure and function of human organs on a small scale. Their importance in scientific research lies in their ability to model human diseases, test drugs, and develop personalized treatments without relying heavily on animal models.
- Understand disease mechanisms: Use organoids to study how diseases develop and progress in human-like environments, which can provide deeper insights than traditional cell or animal models.
- Advance personalized medicine: Leverage patient-specific organoids to test drug responses and create tailored treatments, improving outcomes and reducing exposure to harmful therapies.
- Enhance drug discovery: Employ organoids for high-throughput drug testing, enabling faster identification of effective therapies for complex conditions like cancer and neurodegenerative diseases.
-
-
Recreating the respiratory tract in a dish: Modeling viral infections and testing treatments. Center for iPS Cell Research and Application, Kyoto University. April 24, 2025 Excerpt: RSV is a major respiratory pathogen, particularly among young children, causing severe lower respiratory tract diseases. Current models, such as HEp-2 cells, are commonly used for RSV research, they do not accurately mimic the complex responses of the human respiratory system. Researchers aim to evaluate the utility of respiratory organoids, which are more representative of in vivo human respiratory tract due to the presence of various cell types, in advancing understanding of RSV pathophysiology and evaluating therapeutic and preventive drugs. The study revealed RSV efficiently infected iPS cell-derived respiratory organoids, leading to high viral replication and protein expression. The infected organoids displayed respiratory epithelial layer damage, collagen accumulation, and increased levels of pro-inflammatory cytokines like IL-8 and IFN-γ. Note: Additionally, the researchers found monoclonal antibodies such as nirsevimab, palivizumab, and others targeting RSV F protein were highly effective in inhibiting RSV replication, ribavirin--an antiviral previously used for RSV treatment--showed minimal efficacy. This result highlights the limitations of ribavirin in the organoid model and suggests newer antiviral agents or antibodies may be more promising. The research team also used RNA sequencing and other assays to investigate the host response to RSV infection. RSV-induced changes included a robust innate immune response and activation of genes associated with interferon signaling. The findings demonstrate the organoids' ability to replicate inflammatory and immune responses typically seen during RSV infection in humans. Moreover, the organoids allowed for detailed analysis of cellular interactions and responses, offering an advanced platform for evaluating the effects of antiviral treatments and antibodies on different cell types within the respiratory tract. This study emphasizes the importance of using human iPS cell-derived respiratory organoids for modeling RSV infection. The results suggest organoids are a valuable tool for studying the virus's pathophysiology, testing therapeutic interventions, and advancing the development of effective drugs. The study indicates these models could be utilized to evaluate the efficacy of vaccines and other treatments, providing a more accurate reflection of the human respiratory environment compared to traditional models. Research also suggests incorporating additional immune cell types, such as T cells and neutrophils, may further enhance the model's ability to replicate the complexities of RSV infection, especially in the context of severe inflammation. Refer to enclosed press release to access the paper published 22 April 2025 online. ttps://https://lnkd.in/eSM3mpRR
-
Scientists Create Mini-Brains That Resemble a 40-Day-Old Fetus A Major Leap in Brain Research Researchers at Johns Hopkins University have successfully fused different human organoids to create “mini-brains” that contain approximately 80% of the cell types found in a 40-day-old fetal brain. This breakthrough moves scientists closer than ever to replicating early human brain development in the lab, providing a powerful tool for studying neurological diseases and brain disorders. Bridging the Gap Between Mice and Humans Mini-brains—also known as multi-region brain organoids—are particularly valuable for studying conditions that do not manifest clearly in animal models, such as autism, schizophrenia, and neurodevelopmental disorders. According to lead researcher Annie Kathuria, these lab-grown structures are “a little better than a mouse, a little less than a human”, making them a crucial middle ground for neuroscience research. The ability to replicate human-specific brain development is essential for fields like disease modeling, drug testing, and toxicology studies. By ensuring these organoids closely resemble human fetal brains, scientists can now investigate how environmental factors, genetics, and medications influence early neural development in ways that were previously impossible. Why This Matters for Brain Science • Improved Disease Research – Mini-brains allow scientists to model neurological disorders in human-like systems, leading to better diagnostic and treatment strategies. • Ethical and Practical Advantages – These lab-grown structures offer a way to study human brain development without using fetal tissue. • Toxicology and Environmental Studies – Researchers can now test how drugs, pollutants, and toxins impact the developing brain with greater accuracy. The Future of Lab-Grown Brain Models While these mini-brains are still simplified versions of real human brains, their ability to mimic fetal development represents a major step forward. As organoid technology advances, scientists hope to refine these models further, potentially unlocking new treatments for neurodevelopmental disorders and improving our understanding of the human brain’s earliest stages.
