Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (72)

Search Parameters:
Keywords = patient-derived organoid/PDO

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 2477 KiB  
Review
Deciphering Breast Tumor Heterogeneity Through Patient-Derived Organoids and Circulating Tumor Cells
by Benedetta Policastro, Nikoline Nissen and Carla L. Alves
J. Pers. Med. 2025, 15(7), 271; https://doi.org/10.3390/jpm15070271 - 25 Jun 2025
Viewed by 566
Abstract
Breast cancer is a highly heterogeneous disease, with tumors capable of adapting to shifting conditions, making the development of effective personalized therapies particularly challenging. Patient-derived models, such as patient-derived organoids (PDOs) and circulating tumor cell (CTC) cultures, have emerged as powerful tools for [...] Read more.
Breast cancer is a highly heterogeneous disease, with tumors capable of adapting to shifting conditions, making the development of effective personalized therapies particularly challenging. Patient-derived models, such as patient-derived organoids (PDOs) and circulating tumor cell (CTC) cultures, have emerged as powerful tools for investigating intra- and inter-tumor heterogeneity. These models largely retain the genetic, phenotypic, and microenvironmental features of the original tumors, providing valuable insights into disease progression, drug response, and resistance mechanisms. Furthermore, by enabling tumors’ spatiotemporal molecular profiling, PDOs and CTCs offer a dynamic approach to assess treatment efficacy over time. However, to fully capture the complexity of breast cancer heterogeneity, it is required to develop models from multiple tumor and blood samples collected throughout the course of treatment. This review explores the potential of integrating PDOs and CTC models to better understand intra-tumor heterogeneity while addressing key challenges in developing patient-derived models that accurately recapitulate patients’ tumors to advance personalized care. The integration of PDOs and CTCs could represent a paradigm shift in the personalized management of metastatic breast cancer. Full article
(This article belongs to the Section Disease Biomarker)
Show Figures

Figure 1

23 pages, 913 KiB  
Review
The Role of Organoids in Advancing Colorectal Cancer Research: Insights and Future Directions
by Zahra Heydari, Rex Devasahayam Arokia Balaya, Gobinda Sarkar and Lisa Boardman
Cancers 2025, 17(13), 2129; https://doi.org/10.3390/cancers17132129 - 25 Jun 2025
Viewed by 776
Abstract
Organoids are three-dimensional (3D) structures that mimic the architecture and functionality of human organs, providing a novel approach to study diseases such as colorectal cancer (CRC). This review aims to explore the impact of organoids on understanding CRC and their potential use in [...] Read more.
Organoids are three-dimensional (3D) structures that mimic the architecture and functionality of human organs, providing a novel approach to study diseases such as colorectal cancer (CRC). This review aims to explore the impact of organoids on understanding CRC and their potential use in exploring therapeutic outcomes. Colorectal cancer, characterized by the transformation of colonic epithelial cells into adenomas and carcinomas, remains one of the top causes of cancer-related morbidity and mortality worldwide. Traditional two-dimensional (2D) cell cultures fail to replicate the tumor microenvironment in an effective manner, which highlights the need for advanced 3D models. Organoids preserve the genetic and phenotypic properties of the original tumors, allowing for improved disease modeling, drug screening, and personalized medicine applications. When using patient-derived organoids (PDOs), researchers can gain insights into CRC initiation, progression, and treatment outcome. Ultimately, organoids represent an encouraging platform for improving therapeutic strategies for CRC, potentially leading to better patient outcomes through tailored treatment approaches. Full article
(This article belongs to the Section Transplant Oncology)
Show Figures

