1
|
Zhu D, Brückner D, Sosniok M, Skiba M, Feliu N, Gallego M, Liu Y, Schulz F, Falkenberg G, Parak WJ, Sanchez-Cano C. Size-dependent penetration depth of colloidal nanoparticles into cell spheroids. Adv Drug Deliv Rev 2025; 222:115593. [PMID: 40339992 DOI: 10.1016/j.addr.2025.115593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
The penetration of nanoparticle (NP)-based drugs into tissue is essential for their use as nanomedicines. Systematic studies about how different NP properties, such as size, influence NP penetration are helpful for the development of NP-based drugs. An overview of how NPs of different sizes may penetrate three-dimensional cell spheroids is given. In particular different techniques for experimental analysis are compared, including mass spectrometry, flow cytometry, optical fluorescence microscopy, X-ray fluorescence microscopy, and transmission electron microscopy. An experimental data set is supplemented exclusively made for this review, in which the results of different techniques are visualized. Limitations of the analysis techniques for different types of NPs, including carbon-based materials, are discussed.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany; Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121 Zhejiang, China
| | - Dennis Brückner
- Deutsches Elektronen-Synchrotron DESY, Photon Science, 22607 Hamburg, Germany
| | - Martin Sosniok
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany; Zentrum für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Marvin Skiba
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Neus Feliu
- Zentrum für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Marta Gallego
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE) Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Yang Liu
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Florian Schulz
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron DESY, Photon Science, 22607 Hamburg, Germany.
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany.
| | - Carlos Sanchez-Cano
- Donostia International Physics Center, 20018 Donostia-San Sebastian, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, 20018 Donostia-San Sebastian, Spain.
| |
Collapse
|
2
|
Malakpour-Permlid A, Rodriguez MM, Untracht GR, Andersen PE, Oredsson S, Boisen A, Zór K. High-throughput non-homogenous 3D polycaprolactone scaffold for cancer cell and cancer-associated fibroblast mini-tumors to evaluate drug treatment response. Toxicol Rep 2025; 14:101863. [PMID: 39758801 PMCID: PMC11699757 DOI: 10.1016/j.toxrep.2024.101863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
High-throughput screening (HTS) three-dimensional (3D) tumor models are a promising approach for cancer drug discovery, as they more accurately replicate in vivo cell behavior than two-dimensional (2D) models. However, assessing and comparing current 3D models for drug efficacy remains essential, given the significant influence of cellular conditions on treatment response. To develop in vivo mimicking 3D models, we evaluated two HTS 3D models established in 96-well plates with 3D polycaprolactone (PCL) scaffolds fabricated using two distinct methods, resulting in scaffolds with either homogenous or non-homogenous fiber networks. These models, based on human HeLa cervical cancer cells and cancer-associated fibroblasts (CAFs) cultured as mono- or co-cultures within the 3D scaffolds, revealed that anticancer drug paclitaxel (PTX) exhibited consistently higher inhibitory concentration 50 (IC50) in 3D (≥ 1000 nM) compared to 2D (≥ 100 nM), indicating reduced toxicity on cells cultured in 3D. Interestingly, the toxicity of PTX was significantly lower on mini-tumors in non-homogenous 3D (IC50: 600 or 1000 nM) than in homogenous 3D cultures (IC50 exceeding 1000 nM). Microscopic studies revealed that the non-homogenous scaffolds closely resemble the tumor collagen network than their homogeneous counterpart. Both 3D scaffolds offer optimal pore size, facilitating efficient cell infiltration into the depth of 58.1 ± 1.2 µm (homogenous) and 86.4 ± 9.8 µm (non-homogenous) within 3D cultures. Cells cultured in the 3D non-homogenous systems exhibited drug treatment responses closer to in vivo conditions, highlighting the role of scaffold structure and design on cellular response to drug treatment. The PCL-based 3D models provide a robust, tunable, and efficient approach for the HTS of anti-cancer drugs compared to conventional 2D systems.
Collapse
Affiliation(s)
- Atena Malakpour-Permlid
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Manuel Marcos Rodriguez
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Gavrielle R. Untracht
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Peter E. Andersen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | | | - Anja Boisen
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Kinga Zór
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- BioInnovation Institute Foundation, Copenhagen N 2200, Denmark
- Innovation Acta S.r.l., Siena, Via delle 1-53100, Italy
| |
Collapse
|
3
|
Elsayad KA, Elmasry GF, Mahmoud ST, Awadallah FM, Giovannuzzi S, Supuran CT. Development of novel amino-benzenesulfonamide derivatives and their analogues as carbonic anhydrase inhibitors: Design, synthesis, anticancer activity assessment, and pharmacokinetic studies using UPLC-MS/MS. Bioorg Chem 2025; 159:108335. [PMID: 40086186 DOI: 10.1016/j.bioorg.2025.108335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
The present study outlines the design and synthesis of dual-tail analogues of SLC-0111 as carbonic anhydrase inhibitors (CAIs) targeting tumor isoforms IX and XII 4a-h and 5a-h, along with pharmacokinetic studies. The synthesized compounds were evaluated for their inhibitory activity against four carbonic anhydrase isoforms (hCA I, II, IX, and XII), revealing potent activity, particularly against hCA IX and XII. Notably, compounds 4b, 5a, and 5b demonstrated strong inhibition of hCA IX with Ki values of 20.4, 12.9, and 18.2 nM, respectively, compared to acetazolamide (AAZ), which has a Ki of 25 nM. Additionally, compounds 5a, 5b, 5c, and 5d showed selective inhibition of hCA XII, with Ki values of 26.6, 8.7, 17.2, and 10.9 nM, respectively, relative to AAZ (Ki = 5.7 nM). Moreover, both series were tested for their anti-proliferative activity following the US-NCI protocol against a panel of more than fifty cancer cell lines. Compound 5h met the activity criteria and was automatically scheduled for further evaluation at five concentrations with 10-fold dilutions, revealing high toxicity toward leukemia and lower toxicity against melanoma. In addition, the MTT cytotoxicity assay was performed on 5f, 5d and acetazolamide using WI-38 cells. Furthermore, an in vivo pharmacokinetic study was conducted using UPLC-MS/MS on the most potent derivative, 5d, demonstrating a comparable pharmacokinetic profile compared to the reference drug acetazolamide. Furthermore, molecular docking prediction studies were conducted for the most active compounds, 5d and 5h, to elucidate their interactions with the active site hot spots of the CA isoform.
Collapse
Affiliation(s)
- Khaled A Elsayad
- Pharmacy Department, Cairo University Hospitals, Cairo University, Cairo 11662, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt.
| | - Ghada F Elmasry
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt
| | - Sally T Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt; Pharmaceutical Chemistry Department, School of Pharmacy, New Giza University, New Giza, km 22 Cairo- Alexandria Desert Road, Cairo, Egypt.
| | - Fadi M Awadallah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt
| | - Simone Giovannuzzi
- Department NEUROFARBA - Pharmaceutical and Nutraceutical section, University of Firenze, Università Degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Claudiu T Supuran
- Department NEUROFARBA - Pharmaceutical and Nutraceutical section, University of Firenze, Università Degli Studi di Firenze, Sesto Fiorentino, Italy
| |
Collapse
|
4
|
Maity S, Jewell C, Yilgor C, Kawakita S, Sharma S, Gomez A, Mecwan M, Falcone N, Ermis M, Monirizad M, Kouchehbaghi NH, Zehtabi F, Khorsandi D, Dokmeci MR, Moniz-Garcia D, Quiñones-Hinojosa A, Khademhosseini A, Jucaud V. Deciphering pericyte-induced temozolomide resistance in glioblastoma with a 3D microphysiological system mimicking the biomechanical properties of brain tissue. Acta Biomater 2025:S1742-7061(25)00363-0. [PMID: 40383349 DOI: 10.1016/j.actbio.2025.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/29/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Glioblastoma (GBM) is a highly aggressive malignancy with a poor prognosis and frequent resistance to temozolomide (TMZ), the standard-of-care chemotherapy. The complex mechanisms underlying GBM chemoresistance, particularly the role of pericytes, remain poorly understood due to the lack of physiologically relevant in vitro models replicating the complex tumor microenvironment (TME). Here, we present a biomimetic 3D GBM microphysiological system that replicates the biomechanical properties of brain tissue (G'∼800Pa, G"∼100Pa) and enables the study of pericyte-mediated TMZ resistance. GBM spheroids (U87, LN229, PDM140) were cultured alone or co-cultured with pericytes in a composite hydrogel for 14 days and remained viable and proliferative. In response to TMZ, PDM140 was the most sensitive (IC50=73μM), followed by LN229 (IC50=278μM) and U87 (IC50=446μM). Co-culture with pericytes significantly increased GBM spheroid viability by 22.7% (PDM140), 32.5% (LN229), and 22.1% (U87), confirming pericyte-induced TMZ resistance. Notably, pericytes exhibited a 160-fold upregulation of C-C motif chemokine ligand 5 (CCL5) upon TMZ treatment, implicating the CCL5-mediated pathway in chemoresistance. This innovative brain-mimicking 3D GBM model provides a physiologically relevant platform for studying tumor-pericyte interactions and testing therapeutic strategies targeting CCL5-mediated resistance mechanisms in GBM. STATEMENT OF SIGNIFICANCE: We developed a multicellular 3D glioblastoma microphysiological system mimicking the physicochemical properties of brain tissues and pericyte-mediated TMZ resistance that can be used to screen for standard-of-care chemotherapy. This advanced hydrogel-based platform demonstrated the critical role of the glioblastoma tumor microenvironment in modulating chemotherapy sensitivity, particularly the pericyte-induced CCL5-CCR5 paracrine axis that can lead to the identification of therapeutic targets within the CCL5-CCR5 pathway toward more effective treatments disrupting these resistance mechanisms. Overall, the proposed 3D glioblastoma microphysiological system can transform drug screening and personalized treatment for GBM by offering ethical and cost-effective alternatives to animal testing and more effective drug screening and discovery efforts, ultimately improving GBM patient outcomes.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA.; Department of Orthopedic Surgery, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Saurabh Sharma
- Department of Surgery, Stanford School of Medicine, Stanford University Medical Center, CA 94305, USA
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | | | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA
| | | | - Diogo Moniz-Garcia
- Mayo Clinic Florida, Department of Neurosurgery, Jacksonville, FL, 32224, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA..
