Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Cancer Res ; 84(15): 2397-2399, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39086314

ABSTRACT

Over the past three decades, high-throughput phenotypic cancer cell line screens have revealed unanticipated small-molecule activities and illuminated connections between tumor genotypes and anticancer efficacy. Founded in 1984, the National Cancer Institute's "NCI60" screen laid the conceptual groundwork for the contemporary landscape of phenotypic drug discovery. NCI60 first operated as a primary bioactivity screen, but molecular characterization of the NCI60 cell line panel and development of a small-molecule sensitivity pattern recognition algorithm (called "COMPARE") have enabled subsequent studies into drug mechanisms of action and biomarker identification. In this issue of Cancer Research, Kunkel and colleagues report an updated version of the NCI60 screen, dubbed "HTS384" NCI60, that better aligns with current cell proliferation assay standards and has higher throughput. Changes include the use of a 384-well plate format, automated laboratory equipment, 3 days of compound exposure, and a CellTiter-Glo luminescent endpoint. To confirm that data from the HTS384 and classic NCI60 screen are comparable, the authors tested a library of 1,003 anticancer agents using both protocols and applied COMPARE to analyze patterns of cell line sensitivities. More than three dozen groups of targeted therapies showed high comparability between screens. Modernization of NCI60, and closer integration with other large-scale pharmacogenomic screens and molecular feature sets, will help this public screening service remain pertinent for cancer drug discovery efforts for years to come. See related article by Kunkel et al., p. 2403.


Subject(s)
Antineoplastic Agents , Drug Discovery , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays/methods , Cell Line, Tumor , Antineoplastic Agents/pharmacology , National Cancer Institute (U.S.) , Phenotype , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/genetics , United States , Cell Proliferation/drug effects
2.
Anticancer Res ; 44(8): 3481-3491, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060051

ABSTRACT

BACKGROUND/AIM: Precision medicine aims to revolutionize healthcare by tailoring treatment regimens. This study aimed to integrate comprehensive tumor genomic profiling (CTGP) by targeted-gene panel sequencing and drug screening by circulating tumor cell-derived organoids (CTOs) into clinical practice for the treatment of gastrointestinal (GI) cancers. PATIENTS AND METHODS: Nine patients with various GI cancers underwent CTGP and CTO drug sensitivity testing. CTGP results guided targeted therapy and immunotherapy, while CTO drug sensitivity predicted response to chemotherapy and targeted agents. The drug recommendations from two platforms were correlated with the treatment response to the suggested medications retrospectively. RESULTS: Five patients received therapies aligned with CTGP, including HER2-targeted treatment, immunotherapy, and BRAF/MEK dual inhibition, showing positive responses. CTO drug sensitivity predicted progression under regorafenib (low potential benefit) and good response to chemotherapy with high potential benefit. The combination of CTGP and CTO drug sensitivity may exhibit significant correlation with clinical outcomes during treatment with candidate drugs, demonstrating a sensitivity of 79% and an accuracy of 75%. This encompasses various treatment modalities, such as chemotherapy, targeted therapy, and immunotherapy. CONCLUSION: The present investigation elucidated the integration of CTGP and CTO drug sensitivity screening into clinical practice in a complementary manner, showcasing a significant correlation between treatment response and testing outcomes. Additional prospective evaluation of these two testing modalities in a large cohort is warranted to confirm whether the inclusion of CTO drug sensitivity screening confers enhanced survival benefits compared to utilizing CTGP alone.