Figure 1

11 pages, 2928 KiB  
Communication
Resistance to MAPK Pathway Inhibition in BRAF-V600E Mutant Colorectal Cancer Can Be Overcome with Insulin Receptor/Insulin-like Growth Factor-1 Receptor Inhibitors
by Layla El Bouazzaoui, Daniëlle A. E. Raats, André Verheem, Inne H. M. Borel Rinkes, Hugo J. G. Snippert, Madelon M. Maurice and Onno Kranenburg
Organoids 2025, 4(2), 14; https://doi.org/10.3390/organoids4020014 - 12 Jun 2025
Viewed by 367
Abstract
The current treatment for refractory BRAF-V600E mutant metastatic colorectal cancer (mCRC) involves combined inhibition of BRAF and the epidermal growth factor receptor (EGFR). However, tumour responses are often short-lived due to a rebound in mitogen-activated protein kinase (MAPK) activity. In this study, [...] Read more.
The current treatment for refractory BRAF-V600E mutant metastatic colorectal cancer (mCRC) involves combined inhibition of BRAF and the epidermal growth factor receptor (EGFR). However, tumour responses are often short-lived due to a rebound in mitogen-activated protein kinase (MAPK) activity. In this study, we combined short-term cell viability assays with long-term regrowth assays following drug removal over a period of three weeks. This allowed assessment of regrowth after therapy discontinuation. We tested the effect of combined BRAF inhibition (encorafenib) and EGFR inhibition (afatinib) on BRAF-V600E mutant CRC patient-derived organoids (PDOs). Combined EGFR/BRAF inhibition initially caused a major reduction in PDO growth capacity in BRAF-V600E mutant PDOs. This was followed by rapid regrowth after drug removal, mirroring clinical outcomes. EGFR inhibition in BRAF-V600E mutant PDOs led to activation of the insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R). The IGF1R/IR inhibitor linsitinib prevented the rebound in MAPK activity following removal of afatinib and encorafenib, prevented regrowth of CRC PDOs, and improved the anti-tumour response in an in vivo model. PDO regrowth assays allow the identification of pathways driving tumour recurrence. IR/IGF1R-inhibition prevents regrowth following golden standard MAPK pathway-targeted therapy and provides a strategy to improve the treatment of BRAF-V600E mutant CRC Full article
Show Figures

Figure 1

16 pages, 21692 KiB  
Article
Precision Medicine: IL-1RA and Pancreatic Cancer Organoids
by Annah G. Morgan, Michelle F. Griffin, Michael T. Longaker and Jeffrey A. Norton
Biology 2025, 14(6), 604; https://doi.org/10.3390/biology14060604 - 25 May 2025
Viewed by 1131
Abstract
Cancer organoids have emerged as transformative models for studying tumor biology and therapeutic responses due to the ability to replicate the complexity of the tumor microenvironment (TME). Tumor organoids recapitulate the genetic and phenotypic diversity of cancers, making them invaluable for investigating mechanisms [...] Read more.
Cancer organoids have emerged as transformative models for studying tumor biology and therapeutic responses due to the ability to replicate the complexity of the tumor microenvironment (TME). Tumor organoids recapitulate the genetic and phenotypic diversity of cancers, making them invaluable for investigating mechanisms of resistance and identifying novel therapeutic targets. Patient-derived organoids (PDOs) allow specific treatment methods to be designed based on the properties of each individual tumor in vitro. Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with an immunosuppressive nature. PDAC has a poor prognosis, with the survival rates of metastatic PDAC being improved only minimally over the last few decades. In this study, we demonstrate the antitumor effects of an IL-1 receptor antagonist (IL-1RA) in murine and human PDAC organoids. By reducing the burden of suppressive tumor elements like CAFs, IL-1RA treatment facilitates better immune cell access and response. Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Graphical abstract