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Woodland Hills, CA, 91367, USA..
| |
Collapse
|
5
|
Madsen NH, Nielsen BS, Skandorff I, Rodriguez-Pardo C, Hadrup SR, Ormhøj M, Holmstrøm K, Larsen J, Gad M. Novel approaches to 3D cancer heterospheroid culture and assay development for immunotherapy screening. Exp Cell Res 2025; 449:114604. [PMID: 40379236 DOI: 10.1016/j.yexcr.2025.114604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Advanced 3D heterospheroids, composed of cancer, fibroblast, and immune cells, serve as more physiologically relevant models for anticancer drug screening and immunotherapy research compared to traditional 2D cultures. This study aimed to optimize the culturing, dissociation, and analysis of heterospheroids, addressing limitations that restrict their broader use in immunotherapy research. Our study revealed significant effects of Human Plasma-Like culture medium on cell viability, necrotic core formation, and the spatial organization of cancer and fibroblast cells within heterospheroids compared to DMEM and RPMI media. In HT-29 heterospheroids, cell viability decreased from 75 % in DMEM to 20 % in HPLM, which was accompanied by increased necrotic core formation and elevated PD-L1 expression. TrypLE™ effectively dissociated heterospheroids but compromised immune cell viability and surface marker detection. In comparison, Accutase™ significantly reduced cell yield, while collagenase I preserved immune cell markers but affected those on cancer cells. Furthermore, we developed a luciferase-based assay to measure immune-mediated cancer cell killing in heterospheroids, excluding signals from non-target cells, such as dying fibroblasts and immune cells, without requiring spheroid lysis or dissociation. Our findings highlight the importance of tailoring experimental conditions to reflect specific tumor characteristics, thus enhancing the utility of heterospheroids in drug discovery and immunotherapy research.
Collapse
Affiliation(s)
- Natasha Helleberg Madsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark.
| | - Boye Schnack Nielsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Isabella Skandorff
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | | | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Maria Ormhøj
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Kim Holmstrøm
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Jesper Larsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| | - Monika Gad
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Kogle Allé 2, 2970, Hørsholm, Denmark
| |
Collapse
|
6
|
Kah J, Staffeldt L, Volz T, Schulze K, Heumann A, Rövenstrunk G, Goebel M, Peine S, Dandri M, Lüth S. Classification of the LC4 Primarily-like Cell Line-Recapitulating a CDK4 Overexpressing Immune Evasive HIV-HCV-Induced HCC. Viruses 2025; 17:653. [PMID: 40431665 PMCID: PMC12115383 DOI: 10.3390/v17050653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality. HCC is characterized by high heterogeneity and, subsequently, adaptation by developing resistance to current treatments. Applying individualized models is crucial to understanding the potential of approved therapies. Therefore, we classify a primary-like cell line derived from the core region of an HCC with underlying HIV-HCV co-infection employing deep analysis on the pathway regulation level. METHODS We employed DEG analysis, followed by pathway analysis, to characterize the preservation level of the LC4 cells and the level of adoption. Next, we classify the new model for HCC research by employing healthy donor samples, commonly used HCC cell lines, and global RNAseq datasets. RESULTS LC4 cells reflect the characteristics of the parental cancer region, including immunosuppression and metabolic reprogramming, characterized by the downregulation of drug-metabolizing enzymes compared to healthy individuals, indicating a transition to alternate metabolic pathways. Moreover, we identified specific biomarkers equally regulated in the parental tissue, in global datasets of the same entities as well as in LC4 cells. CONCLUSIONS We classified LC4 cells as an individual immunosuppressive and highly progressive primary-like HCC cell line. LC4 cells are applicable as a model for preclinical drug testing, minimizing the lack of preclinical models in HCV-HIV-induced HCC research.
Collapse
Affiliation(s)
- Janine Kah
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
- Department of Gastroenterology, Center for Translational Medicine, University Hospital Brandenburg, 14770 Brandenburg, Germany
| | - Lisa Staffeldt
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
| | - Tassilo Volz
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, 38124 Braunschweig, Germany
| | - Kornelius Schulze
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
| | - Asmus Heumann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Götz Rövenstrunk
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Meike Goebel
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sven Peine
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maura Dandri
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, 38124 Braunschweig, Germany
| | - Stefan Lüth
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
| |
Collapse
|
7
|
Cao L, Leclercq-Cohen G, Klein C, Sorrentino A, Bacac M. Mechanistic insights into resistance mechanisms to T cell engagers. Front Immunol 2025; 16:1583044. [PMID: 40330489 PMCID: PMC12053166 DOI: 10.3389/fimmu.2025.1583044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
T cell engagers (TCEs) represent a groundbreaking advancement in the treatment of B and plasma cell malignancies and are emerging as a promising therapeutic approach for the treatment of solid tumors. These molecules harness T cells to bind to and eliminate cancer cells, effectively bypassing the need for antigen-specific T cell recognition. Despite their established clinical efficacy, a subset of patients is either refractory to TCE treatment (e.g. primary resistance) or develops resistance during the course of TCE therapy (e.g. acquired or treatment-induced resistance). In this review we comprehensively describe the resistance mechanisms to TCEs, occurring in both preclinical models and clinical trials with a particular emphasis on cellular and molecular pathways underlying the resistance process. We classify these mechanisms into tumor intrinsic and tumor extrinsic ones. Tumor intrinsic mechanisms encompass changes within tumor cells that impact the T cell-mediated cytotoxicity, including tumor antigen loss, the expression of immune checkpoint inhibitory ligands and intracellular pathways that render tumor cells resistant to killing. Tumor extrinsic mechanisms involve factors external to tumor cells, including the presence of an immunosuppressive tumor microenvironment (TME) and reduced T cell functionality. We further propose actionable strategies to overcome resistance offering potential avenues for enhancing TCE efficacy in the clinic.
Collapse
Affiliation(s)
- Linlin Cao
- Roche Innovation Center, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
8
|
Mali AK, Murugappan S, Prasad JR, Tofail SAM, Thorat ND. A deep learning pipeline for morphological and viability assessment of 3D cancer cell spheroids. Biol Methods Protoc 2025; 10:bpaf030. [PMID: 40352793 PMCID: PMC12064216 DOI: 10.1093/biomethods/bpaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Three-dimensional (3D) spheroid models have advanced cancer research by better mimicking the tumour microenvironment compared to traditional two-dimensional cell cultures. However, challenges persist in high-throughput analysis of morphological characteristics and cell viability, as traditional methods like manual fluorescence analysis are labour-intensive and inconsistent. Existing AI-based approaches often address segmentation or classification in isolation, lacking an integrated workflow. We propose a scalable, two-stage deep learning pipeline to address these gaps: (i) a U-Net model for precise detection and segmentation of 3D spheroids from microscopic images, achieving 95% prediction accuracy, and (ii) a CNN Regression Hybrid method for estimating live/dead cell percentages and classifying spheroids, with anR 2 value of 98%. This end-to-end pipeline automates cell viability quantification and generates key morphological parameters for spheroid growth kinetics. By integrating segmentation and analysis, our method addresses environmental variability and morphological characterization challenges, offering a robust tool for drug discovery, toxicity screening, and clinical research. This approach significantly improves efficiency and scalability of 3D spheroid evaluations, paving the way for advancements in cancer therapeutics.
Collapse
Affiliation(s)
- Ajay K Mali
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| | - Sivasubramanian Murugappan
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| | - Jayashree Rajesh Prasad
- Computer Science and Engineering, School of Computing, MIT Art Design and Technology University, Pune, Maharashtra, 412201, India
| | - Syed A M Tofail
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| | - Nanasaheb D Thorat
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| |
Collapse
|
9
|
Luanpitpong S, Janan M, Poohadsuan J, Rodboon N, Samart P, Rungarunlert S, Issaragrisil S. A High-Throughput, Three-Dimensional Multiple Myeloma Model Recapitulating Tumor-Stroma Interactions for CAR-Immune Cell-Mediated Cytotoxicity Assay. Immunotargets Ther 2025; 14:321-338. [PMID: 40182067 PMCID: PMC11967349 DOI: 10.2147/itt.s503984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Background Multiple myeloma (MM) is characterized by an excessive proliferation of clonal plasma cells in the bone marrow (BM). Components in BM niche contribute to the immunosuppressive tumor microenvironment (TME), but three-dimensional (3D) MM models that recreate the complex TME and enable high-throughput cytotoxicity assay of chimeric antigen receptor (CAR)-engineered immune cells are still lacking. Methods Stable, luciferase (Luc)-labeled target MM cells were generated using Luc/RFP dual reporter system to track MM growth. 3D spheroids were formed in a 96-well plate in the presence or absence of cancer-associated fibroblast (CAF)-like stromal cells activated by MM-derived conditioned medium and the cytotoxicity of CAR-immune cells, which were represented by third-generation anti-CD138 CAR-NK-92 cells, was evaluated by luciferase assay using a multimode microplate reader. Immune cell infiltration was visualized under a fluorescence microscope by using multiple fluorescent dyes. Results We first showed that luciferase assay provides a relatively simple and robust means to specifically monitor Luc-labeled tumor cell growth in a coculture system, allowing the high-throughput assessment of CAR-immune cytotoxicity. Through this assay, we demonstrated that CAF-like stromal cells impaired NK cell effector function in 2D culture and 3D spheroids, likely via paracrine signaling and physical barrier function. Importantly, we showed that 3D spheroids consisting of MM cells and CAF-like stromal cells provide a more comprehensive, physiologically relevant immuno-oncology model. Our established model could also be used to investigate the trafficking and infiltration of immune cells into the core of spheroids. Herein, we showed that CAR incorporation did improve the ability of NK cells to infiltrate 3D spheroids. Conclusion Our established 3D spheroid model, which partially recapitulates the complex TME with immunosuppressive environment, is suitable for high-throughput screening of CAR-immune cytotoxicity and could be important in accelerating immuno-oncology drug discovery for MM since there is a pressing need to establish innovative CAR-immune cells.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Cell Factory for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Cell Factory for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirarat Poohadsuan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Napachai Rodboon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parinya Samart
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sasitorn Rungarunlert
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- BDMS Center of Excellence for Hematology, Wattanosoth Cancer Hospital, Bangkok, Thailand
| |
Collapse
|
10
|
Diosdi A, Piccinini F, Boroczky T, Dobra G, Castellani G, Buzas K, Horvath P, Harmati M. Single-cell light-sheet fluorescence 3D images of tumour-stroma spheroid multicultures. Sci Data 2025; 12:492. [PMID: 40128531 PMCID: PMC11933373 DOI: 10.1038/s41597-025-04832-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Spheroids are widely used in oncology for testing drugs, but models composed of a single cell line do not fully capture the complexity of the in vivo tumours targeted by chemotherapy. Developing 3D in vitro models that better mimic tumour architecture is a crucial step for the scientific community. To enable more reliable drug testing, we generated multiculture spheroids and analysed cell morphology and distribution over time. This dataset is the first publicly available single-cell light-sheet fluorescence microscopy image collection of 3D multiculture tumour models comprising of three different cell lines analysed at different time points. Specifically, we created models composed of one cancer cell line (melanoma, breast cancer, or osteosarcoma) alongside two stromal cell lines (fibroblasts and endothelial cells). Then, we acquired single-cell resolution light-sheet fluorescence 3D images of the spheroids to analyse spheroid morphology after 24, 48, and 96 hours. The image collection, whole spheroid annotations, and extracted features are publicly available for further research and can support the development of automated analysis models.