Subject(s)
Gastrointestinal Neoplasms , Neoplastic Cells, Circulating , Organoids , Humans , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/blood , Female , Male , Organoids/pathology , Organoids/drug effects , Middle Aged , Aged , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/drug effects , Precision Medicine/methods , Genomics/methods , Retrospective Studies , Gene Expression Profiling/methods , Biomarkers, Tumor/genetics , Molecular Targeted Therapy/methods , Adult , Drug Screening Assays, Antitumor/methods
3.
Cancer Lett ; 598: 217122, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39029781

ABSTRACT

Tumor organoids have emerged as a crucial preclinical model for multiple cancer research. Their high establishment rates, stability, and ability to replicate key biological features of original tumor cells in vivo render them invaluable for exploring tumor molecular mechanisms, discovering potential anti-tumor drugs, and predicting clinical drug efficacy. Here, we review the establishment of tumor organoid models and provide an extensive overview of organoid culturing strategies. We also emphasize the significance of integrating cellular components of the tumor microenvironment and physicochemical factors in the organoid culturing system, highlighting the importance of artificial intelligence technology in advancing organoid construction. Moreover, we summarize recent advancements in utilizing organoid systems for novel anti-cancer drug screening and discuss promising trends for enhancing advanced organoids in next-generation disease modeling.


Subject(s)
Neoplasms , Organoids , Tumor Microenvironment , Organoids/drug effects , Organoids/pathology , Humans , Neoplasms/pathology , Neoplasms/drug therapy , Animals , Artificial Intelligence , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology
4.
Biochem Biophys Res Commun ; 730: 150369, 2024 Oct 20.
Article in English | MEDLINE | ID: mdl-39013264

ABSTRACT

Gemcitabine is a nucleoside analog widely used as an anticancer agent against several types of cancer. Although gemcitabine sometimes shows excellent effectiveness, cancer cells are often poorly responsive to or resistant to the drug. Recently, specific strains or dysbiosis of the human microbiome were correlated with drug reactivity and resistance acquisition. Therefore, we aimed to identify antibiotic compounds that can modulate the microbiome to enhance the responsiveness to gemcitabine. To achieve this, we confirmed the gemcitabine responsiveness based on public data and conducted drug screening on a set of 250 antibiotics compounds. Subsequently, we performed experiments to investigate whether the selected compounds could enhance the responsiveness to gemcitabine. First, we grouped a total of seven tumor cell lines into resistant and sensitive group based on the IC50 value (1 µM) of gemcitabine obtained from the public data. Second, we performed high-throughput screening with compound treatments, identifying seven compounds from the resistant group and five from the sensitive group based on dose dependency. Finally, the combination of the selected compound, puromycin dihydrochloride, with gemcitabine in gemcitabine-resistant cell lines resulted in extensive cell death and a significant increase in cytotoxic efficacy. Additionally, mRNA levels associated with cell viability and stemness were reduced. Through this study, we screened antibiotics to further improve the efficacy of existing anticancer drugs and overcome resistance. By combining existing anticancer agents and antibiotic substances, we hope to establish various drug combination therapies and ultimately improve cancer treatment efficacy.


Subject(s)
Anti-Bacterial Agents , Deoxycytidine , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Gemcitabine , High-Throughput Screening Assays , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , High-Throughput Screening Assays/methods , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Anti-Bacterial Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Puromycin/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Drug Synergism , Antineoplastic Agents/pharmacology , Small Molecule Libraries/pharmacology
5.
Trends Pharmacol Sci ; 45(8): 723-738, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39013672

ABSTRACT

Before molecular pathways in cancer were known to a depth that could predict targets, drug development relied on phenotypic screening, where the effectiveness of candidate chemicals is judged from functional readouts without considering the mechanisms of action. The unraveling of tumor-specific pathways has brought targets for molecularly driven drug discovery, but precedents in the field have shown that awareness of pathways does not necessarily predict therapeutic efficacy, and many cancers still lack druggable targets. Phenotypic screening therefore retains a niche in drug development where a targeted approach is not informative. We analyze the unique advantages of phenotypic screens, and how technological advances have improved their discovery power. Notable advances include the use of larger biological panels and refined protocols that address the disease-relevance and increase data content with imaging and omic approaches.