-
Revolutionizing Cancer Care: Real-Time Tumor Avatars to Predict Treatment Success Patient tumor-derived organoids are transforming cancer research by closely mimicking patient tumors, offering a more precise and safe way to study treatment responses compared to traditional models. While their potential has been explored in retrospective studies, real-time application in clinical settings has remained rare—especially for challenging cancers like glioblastoma (GBM), a brain tumor with limited treatment options and poor survival rates. Researchers have now developed patient-derived glioblastoma organoids (GBOs) that can be generated rapidly, maintaining the tumor’s cellular and genetic complexity. For the first time, these GBOs are being used in a clinical trial to test a novel CAR-T cell therapy targeting GBM. By matching organoid studies with patient treatments in real-time, researchers aim to predict therapy effectiveness and improve patient outcomes faster. This innovative approach balances patient safety and cost-efficiency, addressing critical unmet needs in GBM treatment while paving the way for personalized medicine. https://bit.ly/4g5jrlN
-
Patient-derived organoids for precision cancer medicine. PDOs are now central to cancer research—enabling drug testing, biomarker discovery, gene editing, and modeling the tumor microenvironment. Wasatch Biolabs supports organoid studies with RNA, cDNA, and methylation sequencing—optimized for complex 3D models and translational workflows. By replicating the 3D structure and function of real tumors, PDOs capture heterogeneity and spatial organization that 2D cultures and animal models often miss. This review explores advances in organoid culture methods—including air-liquid interface, microfluidics, and organ-on-chip—that are accelerating clinical translation. Read the full publication: https://lnkd.in/gths5JrD #Organoids #CancerResearch #PrecisionMedicine
-
Scientists are growing human brains in labs that can actually think. And they just made a breakthrough that will transform medicine forever: Dutch researchers successfully grew 3D mini-organs from human fetal brain tissue that are revolutionizing how we study diseases. These aren't just clumps of neurons - they're self-organizing structures that replicate early brain development. Each one is just 3-4mm in diameter (size of a rice grain), but what they can do is extraordinary. These mini-brains have achieved what was previously impossible: • Cell density matching real brain conditions • 40% myelination of axons (close to human levels) • Spontaneous electrical activity They form neural networks that communicate with each other and generate patterns similar to early brain development. This breakthrough is transforming how we study Alzheimer's disease. Animal models often fail to predict treatment efficacy in humans - that's why so many promising drugs that worked in animals failed in human trials. But now scientists can: • Create mini-brains from Alzheimer's patients' cells • Watch the disease develop in real-time • Test treatments on actual human tissue The most remarkable discovery? These organoids develop the two hallmark features of Alzheimer's: • Amyloid-β plaques • Tau tangles In just 2 months, we can now study disease progression and test multiple drug combinations rapidly. This raises fascinating ethical questions about consciousness as these mini-brains become more complex. While they're far from achieving consciousness, they're advanced enough to revolutionize treatment of brain diseases by helping us understand: • Neural communication • Disease progression • Drug effects As AI and biotech converge, we're entering a new era where labs must evolve to handle: • Complex experimental data • Multiple data sources • AI integration The future belongs to labs that treat their data as a product - not just collecting it, but making it AI-ready and building scalable systems. Want to accelerate scientific discovery?