Collapse
Affiliation(s)
- Akos Diosdi
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Filippo Piccinini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, (FC), Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Timea Boroczky
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Immunology, University of Szeged, Szeged, Hungary
- Doctoral School of Interdisciplinary Medicine, University of Szeged, Szeged, Hungary
| | - Gabriella Dobra
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gastone Castellani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna S.Orsola, Bologna, Italy
| | - Krisztina Buzas
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Immunology, University of Szeged, Szeged, Hungary
| | - Peter Horvath
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute of AI for Health, Helmholtz Zentrum München, Neuherberg, Germany
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Maria Harmati
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
11
|
Momoli C, Costa B, Lenti L, Tubertini M, Parenti MD, Martella E, Varchi G, Ferroni C. The Evolution of Anticancer 3D In Vitro Models: The Potential Role of Machine Learning and AI in the Next Generation of Animal-Free Experiments. Cancers (Basel) 2025; 17:700. [PMID: 40002293 PMCID: PMC11853635 DOI: 10.3390/cancers17040700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The development of anticancer therapies has increasingly relied on advanced 3D in vitro models, which more accurately mimic the tumor microenvironment compared to traditional 2D cultures. This review describes the evolution of these 3D models, highlighting significant advancements and their impact on cancer research. We discuss the integration of machine learning (ML) and artificial intelligence (AI) in enhancing the predictive power and efficiency of these models, potentially reducing the dependence on animal testing. ML and AI offer innovative approaches for analyzing complex data, optimizing experimental conditions, and predicting therapeutic outcomes with higher accuracy. By leveraging these technologies, the next generation of 3D in vitro models could revolutionize anticancer drug development, offering effective alternatives to animal experiments.
Collapse
Affiliation(s)
| | | | | | | | | | - Elisa Martella
- Institute for the Organic Synthesis and Photoreactivity—Italian National Research Council, 40129 Bologna, Italy; (C.M.); (B.C.); (L.L.); (M.T.); (M.D.P.); (C.F.)
| | - Greta Varchi
- Institute for the Organic Synthesis and Photoreactivity—Italian National Research Council, 40129 Bologna, Italy; (C.M.); (B.C.); (L.L.); (M.T.); (M.D.P.); (C.F.)
| | | |
Collapse
|
12
|
Shah S, Osuala KO, Brock EJ, Ji K, Sloane BF, Mattingly RR. Three-Dimensional Models: Biomimetic Tools That Recapitulate Breast Tissue Architecture and Microenvironment to Study Ductal Carcinoma In Situ Transition to Invasive Ductal Breast Cancer. Cells 2025; 14:220. [PMID: 39937011 PMCID: PMC11817749 DOI: 10.3390/cells14030220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Diagnosis of ductal carcinoma in situ (DCIS) presents a challenge as we cannot yet distinguish between those lesions that remain dormant from cases that may progress to invasive ductal breast cancer (IDC) and require therapeutic intervention. Our overall interest is to develop biomimetic three-dimensional (3D) models that more accurately recapitulate the structure and characteristics of pre-invasive breast cancer in order to study the underlying mechanisms driving malignant progression. These models allow us to mimic the microenvironment to investigate many aspects of mammary cell biology, including the role of the extracellular matrix (ECM), the interaction between carcinoma-associated fibroblasts (CAFs) and epithelial cells, and the dynamics of cytoskeletal reorganization. In this review article, we outline the significance of 3D culture models as reliable pre-clinical tools that mimic the in vivo tumor microenvironment and facilitate the study of DCIS lesions as they progress to invasive breast cancer. We also discuss the role of CAFs and other stromal cells in DCIS transition as well as the clinical significance of emerging technologies like tumor-on-chip and co-culture models.
Collapse
Affiliation(s)
- Seema Shah
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
| | | | - Ethan J. Brock
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
| | - Kyungmin Ji
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Bonnie F. Sloane
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Raymond R. Mattingly
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
13
|
Puente-Rivera J, Nuñez-Olvera SI, Fernández-Sánchez V, Cureño-Díaz MA, Gómez-Zamora E, Plascencia-Nieto ES, Figueroa-Angulo EE, Alvarez-Sánchez ME. miRNA Signatures as Predictors of Therapy Response in Castration-Resistant Prostate Cancer: Insights from Clinical Liquid Biopsies and 3D Culture Models. Genes (Basel) 2025; 16:180. [PMID: 40004509 PMCID: PMC11855684 DOI: 10.3390/genes16020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Prostate cancer (PCa) patients who do not respond to androgen deprivation therapy (ADT), referred to as castration-resistant prostate cancer (CRPC), remain a clinical challenge due to confirm the aggressive nature of CRPC and its resistance to conventional therapies. This study aims to investigate the potential of microRNAs (miRNAs) as biomarkers for predicting therapeutic response in CRPC patients. METHODS We performed miRNA and mRNA expression analyses using publicly available datasets and applied 3D cell culture models to replicate more physiologically relevant tumor conditions. Genetic analysis techniques were employed on publicly available data, and expression profiles from 3D cell culture models were examined. RESULTS Eighteen miRNAs with differential expression were identified between patients who responded favorably to abiraterone therapy (responders) and those with advanced CRPC (non-responders). Specifically, miRNAs such as hsa-miR-152-3p and hsa-miR-34a-3p were found to be associated with critical pathways, including TGF-β signaling and P53, which are linked to therapeutic resistance. Several miRNAs were identified as potential predictors of treatment efficacy, including therapies like abiraterone. CONCLUSIONS These results indicate that miRNAs could serve as non-invasive biomarkers for predicting therapeutic outcomes, facilitating a more personalized approach to CRPC treatment. This study provides a novel perspective on treatment strategies for CRPC, emphasizing the role of miRNAs in improving therapeutic precision and efficacy in this complex disease.
Collapse
Affiliation(s)
- Jonathan Puente-Rivera
- Laboratorio de Patogénesis Celular y Molecular Humana y Veterinaria, Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo 290, Col. Del Valle, Mexico City 03100, Mexico; (J.P.-R.)
- División de Investigación, Hospital Juárez De México, Mexico City 07760, Mexico
| | - Stephanie I. Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Verónica Fernández-Sánchez
- División de Investigación, Hospital Juárez De México, Mexico City 07760, Mexico
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla de Baz 54090, Mexico
| | - Monica Alethia Cureño-Díaz
- Dirección de Investigación y Enseñanza, Hospital Juárez de Mexico, Mexico City 07760, Mexico
- Departamento de Salud Pública, Facultad de Medicina, Circuito Interior, Ciudad Universitaria UNAM, Mexico City 04510, Mexico
| | | | - Estibeyesbo Said Plascencia-Nieto
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Elisa Elvira Figueroa-Angulo
- Laboratorio de Patogénesis Celular y Molecular Humana y Veterinaria, Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo 290, Col. Del Valle, Mexico City 03100, Mexico; (J.P.-R.)
| | - María Elizbeth Alvarez-Sánchez
- Laboratorio de Patogénesis Celular y Molecular Humana y Veterinaria, Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), San Lorenzo 290, Col. Del Valle, Mexico City 03100, Mexico; (J.P.-R.)
| |
Collapse
|
14
|
Ruiz-Lorente I, Gimeno L, López-Abad A, López Cubillana P, Fernández Aparicio T, Asensio Egea LJ, Moreno Avilés J, Doñate Iñiguez G, Guzmán Martínez-Valls PL, Server G, Ferri B, Campillo JA, Martínez-Sánchez MV, Minguela A. Differential Role of NKG2A/HLA-E Interaction in the Outcomes of Bladder Cancer Patients Treated with M. bovis BCG or Other Therapies. Biomedicines 2025; 13:156. [PMID: 39857739 PMCID: PMC11760850 DOI: 10.3390/biomedicines13010156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Immunotherapy is gaining great relevance in both non-muscle-invasive bladder cancer (NMIBC), with the use of bacille Calmette-Guerin (BCG), and in muscle-invasive BC (MIBC) with anti-checkpoint therapies blocking PD-1/PD-L1, CTLA-4/CD80-CD86, and, more recently, NKG2A/HLA-E interactions. Biomarkers are necessary to optimize the use of these therapies. Methods: We evaluated killer-cell immunoglobulin-like receptors (KIRs) and HLA-I genotyping and the expression of NK cell receptors in circulating T and NK lymphocytes at diagnosis in 325 consecutive BC patients (151 treated with BCG and 174 treated with other therapies), as well as in 648 patients with other cancers and 973 healthy donors as controls. The proliferation and production of cytokines and cytotoxicity were evaluated in peripheral blood mononuclear cells, stimulated in vitro with anti-CD3/CD28 or BCG, from selected patients based on HLA-B -21M/T dimorphism (NKG2A ligands). Results: The HLA-B -21M/T genotype showed opposing results in BC patients treated with BCG or other therapies. The MM genotype, compared to MT and TT, was associated with a longer 75th-percentile overall survival (not reached vs. 68.0 ± 13.7 and 52.0 ± 8.3 months, p = 0.034) in BCG, but a shorter (8.0 ± 2.4 vs. 21.0 ± 3.4 and 19.0 ± 4.9 months, p = 0.131) survival in other treatments. The HLA-B -21M/T genotype was an independent predictive parameter of the progression-free survival (HR = 2.08, p = 0.01) and the OS (HR = 2.059, p = 0.039) of BC patients treated with BCG, together with age and tumor histopathologic characteristics. The MM genotype was associated with higher counts of circulating CD56bright, fewer KIR2DL1/L2+ NK cells, and lower NKG2A expression, but not with differential in vitro NK cell functionality. Conclusions: The HLA-B -21M/T is independently associated with BC patient outcomes and can help to optimize the use of new immunotherapies in these patients.
Collapse
Affiliation(s)
- Inmaculada Ruiz-Lorente
- Immunology Service, Clinical University Hospital Virgen de la Arrixaca (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (I.R.-L.); (L.G.); (J.A.C.); (M.V.M.-S.)
| | - Lourdes Gimeno
- Immunology Service, Clinical University Hospital Virgen de la Arrixaca (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (I.R.-L.); (L.G.); (J.A.C.); (M.V.M.-S.)