Subject(s)
Antineoplastic Agents , Drug Discovery , Neoplasms , Phenotype , Humans , Drug Discovery/methods , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Animals , Drug Screening Assays, Antitumor/methods
6.
Methods Mol Biol ; 2823: 95-108, 2024.
Article in English | MEDLINE | ID: mdl-39052216

ABSTRACT

Three-dimensional (3D) cell culture creates a more physiologically relevant environment for enhanced drug screening capabilities using microcarriers. An automated 3D system that integrates robotic manipulators, liquid handling systems, sensors, and environment control systems has the capacity to handle multiple samples in parallel, perform repetitive tasks, and provide real-time monitoring and analysis. This chapter describes a potential 3D cell culture drug screening model by combining renal proximal tubule cells as a representative normal cell line with cancer cell lines. This combination is subjected to drug screening to evaluate the drug's efficacy in suppressing cancer cells while minimizing impact on normal cells with the added benefit of having the ability to separate the two cell types by magnetic isolation for high content screens including mass spectrometry-based proteomics. This study presents advancements in 3D cell culture techniques, emphasizing the importance of automation and the potential of microcarriers in drug screening and disease modeling.


Subject(s)
Cell Culture Techniques, Three Dimensional , Humans , Cell Culture Techniques, Three Dimensional/methods , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Drug Screening Assays, Antitumor/methods , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Cell Culture Techniques/methods , Antineoplastic Agents/pharmacology , Automation , Automation, Laboratory/methods , Neoplasms/pathology , Neoplasms/drug therapy
7.
Cell Rep Med ; 5(7): 101627, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38964315

ABSTRACT

The efficacy of chemotherapy varies significantly among patients with gastric cancer (GC), and there is currently no effective strategy to predict chemotherapeutic outcomes. In this study, we successfully establish 57 GC patient-derived organoids (PDOs) from 73 patients with GC (78%). These organoids retain histological characteristics of their corresponding primary GC tissues. GC PDOs show varied responses to different chemotherapeutics. Through RNA sequencing, the upregulation of tumor suppression genes/pathways is identified in 5-fluorouracil (FU)- or oxaliplatin-sensitive organoids, whereas genes/pathways associated with proliferation and invasion are enriched in chemotherapy-resistant organoids. Gene expression biomarker panels, which could distinguish sensitive and resistant patients to 5-FU and oxaliplatin (area under the dose-response curve [AUC] >0.8), are identified. Moreover, the drug-response results in PDOs are validated in patient-derived organoids-based xenograft (PDOX) mice and are consistent with the actual clinical response in 91.7% (11/12) of patients with GC. Assessing chemosensitivity in PDOs can be utilized as a valuable tool for screening chemotherapeutic drugs in patients with GC.


Subject(s)
Fluorouracil , Organoids , Precision Medicine , Stomach Neoplasms , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Humans , Organoids/drug effects , Organoids/pathology , Organoids/metabolism , Animals , Precision Medicine/methods , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Male , Female , Xenograft Model Antitumor Assays , Drug Screening Assays, Antitumor/methods , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Middle Aged , Gene Expression Regulation, Neoplastic/drug effects , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Aged , Clinical Relevance
8.
Cancer Med ; 13(12): e7432, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923304