-
🟦 Brain Organoids Illuminate TBI’s Link to Neurodegeneration Source: University of Southern California 🔷 Researchers have advanced our understanding of how traumatic brain injuries (#TBI) contribute to #neurodegenerative diseases using #labgrown #brain #organoids. By simulating TBI in organoids derived from human stem cells, the team observed nerve cell death and pathological changes similar to those in TBI patients, particularly in #proteins associated with #ALS and #dementia. 🔷 The discovery of the #gene #KCNJ2 as a #protective factor against TBI effects opens new avenues for treatments. This study, supported by a blend of federal and private funding, underscores the potential of #organoids in medical research and the critical role of genetics in TBI outcomes #neuroscience #genetics #neurology #tbi #also #dementia #healthtech #brainhealth #digitalhealth #healthcare #healthcare #neurotech #innovation https://lnkd.in/eCM63vSq
-
Dual CAR-T: Brain tumor organoids accurately model patient response to CAR T cell therapy - Penn Medicine, University of Pennsylvania Health System PHILADELPHIA— For the first time, researchers used lab-grown organoids created from tumors of individuals with glioblastoma (GBM) to accurately model a patient’s response to CAR T cell therapy in real time. The organoid’s response to therapy mirrored the response of the actual tumor in the patient’s brain. That is, if the tumor-derived organoid shrunk after treatment, so did the patient’s actual tumor, according to new research from the Perelman School of Medicine at the University of Pennsylvania, published today in Cell Stem Cell. “It’s hard to measure how a patient with GBM responds to treatment because we can’t regularly biopsy the brain, and it can be difficult to discern tumor growth from treatment-related inflammation on MRI imaging,” said Hongjun Song, PhD, the Perelman Professor of Neuroscience and co-senior author of the research. “These organoids reflect what is happening in an individual’s brain with great accuracy, and we hope that they can be used in the future to ‘get to know’ each patient’s distinctly complicated tumor and quickly determine which therapies would be most effective for them for personalized medicine.” Patient-derived glioblastoma organoids as real-time avatars for assessing responses to clinical CAR-T cell therapy Highlights •Rapid GBO generation in a phase 1 clinical trial for real-time treatment assessment •A unique trial design with parallel treatments of patients and patient-derived GBOs •Cell cytolysis and target antigen reduction in GBOs treated with patient CAR-T cells •Cytokine release and the degree of cytolysis in GBOs correlated with patient metrics Summary Patient-derived tumor organoids have been leveraged for disease modeling and preclinical studies but rarely applied in real time to aid with interpretation of patient treatment responses in clinics. We recently demonstrated early efficacy signals in a first-in-human, phase 1 study of dual-targeting chimeric antigen receptor (CAR)-T cells (EGFR-IL13Rα2 CAR-T cells) in patients with recurrent glioblastoma. Here, we analyzed six sets of patient-derived glioblastoma organoids (GBOs) treated concurrently with the same autologous CAR-T cell products as patients in our phase 1 study. We found that CAR-T cell treatment led to target antigen reduction and cytolysis of tumor cells in GBOs, the degree of which correlated with CAR-T cell engraftment detected in patients’ cerebrospinal fluid (CSF). Furthermore, cytokine release patterns in GBOs mirrored those in patient CSF samples over time. https://lnkd.in/eusyxeba https://lnkd.in/es-s4MSt
-
Dopaminergic System Organoid Model Illuminates Parkinson’s and Addiction Summary: Researchers developed a novel organoid model that mimics the human dopaminergic system, providing crucial insights into Parkinson’s disease and the impact of chronic cocaine use. Key Facts: * Replication of the Dopaminergic System: The organoid model successfully replicates the human dopaminergic system’s morphology, nerve projections, and functionality. * Potential in Parkinson’s Research: It offers new possibilities for understanding and treating Parkinson’s disease, particularly in developing more effective cell therapies. * Insights into Addiction: The study reveals enduring changes in the dopaminergic circuit following chronic cocaine exposure, highlighting the model’s utility in addiction research. https://lnkd.in/gFXXPrnq
-
Happy Friday all! Check out this open access Nature Gene Therapy review by Vivienne M. Kaiser & Anai Gonzalez-Cordero, "Organoids – the future of pre-clinical development of AAV gene therapy for CNS disorders." Abstract: Advancements in our understanding of genetic disease and adeno-associated virus has prompted great excitement into the field of AAV-mediated gene therapy, particularly for genetic diseases of the central nervous system, including retinal disorders. Despite significant progress, exemplified by the approval of therapies such as Luxturna® and Zolgensma®, a substantial number of therapies remain in pre-clinical or early clinical stages, with many failing to advance to later phases. Whilst the use of animal models to test safety and delivery route efficacy of AAV treatments is imperative, differences in tissue structure and physiology between humans and animal models has restricted precise disease modelling and gene therapy development for many CNS disorders. Alongside the FDA push for non-animal alternative models, researchers are increasingly turning to human-based models, including stem cell-derived organoids, which can offer a more accurate representation of human cellular microenvironments and niches. As such, this review explores the advantages and limitations of brain and retinal organoids as pre-clinical models of disease, with a primary focus on their utility in identifying novel AAV capsids, cell-specific promoters, and their role in recent pre-clinical AAV gene therapy studies. #drugdiscovery #aav #cns #retina #organoids #scientificresearch