- Human Anatomy Department, Universidad de Murcia and Campus Mare Nostrum, 30071 Murcia, Spain
| | - Alicia López-Abad
- Urology Service, Virgen de la Arrixaca University Clinical Hospital (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (A.L.-A.); (P.L.C.); (G.S.)
| | - Pedro López Cubillana
- Urology Service, Virgen de la Arrixaca University Clinical Hospital (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (A.L.-A.); (P.L.C.); (G.S.)
| | | | | | | | | | | | - Gerardo Server
- Urology Service, Virgen de la Arrixaca University Clinical Hospital (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (A.L.-A.); (P.L.C.); (G.S.)
| | - Belén Ferri
- Pathology Service, Clinical University Hospital Virgen de la Arrixaca (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain;
| | - José Antonio Campillo
- Immunology Service, Clinical University Hospital Virgen de la Arrixaca (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (I.R.-L.); (L.G.); (J.A.C.); (M.V.M.-S.)
| | - María Victoria Martínez-Sánchez
- Immunology Service, Clinical University Hospital Virgen de la Arrixaca (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (I.R.-L.); (L.G.); (J.A.C.); (M.V.M.-S.)
| | - Alfredo Minguela
- Immunology Service, Clinical University Hospital Virgen de la Arrixaca (HCUVA), Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (I.R.-L.); (L.G.); (J.A.C.); (M.V.M.-S.)
| |
Collapse
|
15
|
Nunes M, Ricardo S. Ivermectin Strengthens Paclitaxel Effectiveness in High-Grade Serous Carcinoma in 3D Cell Cultures. Pharmaceuticals (Basel) 2024; 18:14. [PMID: 39861076 PMCID: PMC11769219 DOI: 10.3390/ph18010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Chemoresistance is a major obstacle in high-grade serous carcinoma (HGSC) treatment. Although many patients initially respond to chemotherapy, the majority of them relapse due to Carboplatin and Paclitaxel resistance. Drug repurposing has surfaced as a potentially effective strategy that works synergically with standard chemotherapy to bypass chemoresistance. In a prior study, using 2D cultures and two HGSC chemoresistant cell lines, it was demonstrated that combining Carboplatin or Paclitaxel with Pitavastatin or Ivermectin resulted in the most notable synergy. Acknowledging that 2D culture systems are limited in reflecting the tumor architecture, 3D cultures were generated to provide insights on treatment efficacy tests in more complex models. OBJECTIVES We aimed to investigate whether combining Carboplatin or Paclitaxel with Pitavastatin or Ivermectin offers therapeutic benefits in a Cultrex-based 3D model. METHODS Here, the cytotoxicity of Carboplatin and Paclitaxel, both alone and in combination with Pitavastatin or Ivermectin, were analyzed on two chemoresistant tumor cell lines, OVCAR8 and OVCAR8 PTX R C, in 3D cultures. Cellular viability was assessed using CellTiter-Glo® Luminescent assays. Also, it explored synergistic interactions using zero interaction potency, Loewe, Bliss independence, and High-single agent reference models. RESULTS Our research indicates combining chemotherapeutic drugs with Pitavastatin or Ivermectin yields significantly more cytotoxic effects than chemotherapy alone. For all the combinations tested, at least one model indicated an additive effect; however, only the combination of Paclitaxel and Ivermectin consistently demonstrated an additive effect across all chemoresistant cell lines cultured in 3D models, as well as in all four synergy reference models used to assess drug interactions. CONCLUSIONS Combining Paclitaxel with Ivermectin has the highest cytotoxic and the strongest additive effect for both chemoresistant cell lines compared to Paclitaxel alone.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal;
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal;
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences (IUCS), University Polytechnic Higher Education Cooperative (CESPU), CRL, 4585-116 Gandra, Portugal
- Applied Molecular Biosciences Unit (UCIBIO), Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
- Oral Pathology and Rehabilitation Research Unit (UNIPRO), Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| |
Collapse
|
16
|
Vitacolonna M, Bruch R, Schneider R, Jabs J, Hafner M, Reischl M, Rudolf R. A spheroid whole mount drug testing pipeline with machine-learning based image analysis identifies cell-type specific differences in drug efficacy on a single-cell level. BMC Cancer 2024; 24:1542. [PMID: 39696122 DOI: 10.1186/s12885-024-13329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The growth and drug response of tumors are influenced by their stromal composition, both in vivo and 3D-cell culture models. Cell-type inherent features as well as mutual relationships between the different cell types in a tumor might affect drug susceptibility of the tumor as a whole and/or of its cell populations. However, a lack of single-cell procedures with sufficient detail has hampered the automated observation of cell-type-specific effects in three-dimensional stroma-tumor cell co-cultures. METHODS Here, we developed a high-content pipeline ranging from the setup of novel tumor-fibroblast spheroid co-cultures over optical tissue clearing, whole mount staining, and 3D confocal microscopy to optimized 3D-image segmentation and a 3D-deep-learning model to automate the analysis of a range of cell-type-specific processes, such as cell proliferation, apoptosis, necrosis, drug susceptibility, nuclear morphology, and cell density. RESULTS This demonstrated that co-cultures of KP-4 tumor cells with CCD-1137Sk fibroblasts exhibited a growth advantage compared to tumor cell mono-cultures, resulting in higher cell counts following cytostatic treatments with paclitaxel and doxorubicin. However, cell-type-specific single-cell analysis revealed that this apparent benefit of co-cultures was due to a higher resilience of fibroblasts against the drugs and did not indicate a higher drug resistance of the KP-4 cancer cells during co-culture. Conversely, cancer cells were partially even more susceptible in the presence of fibroblasts than in mono-cultures. CONCLUSION In summary, this underlines that a novel cell-type-specific single-cell analysis method can reveal critical insights regarding the mechanism of action of drug substances in three-dimensional cell culture models.
Collapse
Affiliation(s)
- Mario Vitacolonna
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
| | - Roman Bruch
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | | | - Julia Jabs
- Merck Healthcare KGaA, 64293, Darmstadt, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Medical Technology, Medical Faculty Mannheim of Heidelberg University, Mannheim University of Applied Sciences, 68167, Mannheim, Germany
| | - Markus Reischl
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, 76344, Eggen-stein-Leopoldshafen, Germany
| | - Rüdiger Rudolf
- CeMOS, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
| |
Collapse
|
17
|
Zaman A, Ghosh A, Ghosh AK, Das PK. DON encapsulated carbon dot-vesicle conjugate in therapeutic intervention of lung adenocarcinoma by dual targeting of CD44 and SLC1A5. NANOSCALE 2024; 16:21817-21836. [PMID: 39513401 DOI: 10.1039/d4nr00426d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Lung adenocarcinoma, recognized as one of the most formidable malignancies with a dismal prognosis and low survival rates, poses a significant challenge in its treatment. This article delineates the design and development of a carbon dot-vesicle conjugate (HACD-TMAV) for efficient cytotoxicity towards lung cancer cells by target selective delivery of the glutamine inhibitor 6-diazo-5-oxo-L-norleucine (DON) within CD44-enriched A549 cancer cells. HACD-TMAV is composed of hyaluronic acid-based carbon dots (HACDs) and trimesic acid-based vesicles (TMAV), which are bound via electrostatic interactions. TMAVs are formed by positively charged trimesic acid-based amphiphiles through H-type aggregation in water. HACDs were synthesized through a one-step hydrothermal route. The blue-emitting HACD-TMAV conjugate demonstrated selective bioimaging in CD44-overexpressed A549 lung cancer cells due to specific ligand-receptor interactions between HA and CD44. HACD-TMAV exhibited notably improved DON loading efficiency compared to individual nano-vehicles. HACD-TMAV-DON exhibited remarkable (∼6.0-fold higher) cytotoxicity against CD44-overexpressing A549 cells compared to CD44- HepG2 cells and HEK 293 normal cells. Also, DON-loaded HACD-TMAV showed ∼2.0-fold higher cytotoxicity against A549 cells compared to individual carriers and ∼4.5-fold higher cytotoxicity than by DON. Furthermore, HACD-TMAV-DON induced a ∼3.5-fold reduction in the size of 3D tumor spheroids of A549 cells. The enhanced anticancer effectiveness was attributed to starvation of the A549 cells of glutamine by dual targeting of glutamine metabolism and solute linked carrier family 1 member A5 (SLC1A5) through HA-linked CD44-mediated targeted delivery of DON. This led to over-production of reactive oxygen species (ROS) that induced apoptosis of cancer cells through downregulation of the PI3K/AKT/mTOR signaling cascade.
Collapse
Affiliation(s)
- Afreen Zaman
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700032, India.
| | - Aparajita Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700032, India.
| | - Anup Kumar Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700032, India.
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700032, India.
| |
Collapse
|
18
|
Imran M, Moyle PM, Kamato D, Mohammed Y. Advances in, and prospects of, 3D preclinical models for skin drug discovery. Drug Discov Today 2024; 29:104208. [PMID: 39396673 DOI: 10.1016/j.drudis.2024.104208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
The skin has an important role in regulating homeostasis and protecting the body from endogenous and exogenous microenvironments. Although 3D models for drug discovery have been extensively studied, there is a growing demand for more advanced 3D skin models to enhance skin research. The use of these advanced skin models holds promise across domains such as cosmetics, skin disease treatments, and toxicity testing of new therapeutics. Recent advances include the development of skin-on-a-chip, spheroids, reconstructed skin, organoids, and computational approaches, including quantitative structure-activity relationship (QSAR) and quantitative structure-property relationship (QSPR) research. These innovations are bridging the gap between traditional 2D and advanced 3D models, moving progress from research to clinical applications. In this review, we highlight in vitro and computational skin models with advanced drug discovery for skin-related applications.
Collapse
Affiliation(s)
- Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Peter Michael Moyle
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia; School of Environment and Science, Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD 4111, Australia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| |
Collapse
|
19
|
Qureshi AA, Wehrle CJ, Ferreira-Gonzalez S, Jiao C, Hong H, Dadgar N, Arpi-Palacios J, Phong YP, Kim J, Sun K, Hashimoto K, Kwon DCH, Miller C, Leipzig N, Ma WW, Melenhorst J, Aucejo F, Schlegel A. Tumor organoids for primary liver cancers: A systematic review of current applications in diagnostics, disease modeling, and drug screening. JHEP Rep 2024; 6:101164. [PMID: 39583095 PMCID: PMC11584567 DOI: 10.1016/j.jhepr.2024.101164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 11/26/2024] Open
Abstract
Background & Aims Liver cancer-related deaths are projected to exceed one million annually by 2030. Existing therapies have significant limitations, including severe side effects and inconsistent efficacy. Innovative therapeutic approaches to address primary liver cancer (PLC) have led to the ongoing development of tumor-derived organoids. These are sophisticated three-dimensional structures capable of mimicking native tissue architecture and function in vitro, improving our ability to model in vivo homeostasis and disease. Methods This systematic review consolidates known literature on human and mouse liver organoids across all PLC subtypes, emphasizing diagnostic precision, disease modeling, and drug screening capabilities. Results Across all 39 included studies, organoids were most frequently patient-derived, closely followed by cancer cell line-derived. The literature concentrated on hepatocellular carcinoma and intrahepatic cholangiocarcinoma, while exploration of other subtypes was limited. These studies demonstrate a valuable role for PLC organoid cultures in biomarker discovery, disease modeling, and therapeutic exploration. Conclusions Encouraging advances such as organoid-on-a-chip and co-culturing systems hold promise for advancing treatment regimens for PLC. Standardizing in vitro protocols is crucial to integrate research breakthroughs into practical treatment strategies for PLC. Impact and implications This study provides an overview of the current understanding of tumor-derived organoids in primary liver cancers, emphasizing their potential in diagnostics, disease modeling, and drug screening. The scientific foundation rests on the organoids' ability to replicate the tumor microenvironment and genetic landscape, opening new avenues for personalized therapies. These insights are crucial for both researchers and clinicians, as patient-derived organoids can help identify biomarkers and therapeutic targets. Physicians and policymakers can harness these advances to drive progress in precision medicine, while recognizing the challenges involved in standardizing organoid models for clinical implementation.