ABSTRACT

INTRODUCTION: Kidney cancer is a common urological malignancy worldwide with an increasing incidence in recent years. Among all subtypes, renal cell carcinoma (RCC) represents the most predominant malignancy in kidney. Clinicians faced a major challenge to select the most effective and suitable treatment regime for patients from a wide range of modalities, despite improved understanding and diagnosis of RCC. OBJECTIVE: Recently, organoid culture gained more interest as the 3D model is shown to be highly patient specific which is hypothetically beneficial to the investigation of precision medicine. Nonetheless, the development and application of organotypic culture in RCC is still immature, therefore, the primary objective of this study was to establish an organoid model for RCC. MATERIALS AND METHODS: Patients diagnosed with renal tumor and underwent surgical intervention were recruited. RCC specimen was collected and derived into organoids. Derived organoids were validated by histological examminations, sequencing and xenograft. Drug response of organoids were compared with resistance cell line and patients' clinical outcomes. RESULTS: Our results demonstrated that organoids could be successfully derived from renal tumor and they exhibited high concordance in terms of immunoexpressional patterns. Sequencing results also depicted concordant mutations of driver genes in both organoids and parental tumor tissues. Critical and novel growth factors were discovered during the establishment of organoid model. Besides, organoids derived from renal tumor exhibited tumorigenic properties in vivo. In addition, organoids recapitulated patient's in vivo drug resistance and served as a platform to predict responsiveness of other therapeutic agents. CONCLUSION: Our RCC organoid model recaptiluated histological and genetic features observed in primary tumors. It also served as a potential platform in drug screening for RCC patients, though future studies are necessary before translating the outcomes into clinical practices.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Organoids , Humans , Organoids/drug effects , Organoids/pathology , Kidney Neoplasms/pathology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/genetics , Animals , Mice , Female , Male , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Xenograft Model Antitumor Assays , Middle Aged , Cell Line, Tumor , Aged , Mutation
9.
In Vivo ; 38(4): 1579-1593, 2024.
Article in English | MEDLINE | ID: mdl-38936891

ABSTRACT

BACKGROUND/AIM: Melanoma, a variant of skin cancer, presents the highest mortality rates among all skin cancers. Despite advancements in targeted therapies, immunotherapies, and tissue culture techniques, the absence of an effective early treatment model remains a challenge. This study investigated the impact of dabrafenib on both 2D and 3D cell culture models with distinct molecular profiles. MATERIALS AND METHODS: We developed a high-throughput workflow enabling drug screening on spheroids. Our approach involved cultivating 2D and 3D cultures derived from normal melanocytes and metastatic melanoma cells, treating them with dabrafenib and conducting viability, aggregation, migration, cell cycle, and apoptosis assays. RESULTS: Dabrafenib exerted multifaceted influences, particularly on migration at concentrations of 10 and 25 µM. It induced a decrease in cell viability, impeded cellular adhesion to the matrix, inhibited cellular aggregation and spheroid formation, arrested the cell cycle in the G1 phase, and induced apoptosis. CONCLUSION: These results confirm the therapeutic potential of dabrafenib in treating melanoma with the BRAF V600E mutation and that 3D models are validated models to study the potential of new molecules for therapeutic purposes. Furthermore, our study underscores the relevance of 3D models in simulating physiological in vivo microenvironments, providing insights into varied treatment responses between normal and tumor cells.


Subject(s)
Apoptosis , Cell Movement , Cell Survival , Imidazoles , Melanoma , Oximes , Proto-Oncogene Proteins B-raf , Spheroids, Cellular , Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Culture Techniques , Cell Culture Techniques, Three Dimensional/methods , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor/methods , Imidazoles/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Melanoma/genetics , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Spheroids, Cellular/drug effects
10.
Theranostics ; 14(8): 3300-3316, 2024.
Article in English | MEDLINE | ID: mdl-38855182

ABSTRACT

Patient-derived organoids (PDOs) have emerged as a promising platform for clinical and translational studies. A strong correlation exists between clinical outcomes and the use of PDOs to predict the efficacy of chemotherapy and/or radiotherapy. To standardize interpretation and enhance scientific communication in the field of cancer precision medicine, we revisit the concept of PDO-based drug sensitivity testing (DST). We present an expert consensus-driven approach for medication selection aimed at predicting patient responses. To further standardize PDO-based DST, we propose guidelines for clarification and characterization. Additionally, we identify several major challenges in clinical prediction when utilizing PDOs.