16 pages, 5955 KiB  
Article
The Association of OLFM4 with the Progression and Cisplatin Resistance of Head and Neck Squamous Carcinoma
by Xinlu He, Xi Yao, Keling Pang, Xulin Chen, Zhengbo Wei and Ying Xie
Curr. Oncol. 2025, 32(5), 276; https://doi.org/10.3390/curroncol32050276 - 13 May 2025
Viewed by 548
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly prevalent malignant tumor globally with a poor prognosis. Despite continuous advancements in treatment modalities, the molecular mechanisms underlying its progression and chemotherapy resistance remain unclear. In previous studies, cisplatin drug induction was performed [...] Read more.
Head and neck squamous cell carcinoma (HNSCC) is a highly prevalent malignant tumor globally with a poor prognosis. Despite continuous advancements in treatment modalities, the molecular mechanisms underlying its progression and chemotherapy resistance remain unclear. In previous studies, cisplatin drug induction was performed on HNSCC patient-derived tumor organoids (HNSCC-PDOs), successfully establishing a cisplatin-resistant organoid model (HNSCC-PDOcisR). This study conducted RNA sequencing on cisplatin-resistant HNSCC-PDOcisR and their parental PDOs. Bioinformatic analysis revealed that the oncoprotein olfactomedin 4 (OLFM4) was significantly upregulated in the drug-resistant model. Combined analysis of TCGA and CPTAC databases demonstrated that OLFM4 expression correlates with poor clinical prognosis in HNSCC. In vitro cellular experiments verified that OLFM4 overexpression significantly enhanced HNSCC cell proliferation, migration, and invasion capabilities (p < 0.05), while OLFM4 knockdown inhibited these phenotypes. Additionally, OLFM4 was found to mediate cisplatin resistance by regulating levels of reactive oxygen species (ROS), malondialdehyde (MDA), and ferrous ions (Fe2⁺), suppressing cisplatin-induced oxidative stress and ferroptosis while maintaining mitochondrial membrane potential. This study confirms that OLFM4 enhances tumor cell proliferation, migration, and resistance to cisplatin-induced cell death, thereby promoting HNSCC progression. These findings suggest OLFM4 may serve as a prognostic biomarker for HNSCC and a potential therapeutic target to reverse cisplatin resistance in HNSCC. Full article
(This article belongs to the Section Head and Neck Oncology)
Show Figures

Figure 1

27 pages, 1566 KiB  
Review
Facing the Challenge to Mimic Breast Cancer Heterogeneity: Established and Emerging Experimental Preclinical Models Integrated with Omics Technologies
by Alessia Ciringione and Federica Rizzi
Int. J. Mol. Sci. 2025, 26(10), 4572; https://doi.org/10.3390/ijms26104572 - 10 May 2025
Viewed by 1229
Abstract
Breast cancer (BC) is among the most common neoplasms globally and is the leading cause of cancer-related mortality in women. Despite significant advancements in prevention, early diagnosis, and treatment strategies made over the past two decades, breast cancer continues to pose a significant [...] Read more.
Breast cancer (BC) is among the most common neoplasms globally and is the leading cause of cancer-related mortality in women. Despite significant advancements in prevention, early diagnosis, and treatment strategies made over the past two decades, breast cancer continues to pose a significant global health challenge. One of the major obstacles in the clinical management of breast cancer patients is the high intertumoral and intratumoral heterogeneity that influences disease progression and therapeutic outcomes. The inability of preclinical experimental models to replicate this diversity has hindered the comprehensive understanding of BC pathogenesis and the development of new therapeutic strategies. An ideal experimental model must recapitulate every aspect of human BC to maintain the highest predictive validity. Therefore, a thorough understanding of each model’s inherent characteristics and limitations is essential to bridging the gap between basic research and translational medicine. In this context, omics technologies serve as powerful tools for establishing comparisons between experimental models and human tumors, which may help address BC heterogeneity and vulnerabilities. This review examines the BC models currently used in preclinical research, including cell lines, patient-derived organoids (PDOs), organ-on-chip technologies, carcinogen-induced mouse models, genetically engineered mouse models (GEMMs), and xenograft mouse models. We emphasize the advantages and disadvantages of each model and outline the most important applications of omics techniques to aid researchers in selecting the most relevant model to address their specific research questions. Full article
(This article belongs to the Special Issue Breast Cancer: From Pathophysiology to Novel Therapies)
Show Figures