Collapse
Affiliation(s)
- Ayesha A. Qureshi
- Nationwide Children's Hospital, Abigail Wexner Research Institute, 575 Children's Crossroad, Columbus, OH, 43215, USA
| | | | - Sofia Ferreira-Gonzalez
- CIR Centre for Inflammation Research, University of Edinburgh, 5 Little France Drive Edinburgh, EH16 4UU, UK
| | - Chunbao Jiao
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hanna Hong
- Transplantation Center, Cleveland Clinic, OH, USA
| | - Neda Dadgar
- Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH, USA
- Translational Hematology & Oncology Research, Cleveland Clinic, Enterprise Cancer Institute, Cleveland, OH, USA
| | - Jorge Arpi-Palacios
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH, USA
| | - Yee Phoon Phong
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH, USA
| | - Jaekeun Kim
- Transplantation Center, Cleveland Clinic, OH, USA
| | - Keyue Sun
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | - Nic Leipzig
- The University of Akron, Department of Chemical, Biomolecular, and Corrosion Engineering, Akron, OH, USA
| | - Wen Wee Ma
- Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH, USA
| | - Jos Melenhorst
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH, USA
| | | | - Andrea Schlegel
- Transplantation Center, Cleveland Clinic, OH, USA
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
20
|
Cordeiro S, Oliveira BB, Valente R, Ferreira D, Luz A, Baptista PV, Fernandes AR. Breaking the mold: 3D cell cultures reshaping the future of cancer research. Front Cell Dev Biol 2024; 12:1507388. [PMID: 39659521 PMCID: PMC11628512 DOI: 10.3389/fcell.2024.1507388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Despite extensive efforts to unravel tumor behavior and develop anticancer therapies, most treatments fail when advanced to clinical trials. The main challenge in cancer research has been the absence of predictive cancer models, accurately mimicking the tumoral processes and response to treatments. The tumor microenvironment (TME) shows several human-specific physical and chemical properties, which cannot be fully recapitulated by the conventional 2D cell cultures or the in vivo animal models. These limitations have driven the development of novel in vitro cancer models, that get one step closer to the typical features of in vivo systems while showing better species relevance. This review introduces the main considerations required for developing and exploiting tumor spheroids and organoids as cancer models. We also detailed their applications in drug screening and personalized medicine. Further, we show the transition of these models into novel microfluidic platforms, for improved control over physiological parameters and high-throughput screening. 3D culture models have provided key insights into tumor biology, more closely resembling the in vivo TME and tumor characteristics, while enabling the development of more reliable and precise anticancer therapies.
Collapse
Affiliation(s)
- Sandra Cordeiro
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Beatriz B. Oliveira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ruben Valente
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Daniela Ferreira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - André Luz
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
21
|
Zhao Z, Feng X, Wu H, Chen S, Ma C, Guan Z, Lei L, Tang K, Chen X, Dong Y, Tang Y. Construction of a lung cancer 3D culture model based on alginate/gelatin micro-beads for drug evaluation. Transl Lung Cancer Res 2024; 13:2698-2712. [PMID: 39507032 PMCID: PMC11535844 DOI: 10.21037/tlcr-24-490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024]
Abstract
Background Lung cancer is one of the most common malignant tumors worldwide. Despite advances in lung cancer treatment, patients still face challenges related to drug resistance and recurrence. Current methods for evaluating anti-cancer drug activity are insufficient, as they rely on two-dimensional (2D) cell culture and animal models. Therefore, the development of an in vitro drug evaluation model capable of predicting individual sensitivity to anti-cancer drugs would greatly enhance the success rate of drug treatments for lung cancer patients. The purpose of this research is to utilise conditional reprogramming technology to cultivate patient-derived lung cancer cells and to construct an in vitro 3D culture model using sodium alginate (SA) and gelatin. The aim is to study the biological characteristics of cells in the 3D culture model and to further investigate the sensitivity of anti-cancer drugs based on the alginate-gelatin 3D culture model. This approach provides new means and insights for personalized precision anti-cancer therapy and the development of new anti-cancer drugs. Methods Conditional reprogramming technology was used to generate conditionally reprogrammed lung adenocarcinoma cells (CRLCs). Alginate-gelatin hydrogel micro-beads were created to explore their potential use in the assessment of anti-cancer drugs. Cell proliferation was also examined using the MTS assay method. Live/dead staining was performed to estimate cell distribution and viability using calcein acetoxymethyl ester/propidium iodide (calcein-AM/PI) double staining. Protein expression was assessed by Western blot. Results The cells grown in the three-dimensional (3D) culture were in a state of continuous proliferation, and there was an obvious phenomenon of cell mass growth. The drug sensitivity assay results demonstrated that compared with the 2D-grown cells, the CRLCs grown in the alginate-gelatin hydrogel micro-beads exhibited more resistance to anti-cancer drugs. The results also showed that the 3D-cultured CRLCs showed greater protein expression levels of stem cell hallmarks, such as Nanog Homeobox (NANOG), SRY-Box Transcription Factor 2 (SOX-2), and aldehyde dehydrogenase 1 family member A1 (ALDH1A1), than the 2D-grown cells. Conclusions These findings suggest that the 3D hydrogel cell culture models more closely mimicked the in vivo biological and clinical behavior of cells, and demonstrated higher innate resistance to anti-cancer drugs than the 2D cell culture models, and thus could serve as valuable tools for diagnosis, drug screening, and personalized medicine.
Collapse
Affiliation(s)
- Ziying Zhao
- Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqing Feng
- Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Huijuan Wu
- Department of Medical Science Research, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Shuisheng Chen
- Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changsong Ma
- Department of Orthopaedics, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ziyun Guan
- Department of Emergency, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Luwen Lei
- Department of Pharmacy, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Kejing Tang
- Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Division of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiao Chen
- Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Dong
- Division of Interventional Radiology, the First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, China
| | - Yubo Tang
- Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
Sanz-Villafruela J, Bermejo-Casadesus C, Zafon E, Martínez-Alonso M, Durá G, Heras A, Soriano-Díaz I, Giussani A, Ortí E, Tebar F, Espino G, Massaguer A. Insights into the anticancer photodynamic activity of Ir(III) and Ru(II) polypyridyl complexes bearing β-carboline ligands. Eur J Med Chem 2024; 276:116618. [PMID: 38972079 DOI: 10.1016/j.ejmech.2024.116618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/31/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
Ir(III) and Ru(II) polypyridyl complexes are promising photosensitizers (PSs) for photodynamic therapy (PDT) due to their outstanding photophysical properties. Herein, one series of cyclometallated Ir(III) complexes and two series of Ru(II) polypyridyl derivatives bearing three different thiazolyl-β-carboline N^N' ligands have been synthesized, aiming to evaluate the impact of the different metal fragments ([Ir(C^N)2]+ or [Ru(N^N)2]2+) and N^N' ligands on the photophysical and biological properties. All the compounds exhibit remarkable photostability under blue-light irradiation and are emissive (605 < λem < 720 nm), with the Ru(II) derivatives displaying higher photoluminescence quantum yields and longer excited state lifetimes. The Ir PSs display pKa values between 5.9 and 7.9, whereas their Ru counterparts are less acidic (pKa > 9.3). The presence of the deprotonated form in the Ir-PSs favours the generation of reactive oxygen species (ROS) since, according to theoretical calculations, it features a low-lying ligand-centered triplet excited state (T1 = 3LC) with a long lifetime. All compounds have demonstrated anticancer activity. Ir(III) complexes 1-3 exhibit the highest cytotoxicity in dark conditions, comparable to cisplatin. Their activity is notably enhanced by blue-light irradiation, resulting in nanomolar IC50 values and phototoxicity indexes (PIs) between 70 and 201 in different cancer cell lines. The Ir(III) PSs are also activated by green (with PI between 16 and 19.2) and red light in the case of complex 3 (PI = 8.5). Their antitumor efficacy is confirmed by clonogenic assays and using spheroid models. The Ir(III) complexes rapidly enter cells, accumulating in mitochondria and lysosomes. Upon photoactivation, they generate ROS, leading to mitochondrial dysfunction and lysosomal damage and ultimately cell apoptosis. Additionally, they inhibit cancer cell migration, a crucial step in metastasis. In contrast, Ru(II) complex 6 exhibits moderate mitochondrial activity. Overall, Ir(III) complexes 1-3 show potential for selective light-controlled cancer treatment, providing an alternative mechanism to chemotherapy and the ability to inhibit lethal cancer cell dissemination.
Collapse
Affiliation(s)
- Juan Sanz-Villafruela
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Cristina Bermejo-Casadesus
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain
| | - Elisenda Zafon
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain
| | - Marta Martínez-Alonso
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Gema Durá
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica. Facultad de Químicas, Avda. Camilo J. Cela 10, 13071, Ciudad Real, Spain
| | - Aranzazu Heras
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain
| | - Iván Soriano-Díaz
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain
| | - Angelo Giussani
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain
| | - Enrique Ortí
- Instituto de Ciencia Molecular, Universidad de Valencia, Catedrático José Beltrán 2, 46980, Paterna, Spain.
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain.
| | - Gustavo Espino
- Universidad de Burgos, Departamento de Química, Facultad de Ciencias, Plaza Misael Bañuelos S/n, 09001, Burgos, Spain.
| | - Anna Massaguer
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003, Girona, Spain.
| |
Collapse
|
23
|
Esposito A, Ferraresi A, Vallino L, Garavaglia B, Dhanasekaran DN, Isidoro C. Three-Dimensional In Vitro Cell Cultures as a Feasible and Promising Alternative to Two-Dimensional and Animal Models in Cancer Research. Int J Biol Sci 2024; 20:5293-5311. [PMID: 39430243 PMCID: PMC11488579 DOI: 10.7150/ijbs.96469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/25/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer represents one of the diseases with the highest mortality rate worldwide. The burden of cancer continues to increase, not only affecting the health-related quality of life of patients but also causing an elevated global financial impact. The complexity and heterogeneity of cancer pose significant challenges in research and clinical practice, contributing to increase the failure rate of clinical trials for antitumoral drugs. This is partially due to the fact that preclinical models still present important limitations in faithfully recapitulating human tumors to serve as reliable indicators of drug effectiveness. Up to now, research and development strategies employ expensive animal models (including the so-called "humanized mice") that not only raise ethical concerns, but also frequently fail to accurately predict responses to anticancer drugs because they do not faithfully replicate human physiology as well as the patient's tumor microenvironment. On the other side, traditional two-dimensional (2D) cell cultures fail to adequately reproduce the structural organization of tumor and the cellular heterogeneity found in vivo. The growing necessity to develop more accurate cancer models has increasingly emphasized the importance of three-dimensional (3D) in vitro cell cultures, such as cancer-derived spheroids and organoids, as promising alternatives to bridge the gap between 2D and animal models. In this review, we provide a brief overview focusing on 3D in vitro cell cultures as preclinical models capable of properly reproducing the tissue organization, biological composition, and complexity of in vivo tumors in a fine-tuned microenvironment. Despite their limitations, these models collectively enhance our understanding of the mechanisms underlying cancer and may offer the potential for a more reliable assessment of drug efficacy before clinical testing and, consequently, improve therapeutic outcomes for cancer patients.
Collapse
Affiliation(s)
- Andrea Esposito
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Beatrice Garavaglia
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| |
Collapse
|
24
|
Emond R, West J, Grolmusz V, Cosgrove P, Nath A, Anderson AR, Bild AH. A novel combination therapy for ER+ breast cancer suppresses drug resistance via an evolutionary double-bind. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611032. [PMID: 39282402 PMCID: PMC11398327 DOI: 10.1101/2024.09.03.611032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Chemotherapy remains a commonly used and important treatment option for metastatic breast cancer. A majority of ER+ metastatic breast cancer patients ultimately develop resistance to chemotherapy, resulting in disease progression. We hypothesized that an "evolutionary double-bind", where treatment with one drug improves the response to a different agent, would improve the effectiveness and durability of responses to chemotherapy. This approach exploits vulnerabilities in acquired resistance mechanisms. Evolutionary models can be used in refractory cancer to identify alternative treatment strategies that capitalize on acquired vulnerabilities and resistance traits for improved outcomes. To develop and test these models, ER+ breast cancer cell lineages sensitive and resistant to chemotherapy are grown in spheroids with varied initial population frequencies to measure cross-sensitivity and efficacy of chemotherapy and add-on treatments such as disulfiram combination treatment. Different treatment schedules then assessed the best strategy for reducing the selection of resistant populations. We developed and parameterized a game-theoretic mathematical model from this in vitro experimental data, and used it to predict the existence of a double-bind where selection for resistance to chemotherapy induces sensitivity to disulfiram. The model predicts a dose-dependent re-sensitization (a double-bind) to chemotherapy for monotherapy disulfiram.
Collapse
Affiliation(s)
- Rena Emond
- City of Hope, Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, 91016, USA
| | - Jeffrey West
- Integrated Mathematical Oncology Dept. Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL 33612
| | - Vince Grolmusz
- City of Hope, Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, 91016, USA
| | - Patrick Cosgrove
- City of Hope, Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, 91016, USA
| | - Aritro Nath
- City of Hope, Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, 91016, USA
| | - Alexander R.A. Anderson
- Integrated Mathematical Oncology Dept. Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL 33612
| | - Andrea H. Bild
- City of Hope, Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, 91016, USA
| |
Collapse
|
25
|
Ostapowicz J, Ostrowska K, Golusiński W, Kulcenty K, Suchorska WM. Improving therapeutic strategies for Head and Neck Cancer: Insights from 3D hypoxic cell culture models in treatment response evaluation. Adv Med Sci 2024; 69:368-376. [PMID: 39047970 DOI: 10.1016/j.advms.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Hypoxia in the tumor core negatively affects the outcome of patients with head and neck squamous cell carcinoma (HNSCC). Nevertheless, its role in predicting treatment response requires further exploration. Typically, reduced oxygen levels in the tumor core correlate with diminished efficacy of radiotherapy, chemotherapy, and immunotherapy, which are commonly used for HNSCC patients' treatment. Understanding the mechanistic underpinnings of these varied treatment responses in HNSCC is crucial for enhancing therapeutic outcomes and extending patients' overall survival (OS) rates. Standard monolayer cell culture conditions have major limitations in mimicking tumor physiological features and the complexity of the tumor microenvironment. Three-dimensional (3D) cell cultures enable the recreation of the in vivo tumor attributes, encompassing oxygen and nutrient gradients, cellular morphology, and intracellular connections. It is vital to use the 3D model in treatment response studies to mimic the tumor microenvironment, as evidenced by the decreased sensitivity of 3D structures to anticancer therapy. Accordingly, the aim of the study was to delineate the utility of the 3D models of hypoxic head and neck tumors in drug screening and treatment response studies.
Collapse
Affiliation(s)
- Julia Ostapowicz
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland; Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| | - Kamila Ostrowska
- Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland; Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland
| | - Wojciech Golusiński
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland
| | - Wiktoria M Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland; Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
26
|
Pong KCC, Lai YS, Wong RCH, Lee ACK, Chow SCT, Lam JCW, Ho HP, Wong CTT. Automated Uniform Spheroid Generation Platform for High Throughput Drug Screening Process. BIOSENSORS 2024; 14:392. [PMID: 39194621 DOI: 10.3390/bios14080392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
Three-dimensional (3D) spheroid models are crucial for cancer research, offering more accurate insights into tumour biology and drug responses than traditional 2D cell cultures. However, inconsistent and low-throughput spheroid production has hindered their application in drug screening. Here, we present an automated high-throughput platform for a spheroid selection, fabrication, and sorting system (SFSS) to produce uniform gelatine-encapsulated spheroids (GESs) with high efficiency. SFSS integrates advanced imaging, analysis, photo-triggered fabrication, and microfluidic sorting to precisely control spheroid size, shape, and viability. Our data demonstrate that our SFSS can produce over 50 GESs with consistent size and circularity in 30 min with over 97% sorting accuracy while maintaining cell viability and structural integrity. We demonstrated that the GESs can be used for drug screening and potentially for various assays. Thus, the SFSS could significantly enhance the efficiency of generating uniform spheroids, facilitating their application in drug development to investigate complex biological systems and drug responses in a more physiologically relevant context.
Collapse
Affiliation(s)
- Kelvin C C Pong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, New Territories, Hong Kong, China
- BioArchitec Group Limited, Hong Kong, China
| | - Yuen Sze Lai
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Roy Chi Hang Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Alan Chun Kit Lee
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | | | | | - Ho Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, New Territories, Hong Kong, China
| | - Clarence T T Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| |
Collapse
|
27
|
de Roode KE, Hashemi K, Verdurmen WPR, Brock R. Tumor-On-A-Chip Models for Predicting In Vivo Nanoparticle Behavior. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402311. [PMID: 38700060 DOI: 10.1002/smll.202402311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Indexed: 05/05/2024]
Abstract
Nanosized drug formulations are broadly explored for the improvement of cancer therapy. Prediction of in vivo nanoparticle (NP) behavior, however, is challenging, given the complexity of the tumor and its microenvironment. Microfluidic tumor-on-a-chip models are gaining popularity for the in vitro testing of nanoparticle targeting under conditions that simulate the 3D tumor (microenvironment). In this review, following a description of the tumor microenvironment (TME), the state of the art regarding tumor-on-a-chip models for investigating nanoparticle delivery to solid tumors is summarized. The models are classified based on the degree of compartmentalization (single/multi-compartment) and cell composition (tumor only/tumor microenvironment). The physiological relevance of the models is critically evaluated. Overall, microfluidic tumor-on-a-chip models greatly improve the simulation of the TME in comparison to 2D tissue cultures and static 3D spheroid models and contribute to the understanding of nanoparticle behavior. Interestingly, two interrelated aspects have received little attention so far which are the presence and potential impact of a protein corona as well as nanoparticle uptake through phagocytosing cells. A better understanding of their relevance for the predictive capacity of tumor-on-a-chip systems and development of best practices will be a next step for the further refinement of advanced in vitro tumor models.
Collapse
Affiliation(s)
- Kim E de Roode
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Khadijeh Hashemi
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Roland Brock
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, 329, Bahrain
| |
Collapse
|
28
|
Joy JD, Malacrida B, Laforêts F, Kotantaki P, Maniati E, Manchanda R, Annibaldi A, Hopkins S, Garrobo-Calleja I, Gautrot J, Balkwill FR. Human 3D Ovarian Cancer Models Reveal Malignant Cell-Intrinsic and -Extrinsic Factors That Influence CAR T-cell Activity. Cancer Res 2024; 84:2432-2449. [PMID: 38819641 PMCID: PMC11292204 DOI: 10.1158/0008-5472.can-23-3007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/29/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
In vitro preclinical testing of chimeric antigen receptor (CAR) T cells is mostly carried out in monolayer cell cultures. However, alternative strategies are needed to take into account the complexity and the effects of the tumor microenvironment. Here, we describe the modulation of CAR T-cell activity by malignant cells and fibroblasts in human three-dimensional (3D) in vitro cell models of increasing complexity. In models combining mucin-1 (MUC1) and TnMUC1 CAR T cells with human high-grade serous ovarian cancer cell spheroids, malignant cell-intrinsic resistance to CAR T-cell killing was due to defective death receptor signaling involving TNFα. Adding primary human fibroblasts to spheroids unexpectedly increased the ability of CAR T cells to kill resistant malignant cells as CCL2 produced by fibroblasts activated CCR2/4+ CAR T cells. However, culturing malignant cells and fibroblasts in collagen gels engendered production of a dense extracellular matrix that impeded CAR T-cell activity in a TGFβ-dependent manner. A vascularized microfluidic device was developed that allowed CAR T cells to flow through the vessels and penetrate the gels in a more physiological way, killing malignant cells in a TNFα-dependent manner. Complex 3D human cell models may provide an efficient way of screening multiple cytotoxic human immune cell constructs while also enabling evaluation of mechanisms of resistance involving cell-cell and cell-matrix interactions, thus accelerating preclinical research on cytotoxic immune cell therapies in solid tumors. Significance: Three-dimensional in vitro models of increasing complexity uncover mechanisms of resistance to CAR T cells in solid tumors, which could help accelerate development of improved CAR T-cell constructs.
Collapse
Affiliation(s)
- Joash D. Joy
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Beatrice Malacrida
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Florian Laforêts
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Panoraia Kotantaki
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Cancer Research UK, Barts Centre, Queen Mary University of London, London, United Kingdom.
- Department of Gynaecological Oncology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom.
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | | | - Sarah Hopkins
- GlaxoSmithKline Medicines Research Centre, Stevenage, United Kingdom.
| | | | - Julien Gautrot
- School of Engineering and Material Science, Centre for Bioengineering, Queen Mary University of London, London, United Kingdom.
| | - Frances R. Balkwill
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
29
|
Gonçalves PP, da Silva CL, Bernardes N. Advancing cancer therapeutics: Integrating scalable 3D cancer models, extracellular vesicles, and omics for enhanced therapy efficacy. Adv Cancer Res 2024; 163:137-185. [PMID: 39271262 DOI: 10.1016/bs.acr.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Cancer remains as one of the highest challenges to human health. However, anticancer drugs exhibit one of the highest attrition rates compared to other therapeutic interventions. In part, this can be attributed to a prevalent use of in vitro models with limited recapitulative potential of the in vivo settings. Three dimensional (3D) models, such as tumor spheroids and organoids, offer many research opportunities to address the urgent need in developing models capable to more accurately mimic cancer biology and drug resistance profiles. However, their wide adoption in high-throughput pre-clinical studies is dependent on scalable manufacturing to support large-scale therapeutic drug screenings and multi-omic approaches for their comprehensive cellular and molecular characterization. Extracellular vesicles (EVs), which have been emerging as promising drug delivery systems (DDS), stand to significantly benefit from such screenings conducted in realistic cancer models. Furthermore, the integration of these nanomedicines with 3D cancer models and omics profiling holds the potential to deepen our understanding of EV-mediated anticancer effects. In this chapter, we provide an overview of the existing 3D models used in cancer research, namely spheroids and organoids, the innovations in their scalable production and discuss how omics can facilitate the implementation of these models at different stages of drug testing. We also explore how EVs can advance drug delivery in cancer therapies and how the synergy between 3D cancer models and omics approaches can benefit in this process.
Collapse
Affiliation(s)
- Pedro P Gonçalves
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Nuno Bernardes
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
30
|
Angeli C, Wroblewska JP, Klein E, Margue C, Kreis S. Protocol to generate scaffold-free, multicomponent 3D melanoma spheroid models for preclinical drug testing. STAR Protoc 2024; 5:103058. [PMID: 38748881 PMCID: PMC11109877 DOI: 10.1016/j.xpro.2024.103058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/06/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
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.
Collapse
Affiliation(s)
- Cristian Angeli
- Department of Life Sciences and Medicine, University of Luxembourg, 4367 Belvaux, Luxembourg.
| | | | - Eliane Klein
- Department of Life Sciences and Medicine, University of Luxembourg, 4367 Belvaux, Luxembourg.
| | - Christiane Margue
- Department of Life Sciences and Medicine, University of Luxembourg, 4367 Belvaux, Luxembourg
| | - Stephanie Kreis
- Department of Life Sciences and Medicine, University of Luxembourg, 4367 Belvaux, Luxembourg.
| |
Collapse
|
31
|
Bloise N, Giannaccari M, Guagliano G, Peluso E, Restivo E, Strada S, Volpini C, Petrini P, Visai L. Growing Role of 3D In Vitro Cell Cultures in the Study of Cellular and Molecular Mechanisms: Short Focus on Breast Cancer, Endometriosis, Liver and Infectious Diseases. Cells 2024; 13:1054. [PMID: 38920683 PMCID: PMC11201503 DOI: 10.3390/cells13121054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Over the past decade, the development of three-dimensional (3D) models has increased exponentially, facilitating the unravelling of fundamental and essential cellular mechanisms by which cells communicate with each other, assemble into tissues and organs and respond to biochemical and biophysical stimuli under both physiological and pathological conditions. This section presents a concise overview of the most recent updates on the significant contribution of different types of 3D cell cultures including spheroids, organoids and organ-on-chip and bio-printed tissues in advancing our understanding of cellular and molecular mechanisms. The case studies presented include the 3D cultures of breast cancer (BC), endometriosis, the liver microenvironment and infections. In BC, the establishment of 3D culture models has permitted the visualization of the role of cancer-associated fibroblasts in the delivery of exosomes, as well as the significance of the physical properties of the extracellular matrix in promoting cell proliferation and invasion. This approach has also become a valuable tool in gaining insight into general and specific mechanisms of drug resistance. Given the considerable heterogeneity of endometriosis, 3D models offer a more accurate representation of the in vivo microenvironment, thereby facilitating the identification and translation of novel targeted therapeutic strategies. The advantages provided by 3D models of the hepatic environment, in conjunction with the high throughput characterizing various platforms, have enabled the elucidation of complex molecular mechanisms underlying various threatening hepatic diseases. A limited number of 3D models for gut and skin infections have been developed. However, a more profound comprehension of the spatial and temporal interactions between microbes, the host and their environment may facilitate the advancement of in vitro, ex vivo and in vivo disease models. Additionally, it may pave the way for the development of novel therapeutic approaches in diverse research fields. The interested reader will also find concluding remarks on the challenges and prospects of using 3D cell cultures for discovering cellular and molecular mechanisms in the research areas covered in this review.
Collapse
Affiliation(s)
- Nora Bloise
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| | - Marialaura Giannaccari
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Giuseppe Guagliano
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
| | - Emanuela Peluso
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Elisa Restivo
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
| | - Silvia Strada
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Cristina Volpini
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Paola Petrini
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, P.zza L. Da Vinci 32, 20133 Milan, Italy; (G.G.); (P.P.)
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of Politecnico di Milano, 20133 Milan, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Centre for Health Technologies (CHT), Unità di Ricerca (UdR) INSTM, University of Pavia, 27100 Pavia, Italy; (M.G.); (E.P.); (E.R.); (S.S.); (C.V.)
- UOR6 Nanotechnology Laboratory, Department of Prevention and Rehabilitation in Occupational Medicine and Specialty Medicine, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), Operative Unit (OU) of University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
32
|
Yadav R, Mahajan S, Singh H, Mehra NK, Madan J, Doijad N, Singh PK, Guru SK. Emerging In Vitro and In Vivo Models: Hope for the Better Understanding of Cancer Progression and Treatment. Adv Biol (Weinh) 2024; 8:e2300487. [PMID: 38581078 DOI: 10.1002/adbi.202300487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Various cancer models have been developed to aid the understanding of the underlying mechanisms of tumor development and evaluate the effectiveness of various anticancer drugs in preclinical studies. These models accurately reproduce the critical stages of tumor initiation and development to mimic the tumor microenvironment better. Using these models for target validation, tumor response evaluation, resistance modeling, and toxicity comprehension can significantly enhance the drug development process. Herein, various in vivo or animal models are presented, typically consisting of several mice and in vitro models ranging in complexity from transwell models to spheroids and CRISPR-Cas9 technologies. While in vitro models have been used for decades and dominate the early stages of drug development, they are still limited primary to simplistic tests based on testing on a single cell type cultivated in Petri dishes. Recent advancements in developing new cancer therapies necessitate the generation of complicated animal models that accurately mimic the tumor's complexity and microenvironment. Mice make effective tumor models as they are affordable, have a short reproductive cycle, exhibit rapid tumor growth, and are simple to manipulate genetically. Human cancer mouse models are crucial to understanding the neoplastic process and basic and clinical research improvements. The following review summarizes different in vitro and in vivo metastasis models, their advantages and disadvantages, and their ability to serve as a model for cancer research.
Collapse
Affiliation(s)
- Rachana Yadav
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Hoshiyar Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Neelesh Kumar Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Nandkumar Doijad
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
33
|
Avci CB, Bagca BG, Shademan B, Takanlou LS, Takanlou MS, Nourazarian A. The future of cancer therapy: exploring the potential of patient-derived organoids in drug development. Front Cell Dev Biol 2024; 12:1401504. [PMID: 38835507 PMCID: PMC11149425 DOI: 10.3389/fcell.2024.1401504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Cancer therapy is on the brink of a significant transformation with the inclusion of patient-derived organoids (PDOs) in drug development. These three-dimensional cell cultures, directly derived from a patient's tumor, accurately replicate the complex structure and genetic makeup of the original cancer. This makes them a promising tool for advancing oncology. In this review, we explore the practical applications of PDOs in clinical drug screening and pharmacognostic assessment, as well as their role in refining therapeutic strategies. We provide insights into the latest advancements in PDO technology and its implications for predicting treatment responses and facilitating novel drug discoveries. Additionally, we address the operational challenges associated with incorporating PDOs into the drug development process, such as scaling up organoid cultures, ensuring consistent results, and addressing the ethical use of patient-derived materials. Aimed at researchers, clinicians, and key stakeholders in oncology, this article aims to succinctly present both the extraordinary potential and the obstacles to integrating PDOs, thereby shedding light on their prospective impact on the future of cancer treatment.
Collapse
Affiliation(s)
- Cigir Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Türkiye
| | - Bakiye Goker Bagca
- Department of Medical Biology, Faculty of Medicine, Adnan Menderes University, Aydın, Türkiye
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
34
|
Yayan J, Franke KJ, Berger M, Windisch W, Rasche K. Adhesion, metastasis, and inhibition of cancer cells: a comprehensive review. Mol Biol Rep 2024; 51:165. [PMID: 38252369 PMCID: PMC10803487 DOI: 10.1007/s11033-023-08920-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
This comprehensive review delves into cancer's complexity, focusing on adhesion, metastasis, and inhibition. It explores the pivotal role of these factors in disease progression and therapeutic strategies. This review covers cancer cell migration, invasion, and colonization of distant organs, emphasizing the significance of cell adhesion and the intricate metastasis process. Inhibition approaches targeting adhesion molecules, such as integrins and cadherins, are discussed. Overall, this review contributes significantly to advancing cancer research and developing targeted therapies, holding promise for improving patient outcomes worldwide. Exploring different inhibition strategies revealed promising therapeutic targets to alleviate adhesion and metastasis of cancer cells. The effectiveness of integrin-blocking antibodies, small molecule inhibitors targeting Focal adhesion kinase (FAK) and the Transforming Growth Factor β (TGF-β) pathway, and combination therapies underscores their potential to disrupt focal adhesions and control epithelial-mesenchymal transition processes. The identification of as FAK, Src, β-catenin and SMAD4 offers valuable starting points for further research and the development of targeted therapies. The complex interrelationships between adhesion and metastatic signaling networks will be relevant to the development of new treatment approaches.
Collapse
Affiliation(s)
- Josef Yayan
- Department of Internal Medicine, Division of Pulmonary, Allergy, and Sleep Medicine, Witten/Herdecke University, HELIOS Clinic Wuppertal, Heusnerstr. 40, 42283, Wuppertal, Germany.
| | - Karl-Josef Franke
- Department of Internal Medicine, Pulmonary Division, Internal Intensive Care Medicine, Infectiology, and Sleep Medicine, Märkische Clinics Health Holding Ltd, Clinic Lüdenscheid, Witten/Herdecke University, Lüdenscheid, Germany
| | - Melanie Berger
- Department of Pneumology, Cologne Merheim Hospital, Witten/Herdecke University, Cologne, Germany
| | - Wolfram Windisch
- Department of Pneumology, Cologne Merheim Hospital, Witten/Herdecke University, Cologne, Germany
| | - Kurt Rasche
- Department of Internal Medicine, Division of Pulmonary, Allergy, and Sleep Medicine, Witten/Herdecke University, HELIOS Clinic Wuppertal, Heusnerstr. 40, 42283, Wuppertal, Germany
| |
Collapse
|
35
|
Fontana F, Sommariva M, Anselmi M, Bianchi F, Limonta P, Gagliano N. Differentiation States of Phenotypic Transition of Melanoma Cells Are Revealed by 3D Cell Cultures. Cells 2024; 13:181. [PMID: 38247872 PMCID: PMC10814891 DOI: 10.3390/cells13020181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Melanoma is characterized by high metastatic potential favored by the epithelial-to-mesenchymal transition (EMT), leading melanoma cells to exhibit a spectrum of typical EMT markers. This study aimed to analyze the expression of EMT markers in A375 and BLM melanoma cell lines cultured in 2D monolayers and 3D spheroids using morphological and molecular methods. The expression of EMT markers was strongly affected by 3D arrangement and revealed a hybrid phenotype for the two cell lines. Indeed, although E-cadherin was almost undetectable in both A375 and BLM cells, cortical actin was detected in A375 2D monolayers and 3D spheroids and was strongly expressed in BLM 3D spheroids. The mesenchymal marker N-cadherin was significantly up-regulated in A375 3D spheroids while undetectable in BLM cells, but vimentin was similarly expressed in both cell lines at the gene and protein levels. This pattern suggests that A375 cells exhibit a more undifferentiated/mesenchymal phenotype, while BLM cells have more melanocytic/differentiated characteristics. Accordingly, the Zeb1 and 2, Slug, Snail and Twist gene expression analyses showed that they were differentially expressed in 2D monolayers compared to 3D spheroids, supporting this view. Furthermore, A375 cells are characterized by a greater invasive potential, strongly influenced by 3D arrangement, compared to the BLM cell line, as evaluated by SDS-zymography and TIMPs gene expression analysis. Finally, TGF-β1, a master controller of EMT, and lysyl oxidase (LOX), involved in melanoma progression, were strongly up-regulated by 3D arrangement in the metastatic BLM cells alone, likely playing a role in the metastatic phases of melanoma progression. Overall, these findings suggest that A375 and BLM cells possess a hybrid/intermediate phenotype in relation to the expression of EMT markers. The former is characterized by a more mesenchymal/undifferentiated phenotype, while the latter shows a more melanocytic/differentiated phenotype. Our results contribute to the characterization of the role of EMT in melanoma cells and confirm that a 3D cell culture model could provide deeper insight into our understanding of the biology of melanoma.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy; (F.F.); (M.A.); (P.L.)
| | - Michele Sommariva
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (M.S.); (F.B.)
| | - Martina Anselmi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy; (F.F.); (M.A.); (P.L.)
| | - Francesca Bianchi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (M.S.); (F.B.)
- U. O. Laboratorio Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy; (F.F.); (M.A.); (P.L.)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (M.S.); (F.B.)
| |
Collapse
|
36
|
Ghosh A, Ghosh AK, Zaman A, Das PK. Metformin-Loaded Hyaluronic Acid-Derived Carbon Dots for Targeted Therapy against Hepatocellular Carcinoma by Glutamine Metabolic Reprogramming. Mol Pharm 2023; 20:6391-6406. [PMID: 37933877 DOI: 10.1021/acs.molpharmaceut.3c00772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Metabolic reprogramming is a significant hallmark of cancer that promotes chemoresistance by allowing tumor tissues to adapt to changes in the tumor microenvironment caused by anticancer therapies. Hepatocellular carcinoma (HCC), one of the most common types of primary tumors, is associated with recurrent metabolic reprogramming that maximizes cancer cell growth and proliferation. Herein, we developed metformin (MET)-loaded hyaluronic acid (HA)-derived carbon dots (HA-CD-MET) by a simple and green method with no involvement of any additives. HA-CD-MET was utilized for specifically binding the CD44 receptor overexpressed in HCC and induced glutamine metabolic rewiring to inhibit HCC cell proliferation. Exposure to HA-CD-MET resulted in ∼6.5-fold better anticancer efficacy against CD44+ Hep3B cells in comparison to CD44-, HepG2, and noncancerous HEK293 cells at a very low dose of 80 μg/mL. Moreover, treatment of three-dimensional (3D) tumor spheroid model of HCC (Hep3B) with HA-CD-MET resulted in ∼4.9-fold reduction in tumor size. This improved anticancer efficacy of HA-CD-MET was attributed to the inhibition of glutaminase-1 (GLS-1), a mitochondrial enzyme that hydrolyzes glutamine into glutamate as confirmed from immunofluorescence and immunoblotting experiments. Furthermore, treatment with HA-CD-MET resulted in downregulation of glucose transporter-1 (GLUT-1) in Hep3B cells. Consequently, cancer cells were starved from essential nutrients, glutamine, and glucose, leading to the enhancement in intracellular ROS generation. This increase in intracellular ROS accumulation activated AMP-activated protein kinase (AMPK) and inhibited AKT phosphorylation, leading to cancer cell apoptosis. Thus, this study offers the targeting of metabolic reprogramming by HA-CD-MET that opens up a promising strategy for therapeutic intervention in hepatocarcinoma.
Collapse
Affiliation(s)
- Aparajita Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science Jadavpur, Kolkata 700032, India
| | - Anup Kumar Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science Jadavpur, Kolkata 700032, India
| | - Afreen Zaman
- School of Biological Sciences, Indian Association for the Cultivation of Science Jadavpur, Kolkata 700032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science Jadavpur, Kolkata 700032, India
| |
Collapse
|
37
|
Miller CP, Fung M, Jaeger-Ruckstuhl CA, Xu Y, Warren EH, Akilesh S, Tykodi SS. Therapeutic targeting of tumor spheroids in a 3D microphysiological renal cell carcinoma-on-a-chip system. Neoplasia 2023; 46:100948. [PMID: 37944353 PMCID: PMC10663960 DOI: 10.1016/j.neo.2023.100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Metastatic renal cell carcinoma (RCC) remains an incurable disease for most patients highlighting an urgent need for new treatments. However, the preclinical investigation of new therapies is limited by traditional two-dimensional (2D) cultures which do not recapitulate the properties of tumor cells within a collagen extracellular matrix (ECM), while human tumor xenografts are time-consuming, expensive and lack adaptive immune cells. We report a rapid and economical human microphysiological system ("RCC-on-a-chip") to investigate therapies targeting RCC spheroids in a 3D collagen ECM. We first demonstrate that culture of RCC cell lines A498 and RCC4 in a 3D collagen ECM more faithfully reproduces the gene expression program of primary RCC tumors compared to 2D culture. We next used bortezomib as a cytotoxin to develop automated quantification of dose-dependent tumor spheroid killing. We observed that viable RCC spheroids exhibited collective migration within the ECM and demonstrated that our 3D system can be used to identify compounds that inhibit spheroid collective migration without inducing cell death. Finally, we demonstrate the RCC-on-a-chip as a platform to model the trafficking of tumor-reactive T cells into the ECM and observed antigen-specific A498 spheroid killing by engineered human CD8+ T cells expressing an ROR1-specific chimeric antigen receptor. In summary, the phenotypic differences between the 3D versus 2D environments, rapid imaging-based readout, and the ability to carefully study the impact of individual variables with quantitative rigor will encourage adoption of the RCC-on-a-chip system for testing a wide range of emerging therapies for RCC.
Collapse
Affiliation(s)
- Chris P Miller
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.
| | - Megan Fung
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Carla A Jaeger-Ruckstuhl
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Yuexin Xu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Edus H Warren
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States; Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, United States
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States; Kidney Research Institute, University of Washington, Seattle, WA, United States
| | - Scott S Tykodi
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
38
|
Green V, Baldwin L, England J, Marshall G, Frost L, Moore C, Greenman J. Head and Neck Squamous Cell Carcinoma Biopsies Maintained Ex Vivo on a Perfusion Device Show Gene Changes with Time and Clinically Relevant Doses of Irradiation. Cancers (Basel) 2023; 15:4575. [PMID: 37760543 PMCID: PMC10527562 DOI: 10.3390/cancers15184575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Advancements in 3-Dimensional (3D) culture models for studying disease have increased significantly over the last two decades, but fully understanding how these models represent in vivo still requires further investigation. The current study investigated differences in gene expression between a baseline sample and that maintained on a tissue-on-chip perfusion device for up to 96 h, with and without clinically-relevant doses of irradiation, to allow differentiation of model and treatment effects. Tumour tissue samples from 7 Head and Neck Squamous Cell Carcinomas (HNSCC) patients were sub-divided and either fixed immediately upon excision or maintained in a tissue-on-chip device for 48 and 96 h, with or without 2 Gray (Gy) or 10 Gy irradiation. Gene expression was measured using an nCounter® PanCancer Progression Panel. Differentially expressed genes between pre- and post-ex vivo culture, and control and irradiated samples were identified using nSolver software (version 4.0). The secretome from the tumour-on-chip was analysed for the presence of cytokines using a Proteome Profiler™ platform. Significant numbers of genes both increased (n = 6 and 64) and decreased (n = 18 and 58) in expression in the tissue maintained on-chip for 48 and 96 h, respectively, compared to fresh tissue; however, the irradiation schedule chosen did not induce significant changes in gene expression or cytokine secretion. Although HNSCC tissue maintained ex vivo shows a decrease in a large proportion of altered genes, 25% and 53% (48 and 96 h) do show increased expression, suggesting that the tissue remains functional. Irradiation of tumour tissue-on-chip needs to be conducted for longer time periods for specific gene changes to be observed, but we have shown, for the first time, the feasibility of using this perfusion platform for studying the genomic response of HNSCC tissue biopsies.
Collapse
Affiliation(s)
- Victoria Green
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| | - Lydia Baldwin
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| | - James England
- Department of Otorhinolaryngology, Head and Neck Surgery, Hull University Teaching Hospitals NHS Trust Hull, Hull HU16 5JQ, UK;
| | - Gayle Marshall
- Medicines Discovery Catapult Ltd., Alderley Park, Alderley Edge, Cheshire SK10 4TG, UK; (G.M.); (L.F.)
| | - Lucy Frost
- Medicines Discovery Catapult Ltd., Alderley Park, Alderley Edge, Cheshire SK10 4TG, UK; (G.M.); (L.F.)
| | - Craig Moore
- Medical Physics Service, Hull University Teaching Hospitals NHS Trust Hull, Hull HU16 5JQ, UK;
| | - John Greenman
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| |
Collapse
|
39
|
Abbas ZK, Naser NH, Atiya RN. IN SILICO STUDY OF NOVEL SULFONAMIDE DERIVATIVES BEARING A 1, 2, 4-TRIAZOLE MOIETY ACT AS CARBONIC ANHYDRASE INHIBITORS WITH PROMISING ANTI-CANCER ACTIVITY. POLSKI MERKURIUSZ LEKARSKI : ORGAN POLSKIEGO TOWARZYSTWA LEKARSKIEGO 2023; 51:527-532. [PMID: 38069854 DOI: 10.36740/merkur202305112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
OBJECTIVE Aim: To evaluate the theoretical binding affinities of four synthetic compounds that target the carbonic anhydrase IX enzyme in solid tumors. PATIENTS AND METHODS Materials and Methods: To accurately depict the molecular structure, we utilized the Chem Draw Professional 12.0 program. We downloaded the carbonic anhydrase IX enzyme (29.25 KDa) (PDB code: 4YWP) from the Protein Data Bank into the Molecular Operating Environment software. Then, the S-score and rmsd were calculated for the proposed compounds. RESULTS Results: The theoretically synthesized compounds demonstrated good binding affinities with the receptor active pockets Sa, Sb, and Sd, with S-scores of -7.6491, -8.3789, and -8.3218, respectively. Substitutions improve compound orientation. The substituted triazoles ring increases flexibility and receptor interaction. In addition, the benzyl chloride derivatives play an important role in the interaction, with varying effects dependent on the groups substituted at position 4 of the benzene ring. CONCLUSION Conclusions: The synthesized compounds Sb with para Br substitution (S-score = -8.37) and Sd with para Cl substitution (S-score = -8.32) are considered the best ones as they exhibit a high affinity for the receptor.
Collapse
Affiliation(s)
- Zainab Kifah Abbas
- PHARMACEUTICAL CHEMISTRY DEPARTMENT, FACULTY OF PHARMACY, KUFA UNIVERSITY, NAJAF, IRAQ
| | - Noor H Naser
- PHARMACEUTICAL CHEMISTRY DEPARTMENT, COLLEGE OF PHARMACY, AL-ZAHRAA UNIVERSITY FOR WOMEN, KARBALA, IRAQ
| | - Rana Neama Atiya
- PHARMACEUTICAL CHEMISTRY DEPARTMENT, FACULTY OF PHARMACY, KUFA UNIVERSITY, NAJAF, IRAQ
| |
Collapse
|