Subject(s)
Antineoplastic Agents , Consensus , Drug Development , Neoplasms , Organoids , Precision Medicine , Organoids/drug effects , Humans , Precision Medicine/methods , Neoplasms/drug therapy , Drug Development/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods
11.
Cancer Med ; 13(11): e7318, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872378

ABSTRACT

BACKGROUND: In vitro drug screening that is more translatable to the in vivo tumor environment can reduce both time and cost of cancer drug development. Here we address some of the shortcomings in screening and show how treatment with 5-fluorouracil (5-FU) in 2D and 3D culture models of colorectal cancer (CRC) and pancreatic ductal adenocarcinomas (PDAC) give different responses regarding growth inhibition. METHODS: The sensitivity of the cell lines at clinically relevant 5-FU concentrations was monitored over 4 days of treatment in both 2D and 3D cultures for CRC (SW948 and HCT116) and PDAC (Panc-1 and MIA-Pa-Ca-2) cell lines. The 3D cultures were maintained beyond this point to enable a second treatment cycle at Day 14, following the timeline of a standard clinical 5-FU regimen. RESULTS: Evaluation after one cycle did not reveal significant growth inhibition in any of the CRC or PDAC 2D models. By the end of the second cycle of treatment the CRC spheroids reached 50% inhibition at clinically achievable concentrations in the 3D model, but not in the 2D model. The PDAC models were not sensitive to clinical doses even after two cycles. High content viability metrics point to even lower response in the resistant PDAC models. CONCLUSION: This study reveals the limitations of testing drugs in 2D cancer models and short exposure in 3D models, and the importance of using appropriate growth inhibition analysis. We found that screening with longer exposure and several cycles of treatment in 3D models suggests a more reliable way to assess drug sensitivity.


Subject(s)
Cell Proliferation , Cell Survival , Fluorouracil , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Fluorouracil/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Spheroids, Cellular/drug effects , Drug Screening Assays, Antitumor/methods , Cell Culture Techniques , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm
12.
J Nanobiotechnology ; 22(1): 344, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890730

ABSTRACT

BACKGROUND: Functional drug testing (FDT) with patient-derived tumor cells in microfluidic devices is gaining popularity. However, the majority of previously reported microfluidic devices for FDT were limited by at least one of these factors: lengthy fabrication procedures, absence of tumor progenitor cells, lack of clinical correlation, and mono-drug therapy testing. Furthermore, personalized microfluidic models based on spheroids derived from oral cancer patients remain to be thoroughly validated. Overcoming the limitations, we develop 3D printed mold-based, dynamic, and personalized oral stem-like spheroids-on-a-chip, featuring unique serpentine loops and flat-bottom microwells arrangement. RESULTS: This unique arrangement enables the screening of seven combinations of three drugs on chemoresistive cancer stem-like cells. Oral cancer patients-derived stem-like spheroids (CD 44+) remains highly viable (> 90%) for 5 days. Treatment with a well-known oral cancer chemotherapy regimen (paclitaxel, 5 fluorouracil, and cisplatin) at clinically relevant dosages results in heterogeneous drug responses in spheroids. These spheroids are derived from three oral cancer patients, each diagnosed with either well-differentiated or moderately-differentiated squamous cell carcinoma. Oral spheroids exhibit dissimilar morphology, size, and oral tumor-relevant oxygen levels (< 5% O2). These features correlate with the drug responses and clinical diagnosis from each patient's histopathological report. CONCLUSIONS: Overall, we demonstrate the influence of tumor differentiation status on treatment responses, which has been rarely carried out in the previous reports. To the best of our knowledge, this is the first report demonstrating extensive work on development of microfluidic based oral cancer spheroid model for personalized combinatorial drug screening. Furthermore, the obtained clinical correlation of drug screening data represents a significant advancement over previously reported personalized spheroid-based microfluidic devices. Finally, the maintenance of patient-derived spheroids with high viability under oral cancer relevant oxygen levels of less than 5% O2 is a more realistic representation of solid tumor microenvironment in our developed device.