Figure 1

15 pages, 2899 KiB  
Article
ECM Stiffness-Induced Redox Signaling Enhances Stearoyl Gemcitabine Efficacy in Pancreatic Cancer
by Shuqing Zhao, Edward Agyare, Xueyou Zhu, Jose Trevino, Sherise Rogers, Enrique Velazquez-Villarreal, Jason Brant, Payam Eliahoo, Jonathan Barajas, Ba Xuan Hoang and Bo Han
Cancers 2025, 17(5), 870; https://doi.org/10.3390/cancers17050870 - 3 Mar 2025
Cited by 1 | Viewed by 1344
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, largely due to its dense fibrotic stroma that promotes drug resistance and tumor progression. While patient-derived organoids (PDOs) have emerged as promising tools for modeling PDAC and evaluating therapeutic responses, the [...] Read more.
Background: Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, largely due to its dense fibrotic stroma that promotes drug resistance and tumor progression. While patient-derived organoids (PDOs) have emerged as promising tools for modeling PDAC and evaluating therapeutic responses, the current PDO models grown in soft matrices fail to replicate the tumor’s stiff extracellular matrix (ECM), limiting their predictive value for advanced disease. Methods: We developed a biomimetic model using gelatin-based matrices of varying stiffness, achieved through modulated transglutaminase crosslinking rates, to better simulate the desmoplastic PDAC microenvironment. Using this platform, we investigated organoid morphology, proliferation, and chemoresistance to gemcitabine (Gem) and its lipophilic derivative, 4-N-stearoyl gemcitabine (Gem-S). Mechanistic studies focused on the interplay between ECM stiffness, hypoxia-inducible factor (HIF) expression, and the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in drug resistance. Results: PDAC organoids in stiffer matrices demonstrated enhanced stemness features, including rounded morphology and elevated cancer stem cell (CSC) marker expression. Matrix stiffness-induced gemcitabine resistance correlated with the upregulation of ABC transporters and oxidative stress adaptive responses. While gemcitabine activated Nrf2 expression, promoting oxidative stress mitigation, Gem-S suppressed Nrf2 levels and induced oxidative stress, leading to increased reactive oxygen species (ROS) and enhanced cell death. Both compounds reduced HIF expression, with gemcitabine showing greater efficacy. Conclusions: Our study reveals ECM stiffness as a critical mediator of PDAC chemoresistance through the promotion of stemness and modulation of Nrf2 and HIF pathways. Gem-S demonstrates promise in overcoming gemcitabine resistance by disrupting Nrf2-mediated adaptive responses and inducing oxidative stress. These findings underscore the importance of biomechanically accurate tumor models and suggest that dual targeting of mechanical and oxidative stress pathways may improve PDAC treatment outcomes. Full article
(This article belongs to the Section Cancer Drug Development)
Show Figures

Figure 1

25 pages, 1451 KiB  
Article
Individualized Pooled CRISPR/Cas9 Screenings Identify CDK2 as a Druggable Vulnerability in a Canine Mammary Carcinoma Patient
by Marine Inglebert, Martina Dettwiler, Chang He, Enni Markkanen, Lennart Opitz, Arunasalam Naguleswaran and Sven Rottenberg
Vet. Sci. 2025, 12(2), 183; https://doi.org/10.3390/vetsci12020183 - 18 Feb 2025
Cited by 1 | Viewed by 1350
Abstract
High-throughput omics approaches have long been used to uncover potential vulnerabilities in human personalized oncology but are often limited by the lack of functional validation. Therefore, we placed our emphasis on functional drug testing using patient-derived organoids (PDOs). However, PDOs generated from tumors [...] Read more.
High-throughput omics approaches have long been used to uncover potential vulnerabilities in human personalized oncology but are often limited by the lack of functional validation. Therefore, we placed our emphasis on functional drug testing using patient-derived organoids (PDOs). However, PDOs generated from tumors mostly lack comparison with matching normal tissue, and the number of testable drugs is limited. Here, we demonstrate how matching the neoplastic and non-neoplastic mammary PDOs derived from the same dog can utilize targeted CRISPR/Cas9 screens to unveil cancer cell specific vulnerabilities. We performed two independent CRISPR/Cas9 dropout screens using sub-libraries targeting the epigenome (n = 1269) or druggable genes (n = 834) in paired PDOs derived from both carcinoma and normal mammary tissues from the same dog. A comparison of essential genes for tumor cells survival identified CDK2 as a functional vulnerability in canine mammary tumors (CMTs) that can be targeted with the PF3600 inhibitor. Additional potential targets were also uncovered, providing insights for personalized cancer treatments in dogs. Full article
(This article belongs to the Special Issue Focus on Tumours in Pet Animals)
Show Figures

Figure 1

26 pages, 1886 KiB  
Review
Establishing Pancreatic Cancer Organoids from EUS-Guided Fine-Needle Biopsy Specimens
by Mei-Juan Wang, Chao Gao, Xin Huang, Min Wang, Shuai Zhang, Xiao-Pei Gao, Chang-Qing Zhong and Lian-Yong Li
Cancers 2025, 17(4), 692; https://doi.org/10.3390/cancers17040692 - 18 Feb 2025
Viewed by 1218
Abstract
Pancreatic cancer is a highly malignant digestive system tumor characterized by covert onset and rapid progression, with a 5-year survival rate of less than 10%. Most patients have already reached an advanced or metastatic stage at the time of diagnosis. Therefore, it is [...] Read more.
Pancreatic cancer is a highly malignant digestive system tumor characterized by covert onset and rapid progression, with a 5-year survival rate of less than 10%. Most patients have already reached an advanced or metastatic stage at the time of diagnosis. Therefore, it is particularly important to study the occurrence, development, and drug resistance mechanisms of pancreatic cancer. In recent years, the development of 3D tumor cell culture technology has provided new avenues for pancreatic cancer research. Patient-derived organoids (PDOs) are micro-organ structures that are obtained directly from the patient’s body and rapidly expand in vitro. PDOs have the ability to self-renew and self-organize and retain the genetic heterogeneity and molecular characteristics of the original tumor. However, the use of organoids is limited because most patients with pancreatic ductal adenocarcinoma (PDAC) are inoperable. Endoscopic ultrasound-guided fine-needle aspiration/biopsy (EUS-FNA/FNB) is an important method for obtaining tissue samples from non-surgical pancreatic cancer patients. This article reviews the factors that affect the formation of pancreatic cancer organoids using EUS-FNA/FNB. High-quality samples, sterile operations, and optimized culture media are key to successfully generating organoids. Additionally, individual patient differences and disease stages can impact the formation of organoids. Pancreatic cancer organoids constructed using EUS-FNA/FNB have significant potential, suggesting new approaches for research and treatment. Full article
(This article belongs to the Special Issue Endoscopic Management of Pancreatic Neoplasms (2nd Edition))
Show Figures

Figure 1

12 pages, 2142 KiB  
Article
Enhancing Progestin Therapy with a Glucagon-Like Peptide 1 Agonist for the Conservative Management of Endometrial Cancer
by Andrea R. Hagemann, Ian S. Hagemann, David G. Mutch, Eric J. Devor, Paige K. Malmrose, Yuping Zhang, Abigail M. Morrison, Kristina W. Thiel and Kimberly K. Leslie
Cancers 2025, 17(4), 598; https://doi.org/10.3390/cancers17040598 - 10 Feb 2025
Viewed by 1843
Abstract
Objective: Obesity is a major risk factor for endometrial cancer. In addition to hormone therapy with progestins, glucagon like peptide-1 receptor (GLP-1R) agonists such as semaglutide may be helpful to achieve weight loss during conservative treatment of endometrial hyperplasia or cancer. Methods: We [...] Read more.
Objective: Obesity is a major risk factor for endometrial cancer. In addition to hormone therapy with progestins, glucagon like peptide-1 receptor (GLP-1R) agonists such as semaglutide may be helpful to achieve weight loss during conservative treatment of endometrial hyperplasia or cancer. Methods: We theorized that the combination of semaglutide and the progestin levonorgestrel would be useful as a novel treatment or prevention regimen and tested this hypothesis using endometrial cancer cell lines and patient-derived organoids (PDOs). Results: Hec50, KLE, and Ishikawa endometrial cancer cells express GLP-1R, as determined by both qPCR and Western blotting, and GLP-1R agonist treatment induces GLP-1R mRNA transcription through positive feedback mechanisms in cell models. PDOs from six individuals with grade 1 endometrial carcinomas were treated with progesterone, levonorgestrel, semaglutide, or levonorgestrel + semaglutide. Multiple models demonstrated a significant reduction in viability in response to combinatorial treatment, and the effect was noted in models from both PR high- and PR low-expressing tumors. Most interesting was the induction not only of the membrane GLP-1R with treatment, but also the significant upregulation of nuclear and membrane progesterone receptors—PR and PGRMC1/2, respectively—indicating a potential positive feedback loop between semaglutide and progestins such as levonorgestrel. Conclusion: In summary, we identify synergistic molecular cross-talk between the GLP-1R and steroid hormone receptor pathways, with the potential to enhance the anticancer activity of levonorgestrel when combined with semaglutide. Full article
(This article belongs to the Special Issue Gynecologic Cancer: Risk Factors, Interception and Prevention)
Show Figures

Figure 1

22 pages, 3136 KiB  
Article
GATA3-Driven ceRNA Network in Lung Adenocarcinoma Bone Metastasis Progression and Therapeutic Implications
by Yun Liu, Shihui Shen, Xudong Wang, Hansen Chen, Wenjie Ren, Haifeng Wei, Kun Li and Lei Li
Cancers 2025, 17(3), 559; https://doi.org/10.3390/cancers17030559 - 6 Feb 2025
Cited by 3 | Viewed by 1406
Abstract
Background/Objectives: Bone metastasis is a common and severe complication of lung adenocarcinoma (LUAD), impacting prognosis and treatment outcomes. Understanding the molecular mechanisms behind LUAD bone metastasis (LUADBM) is essential for developing new therapeutic strategies. The interactions between long non-coding RNAs (lncRNAs), microRNAs [...] Read more.
Background/Objectives: Bone metastasis is a common and severe complication of lung adenocarcinoma (LUAD), impacting prognosis and treatment outcomes. Understanding the molecular mechanisms behind LUAD bone metastasis (LUADBM) is essential for developing new therapeutic strategies. The interactions between long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and mRNAs in the competing endogenous RNA (ceRNA) network are crucial in cancer progression and metastasis, but the regulatory mechanisms in LUADBM remain unclear. Methods: Microarray analysis was performed on clinical samples, followed by weighted gene co-expression network analysis (WGCNA) and construction of a ceRNA network. Molecular mechanisms were validated using colony formation assays, transwell migration assays, wound healing assays to assess cell migration, and osteoclastogenesis assays to evaluate osteoclast differentiation. Potential therapeutic drugs and their binding affinities were predicted using the CMap database and Kdeep. The interaction between the small-molecule drug and its target protein was confirmed by surface plasmon resonance (SPR) and drug affinity responsive target stability (DARTS) assays. Mechanistic insights and therapeutic efficacy were further validated using patient-derived organoid (PDO) cultures, drug sensitivity assays, and in vivo drug treatments. Results: Our results identified the XLOC_006941/hsa-miR-543/NPRL3 axis as a key regulatory pathway in LUADBM. We also demonstrated that GATA3-driven Th2 cell infiltration creates an immunosuppressive microenvironment that promotes metastasis. Additionally, we confirmed that the inhibitor E7449 effectively targets NPRL3, and its combination with the IL4R-blocking antibody dupilumab resulted in improved therapeutic outcomes in LUADBM. Conclusions: These findings offer new insights into the molecular mechanisms of LUADBM and highlight potential therapeutic targets, including the XLOC_006941/miR-543/NPRL3 axis and GATA3-driven Th2 cell infiltration. The dual-target therapy combining E7449 with dupilumab shows promise for improving patient outcomes in LUADBM, warranting further clinical evaluation. Full article
(This article belongs to the Special Issue Bone and Spine Metastases)
Show Figures

Figure 1

17 pages, 3770 KiB  
Article
Treatment Response to Oncolytic Virus in Patient-Derived Breast Cancer and Hypopharyngeal Cancer Organoids: Evaluation via a Microfluidics Organ-on-a-Chip System
by Yu Sun, Jiaqi Liu, Li Zhu, Fang Huang, Yanbo Dong, Shuang Liu, Siyi Chen, Wei Ji, Jingjing Lu, Liangfa Liu and Shanhu Li
Bioengineering 2025, 12(2), 146; https://doi.org/10.3390/bioengineering12020146 - 4 Feb 2025
Cited by 1 | Viewed by 1683
Abstract
In this study, we present an oncolytic virus (OV) evaluation system established using microfluidic organ-on-a-chip (OOC) systems and patient-derived organoids (PDOs), which was used in the development of a novel oncolytic virus, AD4-GHPE. An OV offers advantages such as good targeting ability and [...] Read more.
In this study, we present an oncolytic virus (OV) evaluation system established using microfluidic organ-on-a-chip (OOC) systems and patient-derived organoids (PDOs), which was used in the development of a novel oncolytic virus, AD4-GHPE. An OV offers advantages such as good targeting ability and minimal side effects, and it has achieved significant breakthroughs when combined with immunotherapy in recent clinical trials. The development of OVs has become an emerging research focus. PDOs can preserve the heterogeneity of in situ tumor tissues, whereas microfluidic OOC systems can automate and standardize various experimental procedures. These systems have been applied in cutting-edge drug screening and cell therapy experiments; however, their use in functionally complex oncolytic viruses remains to be explored. In this study, we constructed a novel recombinant oncolytic adenovirus, AD4-GHPE, and evaluated OOC systems and PDOs through various functional validations in hypopharyngeal and breast cancer organoids. The results confirmed that AD4-GHPE exhibits three antitumor mechanisms, namely, tumor-specific cytotoxicity, a reduction in programmed death ligand 1 (PD-L1) expression in tumor cells to increase CD8+ T-cell activity, and granulocyte–macrophage colony-stimulating factor (GM-CSF) secretion. The evaluation system combining OOC systems and PDOs was efficient and reliable, providing personalized OV treatment recommendations for patients and offering industrialized and standardized research ideas for the development of OVs. Full article
(This article belongs to the Section Regenerative Engineering)
Show Figures

Figure 1

23 pages, 2446 KiB  
Review
Patient-Derived Organoid Models for NKT Cell-Based Cancer Immunotherapy
by Pablo A. Palacios, Iván Flores, Lucas Cereceda, Francisco F. Otero, Marioly Müller, Priscilla Brebi, Héctor R. Contreras and Leandro J. Carreño
Cancers 2025, 17(3), 406; https://doi.org/10.3390/cancers17030406 - 26 Jan 2025
Viewed by 2206
Abstract
Invariant Natural Killer T (iNKT) cells are a unique subset of T cells that bridge innate and adaptive immunity, displaying potent anti-tumor properties through cytokine secretion, direct cytotoxicity, and recruitment of immune effector cells such as CD8+ T cells and NK cells. [...] Read more.
Invariant Natural Killer T (iNKT) cells are a unique subset of T cells that bridge innate and adaptive immunity, displaying potent anti-tumor properties through cytokine secretion, direct cytotoxicity, and recruitment of immune effector cells such as CD8+ T cells and NK cells. Despite their therapeutic potential, the immunosuppressive tumor microenvironment (TME), characterized by regulatory T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), limits iNKT cell efficacy. Patient-derived organoid (PDO) platforms provide an innovative model for dissecting these complex interactions and evaluating strategies to reinvigorate iNKT cell functionality within the TME. PDOs closely mimic the genetic, phenotypic, and structural characteristics of primary tumors, enabling the study of tumor–immune dynamics. Integrating iNKT cells into PDOs offers a robust platform for investigating CD1d-mediated interactions, Th1-biased immune responses driven by glycolipid analogs like α-GalCer, and combination therapies such as immune checkpoint inhibitors. Additionally, PDO systems can assess the effects of metabolic modulation, including reducing lactic acid accumulation or targeting glutamine pathways, on enhancing iNKT cell activity. Emerging innovations, such as organoid-on-a-chip systems, CRISPR-Cas9 gene editing, and multi-omics approaches, further expand the potential of PDO–iNKT platforms for personalized immunotherapy research. Although the application of iNKT cells in PDOs is still undeveloped, these systems hold immense promise for bridging preclinical studies and clinical translation. By addressing the challenges of the TME and optimizing therapeutic strategies, PDO–iNKT platforms offer a transformative avenue for advancing cancer immunotherapy and personalized medicine. Full article
(This article belongs to the Special Issue Novel Targeted Therapies for T-cell Malignancies)
Show Figures

Figure 1

18 pages, 1150 KiB  
Review
Precision Medicine for Peritoneal Carcinomatosis—Current Advances in Organoid Drug Testing and Clinical Applicability
by Harleen Kaur, Josephine A. Wright, Daniel L. Worthley, Elizabeth Murphy and Susan L. Woods
Organoids 2025, 4(1), 2; https://doi.org/10.3390/organoids4010002 - 24 Jan 2025
Cited by 1 | Viewed by 1508
Abstract
Peritoneal carcinomatosis from gastrointestinal tumours is considered a poor prognostic factor, with a median overall survival of six to nine months in the absence of intervention. The advent of patient-derived organoid cultures (PDOs) has provided a breakthrough in personalised medicine, allowing researchers and [...] Read more.
Peritoneal carcinomatosis from gastrointestinal tumours is considered a poor prognostic factor, with a median overall survival of six to nine months in the absence of intervention. The advent of patient-derived organoid cultures (PDOs) has provided a breakthrough in personalised medicine, allowing researchers and clinicians to model the complexity and heterogeneity of individual tumours in vitro. PDOs hold great promise in this field, as variations in the management of peritoneal carcinomatosis due to differences in the method of delivery of chemotherapeutics, drug selection, exposure duration, and tumour pathology make it impractical to use a single, standardised treatment regimen. We aim to summarise the methodologies and limitations of studies encapsulating organoids derived from peritoneal metastases to encourage design considerations that may improve future clinical relevance, standardise protocols, and address translational challenges in personalising treatment strategies. Full article
Show Figures

Figure 1

18 pages, 9612 KiB  
Article
A Two-Step Protocol for Isolation and Maintenance of Lung Cancer Primary 3D Cultures
by Silvia Strocchi, Giacomo Santandrea, Eleonora Zanetti, Giulio Verna, Vincenza Ylenia Cusenza, Davide Nicoli, Valentina Fantini, Alessandra Grieco, Massimiliano Paci, Alessia Ciarrocchi and Valentina Sancisi
Cancers 2025, 17(1), 27; https://doi.org/10.3390/cancers17010027 - 25 Dec 2024
Viewed by 1115
Abstract
Background/Objectives: Despite the introduction of innovative therapeutics, lung cancer is still the leading cause of cancer-related death. For this reason, lung cancer still requires deep characterization to identify cellular and molecular targets that can be used to develop novel therapeutic strategies. Three-dimensional cellular [...] Read more.
Background/Objectives: Despite the introduction of innovative therapeutics, lung cancer is still the leading cause of cancer-related death. For this reason, lung cancer still requires deep characterization to identify cellular and molecular targets that can be used to develop novel therapeutic strategies. Three-dimensional cellular models, including patient-derived organoids (PDOs), represent useful tools to study lung cancer biology and may be employed in the future as predictive tools in therapeutic decisions. However, the successful establishment of lung cancer organoids cultures that faithfully represent the respective patient tissues is still challenging due to low success rate and/or overgrowth of normal airway epithelial cells. Methods: We set up a two-step protocol that allows for establishing both short-term and long-term 3D cultures, with different characteristics and success rates. Results: Cancer tissue-originated spheroids (CTOSs) show a 100% success rate and allow for the concomitant isolation of autologous tumor infiltrating leukocytes (TILs). On the contrary, PDOs can be expanded for a medium-long term and bio-banked but retain a lower success rate and the possibility of contamination with normal airway epithelial cells. To overcome these problems, we set up an optimal medium formulation and we implemented rigorous quality controls, leading to a substantial improvement in the success rate of tumoral PDO establishment. Conclusions: Overall, this protocol guarantees flexibility and reliability, also providing useful guidelines for quality control checks to support different experimental settings. The setting up of a robust protocol for lung cancer PDO culture establishment and expansion is a key requirement for their employment both in cancer research and as predictive tools in clinical practice. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Figure 1

Back to TopTop