Subject(s)
Antineoplastic Agents , Drug Screening Assays, Antitumor , Lab-On-A-Chip Devices , Mouth Neoplasms , Neoplastic Stem Cells , Precision Medicine , Spheroids, Cellular , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Spheroids, Cellular/drug effects , Neoplastic Stem Cells/drug effects , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology , Precision Medicine/methods , Printing, Three-Dimensional , Fluorouracil/pharmacology , Paclitaxel/pharmacology
13.
Cancer Res ; 84(15): 2403-2416, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38861359

ABSTRACT

The NCI60 human tumor cell line screen has been in operation as a service to the cancer research community for more than 30 years. The screen operated with 96-well plates, a 2-day exposure period to test agents, and following cell fixation, a visible absorbance endpoint by the protein-staining dye sulforhodamine B. In this study, we describe the next phase of this important cancer research tool, the HTS384 NCI60 screen. Although the cell lines remain the same, the updated screen is performed with 384-well plates, a 3-day exposure period to test agents, and a luminescent endpoint to measure cell viability based upon cellular ATP content. In this study, a library of 1,003 FDA-approved and investigational small-molecule anticancer agents was screened by the two NCI60 assays. The datasets were compared with a focus on targeted agents with at least six representatives in the library. For many agents, including inhibitors of EGFR, BRAF, MEK, ERK, and PI3K, the patterns of GI50 values were very similar between the screens with strong correlations between those patterns within the dataset from each screen. However, for some groups of targeted agents, including mTOR, BET bromodomain, and NAMPRTase inhibitors, there were limited or no correlations between the two datasets, although the patterns of GI50 values and correlations between those patterns within each dataset were apparent. Beginning in January 2024, the HTS384 NCI60 screen became the free screening service of the NCI to facilitate drug discovery by the cancer research community. Significance: The new NCI60 cell line screen HTS384 shows robust patterns of response to oncology agents and substantial overlap with the classic screen, providing an updated tool for studying therapeutic agents. See related commentary by Colombo and Corsello, p. 2397.


Subject(s)
Antineoplastic Agents , Drug Screening Assays, Antitumor , Humans , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays/methods , Small Molecule Libraries/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Cell Survival/drug effects
14.
STAR Protoc ; 5(2): 103058, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38748881

ABSTRACT

Three-dimensional (3D) models play an increasingly important role in preclinical drug testing as they faithfully mimic interactions between cancer cells and the tumor microenvironment (TME). Here, we present a protocol for generating scaffold-free 3D multicomponent human melanoma spheroids. We describe steps for characterizing models using live-cell imaging and histology, followed by drug testing and assessment of cell death through various techniques such as imaging, luminescence-based assays, and flow cytometry. Finally, we demonstrate the models' adaptability for co-cultures with immune cells.


Subject(s)
Melanoma , Spheroids, Cellular , Humans , Spheroids, Cellular/pathology , Spheroids, Cellular/metabolism , Melanoma/pathology , Melanoma/metabolism , Drug Evaluation, Preclinical/methods , Tumor Microenvironment , Coculture Techniques/methods , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Cell Culture Techniques/methods
15.
STAR Protoc ; 5(2): 103090, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38809757

ABSTRACT

Drug sensitivity testing of patient-derived tumor organoids (PDTOs) is a promising tool for personalizing cancer treatment. Here, we present a protocol for generation of and high-throughput drug testing with PDTOs. We describe detailed steps for PDTO establishment from colorectal cancer tissues, preparation of PDTOs for high-throughput drug testing, and quantification of drug testing results using image analysis. This protocol provides a standardized workflow for PDTO testing of standard-of-care therapies, along with exploring the activity of new agents, for translational research. For complete details on the use and execution of this protocol, please refer to Tan et al.1.


Subject(s)
Colorectal Neoplasms , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Organoids , Organoids/drug effects , Organoids/pathology , Organoids/metabolism , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , High-Throughput Screening Assays/methods , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
16.
J Cell Mol Med ; 28(9): e18374, 2024 May.
Article in English | MEDLINE | ID: mdl-38722288

ABSTRACT

The majority of advanced breast cancers exhibit strong aggressiveness, heterogeneity, and drug resistance, and currently, the lack of effective treatment strategies is one of the main challenges that cancer research must face. Therefore, developing a feasible preclinical model to explore tailored treatments for refractory breast cancer is urgently needed. We established organoid biobanks from 17 patients with breast cancer and characterized them by immunohistochemistry (IHC) and next generation sequencing (NGS). In addition, we in the first combination of patient-derived organoids (PDOs) with mini-patient-derived xenografts (Mini-PDXs) for the rapid and precise screening of drug sensitivity. We confirmed that breast cancer organoids are a high-fidelity three-dimension (3D) model in vitro that recapitulates the original tumour's histological and genetic features. In addition, for a heavily pretreated patient with advanced drug-resistant breast cancer, we combined PDO and Mini-PDX models to identify potentially effective combinations of therapeutic agents for this patient who were alpelisib + fulvestrant. In the drug sensitivity experiment of organoids, we observed changes in the PI3K/AKT/mTOR signalling axis and oestrogen receptor (ER) protein expression levels, which further verified the reliability of the screening results. Our study demonstrates that the PDO combined with mini-PDX model offers a rapid and precise drug screening platform that holds promise for personalized medicine, improving patient outcomes and addressing the urgent need for effective therapies in advanced breast cancer.


Subject(s)
Breast Neoplasms , Organoids , Precision Medicine , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Organoids/drug effects , Organoids/pathology , Organoids/metabolism , Precision Medicine/methods , Animals , Xenograft Model Antitumor Assays , Mice , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor/methods , Middle Aged
17.
Nat Commun ; 15(1): 4363, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778087

ABSTRACT

Drug screening based on in-vitro primary tumor cell culture has demonstrated potential in personalized cancer diagnosis. However, the limited number of tumor cells, especially from patients with early stage cancer, has hindered the widespread application of this technique. Hence, we developed a digital microfluidic system for drug screening using primary tumor cells and established a working protocol for precision medicine. Smart control logic was developed to increase the throughput of the system and decrease its footprint to parallelly screen three drugs on a 4 × 4 cm2 chip in a device measuring 23 × 16 × 3.5 cm3. We validated this method in an MDA-MB-231 breast cancer xenograft mouse model and liver cancer specimens from patients, demonstrating tumor suppression in mice/patients treated with drugs that were screened to be effective on individual primary tumor cells. Mice treated with drugs screened on-chip as ineffective exhibited similar results to those in the control groups. The effective drug identified through on-chip screening demonstrated consistency with the absence of mutations in their related genes determined via exome sequencing of individual tumors, further validating this protocol. Therefore, this technique and system may promote advances in precision medicine for cancer treatment and, eventually, for any disease.


Subject(s)
Breast Neoplasms , Microfluidics , Precision Medicine , Xenograft Model Antitumor Assays , Precision Medicine/methods , Humans , Animals , Mice , Female , Cell Line, Tumor , Microfluidics/methods , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
18.
Acta Biomater ; 183: 111-129, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38801868

ABSTRACT

The development of high-throughput anticancer drug screening methods using patient-derived cancer cell (PDC) lines that maintain their original characteristics in an in vitro three-dimensional (3D) culture system poses a significant challenge to achieving personalized cancer medicine. Because stromal tissue plays a critical role in the composition and maintenance of the cancer microenvironment, in vitro 3D-culture using reconstructed stromal tissues has attracted considerable attention. Here, a simple and unique in vitro 3D-culture method using heparin and collagen together with fibroblasts and endothelial cells to fabricate vascularized 3D-stromal tissues for in vitro culture of PDCs is reported. Whereas co-treatment with bevacizumab, a monoclonal antibody against vascular endothelial growth factor, and 5-fluorouracil significantly reduced the survival rate of 3D-cultured PDCs to 30%, separate addition of each drug did not induce comparable strong cytotoxicity, suggesting the possibility of evaluating the combined effect of anticancer drugs and angiogenesis inhibitors. Surprisingly, drug evaluation using eight PDC lines with the 3D-culture method resulted in a drug efficacy concordance rate of 75% with clinical outcomes. The model is expected to be applicable to in vitro throughput drug screening for the development of personalized cancer medicine. STATEMENT OF SIGNIFICANCE: To replicate the cancer microenvironment, we constructed a cancer-stromal tissue model in which cancer cells are placed above and inside stromal tissue with vascular network structures derived from vascular endothelial cells in fibroblast tissue using CAViTs method. Using this method, we were able to reproduce the invasion and metastasis processes of cancer cells observed in vivo. Using patient-derived cancer cells, we assessed the possibility of evaluating the combined effect with an angiogenesis inhibitor. Further, primary cancer cells also grew on the stromal tissues with the normal medium. These data suggest that the model may be useful for new in vitro drug screening and personalized cancer medicine.


Subject(s)
Antineoplastic Agents , Drug Screening Assays, Antitumor , Humans , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Cell Line, Tumor , Stromal Cells/drug effects , Stromal Cells/cytology , Stromal Cells/metabolism , Cell Culture Techniques, Three Dimensional/methods , High-Throughput Screening Assays/methods , Tumor Microenvironment/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/cytology , Coculture Techniques
19.
Adv Healthc Mater ; 13(20): e2400203, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38774999

ABSTRACT

The limited recapitulation of critical cancer features in 2D cultures causes poor translatability of preclinical results from in vitro assays to in vivo tumor models. This contributes to slow drug development with a low success rate. 3D cultures better recapitulate the tumor microenvironment, enabling more accurate predictions when screening drug candidates and improving the development of chemotherapeutics. Platinum (Pt) (IV) compounds are promising prodrugs designed to reduce the severe systemic toxicity of widely used Food and Drug Administration (FDA)-approved Pt(II) drugs such as cisplatin. Here, this work presents spatiotemporal evaluations in 3D colorectal cancer (CRC) spheroids of mitochondria-targeting Pt(IV) complexes. CRC spheroids provide a greater pathophysiological recapitulation of in vivo tumors than 2D cultures by a marked upregulation of the ABCG2 chemoresistance marker expression. Furthermore, new 3D-staining protocols are introduced to evaluate the real-time decrease in mitochondria membrane potential (ΔΨ) in CRC spheroids, and a Pt-sensing dye to quantify the Pt mitochondrial accumulation. Finally, this work demonstrates a correlation between in vitro results and the efficacy of the compounds in vivo. Overall, the CRC spheroids represent a fast and cost-effective model to assess the behavior of Pt compounds in vitro and predict their translational potential in CRC treatment.


Subject(s)
Colorectal Neoplasms , Spheroids, Cellular , Humans , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor/methods , Cell Line, Tumor , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Membrane Potential, Mitochondrial/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Fluorescence
20.
Nat Commun ; 15(1): 3599, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678014

ABSTRACT

Targeting the supportive tumor microenvironment (TME) is an approach of high interest in cancer drug development. However, assessing TME-targeted drug candidates presents a unique set of challenges. We develop a comprehensive screening platform that allows monitoring, quantifying, and ranking drug-induced effects in self-organizing, vascularized tumor spheroids (VTSs). The confrontation of four human-derived cell populations makes it possible to recreate and study complex changes in TME composition and cell-cell interaction. The platform is modular and adaptable for tumor entity or genetic manipulation. Treatment effects are recorded by light sheet fluorescence microscopy and translated by an advanced image analysis routine in processable multi-parametric datasets. The system proved to be robust, with strong interassay reliability. We demonstrate the platform's utility for evaluating TME-targeted antifibrotic and antiangiogenic drugs side-by-side. The platform's output enabled the differential evaluation of even closely related drug candidates according to projected therapeutic needs.


Subject(s)
Breast Neoplasms , Microscopy, Fluorescence , Spheroids, Cellular , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Microscopy, Fluorescence/methods , Female , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL