1
|
Regeni I, Bonnet S. Supramolecular approaches for the treatment of hypoxic regions in tumours. Nat Rev Chem 2025:10.1038/s41570-025-00705-7. [PMID: 40185999 DOI: 10.1038/s41570-025-00705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
Supramolecular chemistry provides a range of 'weak' intermolecular interactions that allow drugs and prodrugs to self-assemble. In the complex biological setting of blood and tumours, these interactions must be stable enough for efficient and selective drug delivery to the tumour site, but weak enough to allow the release of the cytotoxic load. The non-covalent nature of supramolecular interactions enables the detachment of smaller (pro)drug monomers that can penetrate cancer cells differently to the original nanoparticles. Hypoxic tumours show low oxygen levels due to poor vascularization, which poses challenges for drug delivery and generates biological resistances. Supramolecular building blocks specifically designed for hypoxic tumours offer targeted activation of prodrug self-assemblies, enhancing effectiveness against hypoxic cancer cells and hypoxic regions in tumours. This Review explores how supramolecular chemistry can improve (pro)drug delivery and activation in hypoxic tumours.
Collapse
Affiliation(s)
- Irene Regeni
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
2
|
Zhang Z, Sanders HS, Dragun V, Cole S, Smith B. Fluorescent Molecular Probe for Imaging Hypoxia in 2D Cell Culture Monolayers and 3D Tumor Spheroids: The Cell Membrane Partition Model for Predicting Probe Distribution in a Spheroid. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18046-18058. [PMID: 40079788 PMCID: PMC12053547 DOI: 10.1021/acsami.4c22228] [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] [Indexed: 03/15/2025]
Abstract
Compared to cultured 2D cell monolayers, 3D multicellular spheroids are more realistic tumor models. Nonetheless, spheroids remain under-utilized in preclinical research, in part, because there is a lack of fluorescence sensors that can noninvasively interrogate all the individual cells within a spheroid. This present study describes a deep-red fluorogenic molecular probe for microscopic imaging of cells that contain a high level of nitroreductase enzyme activity as a biomarker of cell hypoxia. A first-generation version of the probe produced "turn-on" fluorescence in a 2D cell monolayer under hypoxic conditions; however, it was not useful in a 3D multicellular tumor spheroid because it only accumulated in the peripheral cells. To guide the probe structural optimization process, an intuitive theoretical membrane partition model was conceived to predict how a dosed probe will distribute within a 3D spheroid. The model identifies three limiting molecular diffusion pathways that are determined by a probe's membrane partition properties. A lipophilic probe with high membrane affinity rapidly becomes trapped in the membranes of the peripheral cells. In contrast, a very hydrophilic probe molecule with negligible membrane affinity diffuses rapidly through the spheroid intercellular space and rarely enters the cells. However, a probe molecule with intermediate membrane affinity undergoes sequential diffusion in and out of cells and distributes to all the cells within a spheroid. Using the model as a predictive tool, a second-generation fluorescent probe was prepared with a smaller and more hydrophilic molecular structure, and optical sectioning using structured illumination or light sheet microscopy revealed roughly even probe diffusion throughout a tumor spheroid. The membrane permeation model is likely to be broadly applicable for the structural optimization of various classes of molecules and nanoparticles to enable even distribution within a tumor spheroid.
Collapse
Affiliation(s)
- Zhumin Zhang
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Hailey S. Sanders
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Vivienne Dragun
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Sara Cole
- Notre Dame Integrated Imaging Facility, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley Smith
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
3
|
Garg V, Mathew R, Chatterjee A, Ghosh SK. Protocols for translocation processes of flexible polymers through a pore using LAMMPS. STAR Protoc 2025; 6:103568. [PMID: 39826119 PMCID: PMC11787498 DOI: 10.1016/j.xpro.2024.103568] [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: 05/07/2024] [Revised: 11/23/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Large-scale Atomic/Molecular Massively Parallel Simulator (LAMMPS) is an open-source, powerful simulator with a customizable platform for extensive Langevin dynamics simulations. Here, we present a protocol for using LAMMPS to develop coarse-grained models of polymeric systems with macromolecular crowding, an integral part of any soft matter or biophysical system. We describe steps for installing software, using LAMMPS basic commands and code, and translocating polymers. This protocol has potential applications in developing mathematical models for DNA sequencing, controlled drug delivery, and cellular transport processes. For complete details on the use and execution of this protocol, please refer to Garg et al.1.
Collapse
Affiliation(s)
- Vrinda Garg
- Department of Physics, National Institute of Technology, Warangal 506004, India
| | - Rejoy Mathew
- Department of Physics, National Institute of Technology, Warangal 506004, India
| | - Arindam Chatterjee
- Department of Computer Science and Engineering, IIT Patna, Patna, India; Wipro AI Research, Lab45, Bengaluru, India
| | - Surya K Ghosh
- Department of Physics, National Institute of Technology, Warangal 506004, India.
| |
Collapse
|
4
|
Sánchez-Arribas N, Velasco Rodríguez B, Aicart E, Guerrero-Martínez A, Junquera E, Taboada P. Lipid nanoparticles as nano-Trojan-horses for siRNA delivery and gene-knockdown. J Colloid Interface Sci 2025; 679:975-987. [PMID: 39488022 DOI: 10.1016/j.jcis.2024.10.115] [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: 06/20/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
The therapeutic messenger RNA strategies, such as those using small interfering RNAs, take several advantages (versatility, efficiency and selectivity) over plasmid DNA-based strategies. However, the challenge remains to find nanovectors capable of properly loading the genetic material, transporting it through troublesome environments, like a tumoral site, and delivering it into the cytoplasm of target cells. Here, lipid nanoparticles, consisting of a gemini cationic/neutral helper lipid mixture, are proposed as siRNA nanovector. Cells from cervical and brain cancer overexpressing the green fluorescent protein (GFP) were chosen to analyse the biological response as well as the efficiency and safety of the siRNA-loaded nanovector according to the cell phenotype. Flow cytometry and epifluorescence or confocal microscopy were used to follow the gene knockdown in these overexpressed cells. The effect of the nanovector on cellular proliferation was evaluated with cytotoxicity assays while their potential oxidative stress generation was determined by quantifying the generation of reactive oxygen species. To explore the mechanism of cellular uptake, different inhibitors of endocytic pathways were used during incubation with cells. Finally, nanovectors were incubated in 3D-grown cells (spheroids) to see whether they can penetrate the complex tumoral microenvironments, their efficiency to knockdown GFP expression being monitored by confocal microscopy.
Collapse
Affiliation(s)
- Natalia Sánchez-Arribas
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain
| | - Brenda Velasco Rodríguez
- Departamento de Física de Partículas-Facultad de Física, Instituto de Materiales (IMATUS) e Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Campus Vida, E-15782 Santiago de Compostela, Spain
| | - Emilio Aicart
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain
| | - Andrés Guerrero-Martínez
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain
| | - Elena Junquera
- Dpto. Química Física, Fac. CC. Químicas, Universidad Complutense de Madrid, Av. Complutense s/n, 28040 Madrid, Spain.
| | - Pablo Taboada
- Departamento de Física de Partículas-Facultad de Física, Instituto de Materiales (IMATUS) e Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Campus Vida, E-15782 Santiago de Compostela, Spain.
| |
Collapse
|
5
|
Dhariwal R, Jain M, Mir YR, Singh A, Jain B, Kumar P, Tariq M, Verma D, Deshmukh K, Yadav VK, Malik T. Targeted drug delivery in neurodegenerative diseases: the role of nanotechnology. Front Med (Lausanne) 2025; 12:1522223. [PMID: 39963432 PMCID: PMC11831571 DOI: 10.3389/fmed.2025.1522223] [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: 11/04/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Neurodegenerative diseases, characterized by progressive neuronal loss and cognitive impairments, pose a significant global health challenge. This study explores the potential of nanotherapeutics as a promising approach to enhance drug delivery across physiological barriers, particularly the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (B-CSFB). By employing nanoparticles, this research aims to address critical challenges in the diagnosis and treatment of conditions such as Alzheimer's, Parkinson's, and Huntington's diseases. The multifactorial nature of these disorders necessitates innovative solutions that leverage nanomedicine to improve drug solubility, circulation time, and targeted delivery while minimizing off-target effects. The findings underscore the importance of advancing nanomedicine applications to develop effective therapeutic strategies that can alleviate the burden of neurodegenerative diseases on individuals and healthcare systems.
Collapse
Affiliation(s)
- Rupal Dhariwal
- Research and Development Cell, Parul University, Vadodara, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Mukul Jain
- Research and Development Cell, Parul University, Vadodara, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Yaser Rafiq Mir
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Abhayveer Singh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - Bhavik Jain
- Chitkara Centre for Research and Development, Chitkara University, Baddi, India
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Mohd Tariq
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, India
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Devvret Verma
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Khemraj Deshmukh
- Department of Biomedical Engineering, Parul Institute of Technology, Parul University, Vadodara, India
| | | | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
- Division of Research & Development, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
6
|
Arora S, Singh S, Mittal A, Desai N, Khatri DK, Gugulothu D, Lather V, Pandita D, Vora LK. Spheroids in cancer research: Recent advances and opportunities. J Drug Deliv Sci Technol 2024; 100:106033. [DOI: 10.1016/j.jddst.2024.106033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
|
7
|
Demirbüken SE, Öztürk E, Güngör MA, Garipcan B, Kuralay F. Modified Au:Fe-Ni magnetic micromotors improve drug delivery and diagnosis in MCF-7 cells and spheroids. Colloids Surf B Biointerfaces 2024; 241:114019. [PMID: 38897023 DOI: 10.1016/j.colsurfb.2024.114019] [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: 02/29/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Nano/micromotors hold immense potential for revolutionizing drug delivery and detection systems, especially in the realm of cancer diagnosis and treatment, owing to their distinctive features, including precise propulsion, maneuverability, and meticulously designed surface modifications. In this study, we explore the capabilities of modified and magnetically driven micromotors as active drug delivery systems within 2D and 3D cell culture environments and cancer diagnosis. We synthesized gold (Au) and iron-nickel (Fe-Ni) metallic-based magnetic micromotors (Au:Fe-Ni MMs) through electrochemical methods, equipping them with functionalities for controlled doxorubicin (DOX) release and cancer cell recognition. In 2D and spheroids of MCF-7 adenocarcinoma cells, the Au segment of these micromotors was utilized to help DOX loading through poly(sodium-4-styrenesulfonate) (PSS) functionalization, and the attachment of antiHER2 antibodies for specific recognition. This innovative approach enabled controlled drug release within the cancerous microenvironment, coupled with magnetic (Fe-Ni) propulsion for biocompatible drug delivery to MCF-7 cells. Furthermore, antiHER2 immobilized Au:Fe-Ni MMs effectively interacted with receptors, capitalizing on the overexpression of HER2 antigens on MCF-7 cells. Encouraging outcomes were observed, particularly in spheroid models, underscoring the remarkable potential of these multifunctional micromotors for advancing intelligent drug delivery methodologies and diagnostic purposes.
Collapse
Affiliation(s)
| | - Elif Öztürk
- Department of Chemistry, Faculty of Sciences, Hacettepe University, Ankara 06800, Turkey
| | - Mustafa Ali Güngör
- Department of Chemistry, Faculty of Sciences, Hacettepe University, Ankara 06800, Turkey; Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, Polatlı, Ankara 06900, Turkey
| | - Bora Garipcan
- Institute of Biomedical Engineering, Bogazici University, Istanbul 34684, Turkey.
| | - Filiz Kuralay
- Department of Chemistry, Faculty of Sciences, Hacettepe University, Ankara 06800, Turkey.
| |
Collapse
|
8
|
Mukherjee D, Raikwar S. Recent Update on Nanocarrier(s) as the Targeted Therapy for Breast Cancer. AAPS PharmSciTech 2024; 25:153. [PMID: 38961013 DOI: 10.1208/s12249-024-02867-x] [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: 11/28/2023] [Accepted: 06/11/2024] [Indexed: 07/05/2024] Open
Abstract
Despite ongoing advances in cancer therapy, the results for the treatment of breast cancer are not satisfactory. The advent of nanotechnology promises to be an essential tool to improve drug delivery effectiveness in cancer therapy. Nanotechnology provides an opportunity to enhance the treatment modality by preventing degradation, improving tumour targeting, and controlling drug release. Recent advances have revealed several strategies to prevent cancer metastasis using nano-drug delivery systems (NDDS). These strategies include the design of appropriate nanocarriers loaded with anti-cancer drugs that target the optimization of physicochemical properties, modulate the tumour microenvironment, and target biomimetic techniques. Nanocarriers have emerged as a preferential approach in the chemotropic treatment for breast cancer due to their pivotal role in safeguarding the therapeutic agents against degradation. They facilitate efficient drug concentration in targeted cells, surmount the resistance of drugs, and possess a small size. Nevertheless, these nanocarrier(s) have some limitations, such as less permeability across the barrier and low bioavailability of loaded drugs. To overcome these challenges, integrating external stimuli has been employed, encompassing infrared light, thermal stimulation, microwaves, and X-rays. Among these stimuli, ultrasound-triggered nanocarriers have gained significant attention due to their cost-effectiveness, non-invasive nature, specificity, ability to penetrate tissues, and capacity to deliver elevated drug concentrations to intended targets. This article comprehensively reviews recent advancements in different nanocarriers for breast cancer chemotherapy. It also delves into the associated hurdles and offers valuable insights into the prospective directions for this innovative field.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sarjana Raikwar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
9
|
Li C, Rounds CC, Torres VC, He Y, Xu X, Papavasiliou G, Samkoe KS, Brankov JG, Tichauer KM. Quantifying Imaging Agent Binding and Dissociation in 3-D Cancer Spheroid Tissue Culture Using Paired-Agent Principles. Ann Biomed Eng 2024; 52:1625-1637. [PMID: 38409434 PMCID: PMC10174639 DOI: 10.1007/s10439-024-03476-2] [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: 04/04/2023] [Accepted: 02/10/2024] [Indexed: 02/28/2024]
Abstract
Binding kinetics play an important role in cancer diagnosis and therapeutics. However, current methods of quantifying binding kinetics fail to consider the three-dimensional environment that drugs and imaging agents experience in biological tissue. In response, a methodology to assay agent binding and dissociation in 3-D tissue culture was developed using paired-agent molecular imaging principles. To test the methodology, the uptakes of ABY-029 (an IRDye 800CW-labeled epidermal growth factor receptor (EGFR)-targeted antibody mimetic) and IRDye-700DX carboxylate in 3-D spheroids were measured in four different human cancer cell lines throughout staining and rinsing. A compartment model (optimized for the application) was then fit to the kinetic curves of both imaging agents to estimate binding and dissociation rate constants of the EGFR-targeted ABY-029 agent. A statistically significant correlation was observed between apparent association rate constant (k3) and the receptor concentration experimentally and in simulations (r = 0.99, p < 0.05). A statistically significant difference was found between effective k3 (apparent rate constant of ABY-029 binding to EGFR) values for cell lines with varying levels of EGFR expression (p < 0.05), with no significant difference found between cell lines and controls for other fit parameters. Additionally, a similar binding affinity profile compared to a gold standard method was determined by this model. This low-cost methodology to quantify imaging agent or drug binding affinity in clinically relevant 3-D tumor spheroid models can be used to guide timing of imaging in molecular guided surgery and could have implications in drug development.
Collapse
Affiliation(s)
- Chengyue Li
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Cody C Rounds
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Veronica C Torres
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Yusheng He
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Xiaochun Xu
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Georgia Papavasiliou
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Jovan G Brankov
- Electrical and Computer Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Kenneth M Tichauer
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA.
| |
Collapse
|
10
|
Casadidio C, Hartman JEM, Mesquita B, Haegebaert R, Remaut K, Neumann M, Hak J, Censi R, Di Martino P, Hennink WE, Vermonden T. Effect of Polyplex Size on Penetration into Tumor Spheroids. Mol Pharm 2023; 20:5515-5531. [PMID: 37811785 PMCID: PMC10630948 DOI: 10.1021/acs.molpharmaceut.3c00397] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
Ovarian cancer is one of the most lethal gynecological cancers in the world. In recent years, nucleic acid (NA)-based formulations have been shown to be promising treatments for ovarian cancer, including tumor nodules. However, gene therapy is not that far advanced in clinical reality due to unfavorable physicochemical properties of the NAs, such as high molecular weight, poor cellular uptake, rapid degradation by nucleases, etc. One of the strategies used to overcome these drawbacks is the complexation of anionic NAs via electrostatic interactions with cationic polymers, resulting in the formation of so-called polyplexes. In this work, the role of the size of pDNA and siRNA polyplexes on their penetration into ovarian-cancer-based tumor spheroids was investigated. For this, a methoxypoly(ethylene glycol) poly(2-(dimethylamino)ethyl methacrylate) (mPEG-pDMAEMA) diblock copolymer was synthesized as a polymeric carrier for NA binding and condensation with either plasmid DNA (pDNA) or short interfering RNA (siRNA). When prepared in HEPES buffer (10 mM, pH 7.4) at a nitrogen/phosphate (N/P) charge ratio of 5 and pDNA polyplexes were formed with a size of 162 ± 11 nm, while siRNA-based polyplexes displayed a size of 25 ± 2 nm. The polyplexes had a slightly positive zeta potential of +7-8 mV in the same buffer. SiRNA and pDNA polyplexes were tracked in vitro into tumor spheroids, resembling in vivo avascular ovarian tumor nodules. For this purpose, reproducible spheroids were obtained by coculturing ovarian carcinoma cells with primary mouse embryonic fibroblasts in different ratios (5:2, 1:1, and 2:5). Penetration studies revealed that after 24 h of incubation, siRNA polyplexes were able to penetrate deeper into the homospheroids (composed of only cancer cells) and heterospheroids (cancer cells cocultured with fibroblasts) compared to pDNA polyplexes which were mainly located in the rim. The penetration of the polyplexes was slowed when increasing the fraction of fibroblasts present in the spheroids. Furthermore, in the presence of serum siRNA polyplexes encoding for luciferase showed a high cellular uptake in 2D cells resulting in ∼50% silencing of luciferase expression. Taken together, these findings show that self-assembled small siRNA polyplexes have good potential as a platform to test ovarian tumor nodulus penetration..
Collapse
Affiliation(s)
- Cristina Casadidio
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
- School
of Pharmacy, Drug Delivery Division, University
of Camerino, CHiP Research Center, Via Madonna delle Carceri, 62032 Camerino, Macerata, Italy
| | - Jet E. M. Hartman
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Bárbara
S. Mesquita
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Ragna Haegebaert
- Laboratory
of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Katrien Remaut
- Laboratory
of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Myriam Neumann
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Jaimie Hak
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Roberta Censi
- School
of Pharmacy, Drug Delivery Division, University
of Camerino, CHiP Research Center, Via Madonna delle Carceri, 62032 Camerino, Macerata, Italy
- Recusol
Srl, Via del Bastione
16, 62032 Camerino, Macerata, Italy
| | - Piera Di Martino
- Department
of Pharmacy, “G. D’Annunzio”
University of Chieti and Pescara, Via dei Vestini 1, 66100 Chieti, Chieti, Italy
- Recusol
Srl, Via del Bastione
16, 62032 Camerino, Macerata, Italy
| | - Wim E. Hennink
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
11
|
Vakhshiteh F, Bagheri Z, Soleimani M, Ahvaraki A, Pournemat P, Alavi SE, Madjd Z. Heterotypic tumor spheroids: a platform for nanomedicine evaluation. J Nanobiotechnology 2023; 21:249. [PMID: 37533100 PMCID: PMC10398970 DOI: 10.1186/s12951-023-02021-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/23/2023] [Indexed: 08/04/2023] Open
Abstract
Nanomedicine has emerged as a promising therapeutic approach, but its translation to the clinic has been hindered by the lack of cellular models to anticipate how tumor cells will respond to therapy. Three-dimensional (3D) cell culture models are thought to more accurately recapitulate key features of primary tumors than two-dimensional (2D) cultures. Heterotypic 3D tumor spheroids, composed of multiple cell types, have become more popular than homotypic spheroids, which consist of a single cell type, as a superior model for mimicking in vivo tumor heterogeneity and physiology. The stromal interactions demonstrated in heterotypic 3D tumor spheroids can affect various aspects, including response to therapy, cancer progression, nanomedicine penetration, and drug resistance. Accordingly, to design more effective anticancer nanomedicinal therapeutics, not only tumor cells but also stromal cells (e.g., fibroblasts and immune cells) should be considered to create a more physiologically relevant in vivo microenvironment. This review aims to demonstrate current knowledge of heterotypic 3D tumor spheroids in cancer research, to illustrate current advances in utilizing these tumor models as a novel and versatile platform for in vitro evaluation of nanomedicine-based therapeutics in cancer research, and to discuss challenges, guidelines, and future directions in this field.
Collapse
Affiliation(s)
- Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zeinab Bagheri
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 1983969411, Iran.
| | - Marziye Soleimani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 1983969411, Iran
| | - Akram Ahvaraki
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 1983969411, Iran
| | - Parisa Pournemat
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 1983969411, Iran
| | - Seyed Ebrahim Alavi
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
12
|
Arutyunyan IV, Soboleva AG, Kovtunov EA, Kosyreva AM, Kudelkina VV, Alekseeva AI, Elchaninov AV, Jumaniyazova ED, Goldshtein DV, Bolshakova GB, Fatkhudinov TK. Gene Expression Profile of 3D Spheroids in Comparison with 2D Cell Cultures and Tissue Strains of Diffuse High-Grade Gliomas. Bull Exp Biol Med 2023; 175:576-584. [PMID: 37770789 DOI: 10.1007/s10517-023-05906-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 09/30/2023]
Abstract
The use of relevant, accessible, and easily reproducible preclinical models of diffuse gliomas is a prerequisite for the development of successful therapeutic approaches to their treatment. Here we studied the gene expression profile of 3D spheroids in a comparison with 2D cell cultures and tissue strains of diffuse high-grade gliomas. Using real time PCR, we evaluated the expression of Gfap, Cd44, Pten, S100b, Vegfa, Hif1a, Sox2, Melk, Gdnf, and Mgmt genes playing an important role in the progression of gliomas and regulating tumor cell proliferation, adhesion, invasion, plasticity, apoptosis, DNA repair, and recruitment of tumor-associated cells. Gene expression analysis showed that 3D spheroids are more similar to tumor tissue strains by the expression levels of Gfap, Cd44, and Pten, while the expression levels of Hif1a and Sox2 in 3D spheroids did not differ from those of 2D cell cultures, the expression levels S100b and Vegfa in 3D spheroids was higher than in other models, and the expression levels of Melk, Gdnf, and Mgmt genes changed diversely. Thus, 3D spheroid model more closely mimics the tumor tissue than 2D cell culture, but still is not the most relevant, probably due to too small size of spheroids, which does not allow reproducing hypoxia and apoptotic and necrotic processes in the tumor tissue.
Collapse
Affiliation(s)
- I V Arutyunyan
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia.
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia.
| | - A G Soboleva
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - E A Kovtunov
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia
| | - A M Kosyreva
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - V V Kudelkina
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - A I Alekseeva
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - A V Elchaninov
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - E D Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia
| | - D V Goldshtein
- N. P. Bochkov Research Centre for Medical Genetics, Moscow, Russia
| | - G B Bolshakova
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - T Kh Fatkhudinov
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Institute of Medicine, Peoples' Friendship, University of Russia, RUDN University), Moscow, Russia
- A. P. Avtsyn Research Institute of Human Morphology, B. V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| |
Collapse
|
13
|
Lee S, Kim S, Kim D, You J, Kim JS, Kim H, Park J, Song J, Choi I. Spatiotemporally controlled drug delivery via photothermally driven conformational change of self-integrated plasmonic hybrid nanogels. J Nanobiotechnology 2023; 21:191. [PMID: 37316900 DOI: 10.1186/s12951-023-01935-x] [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: 02/28/2023] [Accepted: 05/18/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Spatiotemporal regulation is one of the major considerations for developing a controlled and targeted drug delivery system to treat diseases efficiently. Light-responsive plasmonic nanostructures take advantage due to their tunable optical and photothermal properties by changing size, shape, and spatial arrangement. RESULTS In this study, self-integrated plasmonic hybrid nanogels (PHNs) are developed for spatiotemporally controllable drug delivery through light-driven conformational change and photothermally-boosted endosomal escape. PHNs are easily synthesized through the simultaneous integration of gold nanoparticles (GNPs), thermo-responsive poly (N-isopropyl acrylamide), and linker molecules during polymerization. Wave-optic simulations reveal that the size of the PHNs and the density of the integrated GNPs are crucial factors in modulating photothermal conversion. Several linkers with varying molecular weights are inserted for the optimal PHNs, and the alginate-linked PHN (A-PHN) achieves more than twofold enhanced heat conversion compared with others. Since light-mediated conformational changes occur transiently, drug delivery is achieved in a spatiotemporally controlled manner. Furthermore, light-induced heat generation from cellular internalized A-PHNs enables pinpoint cytosolic delivery through the endosomal rupture. Finally, the deeper penetration for the enhanced delivery efficiency by A-PHNs is validated using multicellular spheroid. CONCLUSION This study offers a strategy for synthesizing light-responsive nanocarriers and an in-depth understanding of light-modulated site-specific drug delivery.
Collapse
Affiliation(s)
- Seungki Lee
- Department of Life Science, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-Gu, Seoul, 02504, Republic of Korea
| | - Subeen Kim
- Department of Mechanical Engineering, Hanbat National University, 125 Dongseodaero, Yuseong-Gu, Daejeon, 34158, Republic of Korea
| | - Doyun Kim
- Department of Life Science, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-Gu, Seoul, 02504, Republic of Korea
| | - Jieun You
- Department of Life Science, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-Gu, Seoul, 02504, Republic of Korea
| | - Ji Soo Kim
- School of Chemical and Biological Engineering, Institute of Chemical Process, Seoul National University, 1 Gwanakro, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Hakchun Kim
- Department of Life Science, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-Gu, Seoul, 02504, Republic of Korea
| | - Jungwon Park
- School of Chemical and Biological Engineering, Institute of Chemical Process, Seoul National University, 1 Gwanakro, Gwanak-Gu, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Jihwan Song
- Department of Mechanical Engineering, Hanbat National University, 125 Dongseodaero, Yuseong-Gu, Daejeon, 34158, Republic of Korea.
| | - Inhee Choi
- Department of Life Science, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-Gu, Seoul, 02504, Republic of Korea.
- Department of Applied Chemistry, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-Gu, Seoul, 02504, Republic of Korea.
| |
Collapse
|
14
|
Saemundsson SA, Ganguly S, Curry SD, Goodwin AP, Cha JN. Controlling Cell Organization in 3D Coculture Spheroids Using DNA Interactions. ACS Biomater Sci Eng 2023. [PMID: 37155244 DOI: 10.1021/acsbiomaterials.3c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The role of stromal and immune cells in transforming the tumor microenvironment is a key consideration in understanding tumor cell behavior and anticancer drug development. To better model these systems in vitro, 3D coculture tumor spheroids have been engineered using a variety of techniques including centrifugation to microwells, hanging drop, low adhesion cultures, and culture of cells in a microfluidic platform. Aside from using bioprinting, however, it has remained more challenging to direct the spatial organization of heterotypic cells in standalone 3D spheroids. To address this, we present an in vitro 3D coculture tumor model where we modulated the interactions between cancer cells and fibroblasts through DNA hybridization. When native heterotypic cells are simply mixed, the cell aggregates typically show cell sorting behavior to form phase separated structures composed of single cell types. In this work, we demonstrate that when MDA-MB-468 breast cancer and NIH/3T3 fibroblasts are directed to associate via complementary DNA, a uniform distribution of the two cell types within a single spheroid was observed. In contrast, in the absence of specific DNA interactions between the cancer cells and fibroblasts, individual clusters of the NIH/3T3 cells formed in each spheroid due to cell sorting. To better understand the effect of heterotypic cell organization on either cell-cell contacts or matrix protein production, the spheroids were further stained with anti-E-cadherin and antifibronectin antibodies. While the amounts of E-cadherin appeared to be similar between the spheroids, a significantly higher amount of fibronectin secretion was observed in the coculture spheroids with uniform mixing of two cell types. This result showed that different heterotypic cell distributions within 3D architecture can influence the ECM protein production that can again alter the properties of the tumor or tumor microenvironment. The present study thus describes the use of DNA templating to direct the organization of cells in coculture spheroids, which can provide mechanistic biological insight into how heterotypic distribution in tumor spheroids can influence tumor progression, metastasis, and drug resistance.
Collapse
Affiliation(s)
- Sven A Saemundsson
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Saheli Ganguly
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Shane D Curry
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Andrew P Goodwin
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
- Materials Science and Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| | - Jennifer N Cha
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
- Materials Science and Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
- Biomedical Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, Colorado 80303, United States
| |
Collapse
|
15
|
Barman S, Chakraborty A, Saha S, Sikder K, Maitra Roy S, Modi B, Bahadur S, Khan AH, Manna D, Bag P, Sarkar AK, Bhattacharya R, Basu A, Maity AR. Efficient Synergistic Antibacterial Activity of α-MSH Using Chitosan-Based Versatile Nanoconjugates. ACS OMEGA 2023; 8:12865-12877. [PMID: 37065019 PMCID: PMC10099120 DOI: 10.1021/acsomega.2c08209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
The application of antimicrobial peptides has emerged as an alternative therapeutic tool to encounter against multidrug resistance of different pathogenic organisms. α-Melanocyte stimulating hormone (α-MSH), an endogenous neuropeptide, is found to be efficient in eradicating infection of various kinds of Staphylococcus aureus, including methicillin-resistant Staphylococcus aureus (MRSA). However, the chemical stability and efficient delivery of these biopharmaceuticals (i.e., α-MSH) to bacterial cells with a significant antibacterial effect remains a key challenge. To address this issue, we have developed a chitosan-cholesterol polymer using a single-step, one-pot, and simple chemical conjugation technique, where α-MSH is loaded with a significantly high amount (37.7%), and the final product is obtained as chitosan-cholesterol α-MSH polymer-drug nanoconjugates. A staphylococcal growth inhibition experiment was performed using chitosan-cholesterol α-MSH and individual controls. α-MSH and chitosan-cholesterol both show bacterial growth inhibition by a magnitude of 50 and 79%, respectively. The killing efficiency of polymer-drug nanoconjugates was very drastic, and almost no bacterial colony was observed (∼100% inhibition) after overnight incubation. Phenotypic alternation was observed in the presence of α-MSH causing changes in the cell structure and shape, indicating stress on Staphylococcus aureus. As a further consequence, vigorous cell lysis with concomitant release of the cellular material in the nearby medium was observed after treatment of chitosan-cholesterol α-MSH nanoconjugates. This vigorous lysis of the cell structure is associated with extensive aggregation of the bacterial cells evident in scanning electron microscopy (SEM). The dose-response experiment was performed with various concentrations of chitosan-cholesterol α-MSH nanoconjugates to decipher the degree of the bactericidal effect. The concentration of α-MSH as low as 1 pM also shows significant inhibition of bacterial growth (∼40% growth inhibition) of Staphylococcus aureus. Despite playing an important role in inhibiting bacterial growth, our investigation on hemolytic assay shows that chitosan-cholesterol α-MSH is significantly nontoxic at a wide range of concentrations. In a nutshell, our analysis demonstrated novel antimicrobial activity of nanoparticle-conjugated α-MSH, which could be used as future therapeutics against multidrug-resistant Staphylococcus aureus and other types of bacterial cells.
Collapse
Affiliation(s)
- Sourav Barman
- Amity
Institute of Biotechnology, Amity University, Kolkata, West Bengal 700135, India
| | - Asmita Chakraborty
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Sujata Saha
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Kunal Sikder
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Sayoni Maitra Roy
- Amity
Institute of Biotechnology, Amity University, Kolkata, West Bengal 700135, India
| | - Barkha Modi
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Sabarnee Bahadur
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Ali Hossain Khan
- S.
N. Bose National Centre for Basic Sciences, Kolkata, West Bengal 700106, India
| | - Dipak Manna
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Pousali Bag
- Amity
Institute of Biotechnology, Amity University, Kolkata, West Bengal 700135, India
| | - Ankan Kumar Sarkar
- School
of Materials Sciences, Indian Association
for the Cultivation of Science, Kolkata, West Bengal 700032, India
| | - Rishi Bhattacharya
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Arnab Basu
- Department
of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research
Institute, Belur Math, Howrah, West
Bengal 711202, India
| | - Amit Ranjan Maity
- Amity
Institute of Biotechnology, Amity University, Kolkata, West Bengal 700135, India
| |
Collapse
|
16
|
Martins C, Pacheco C, Moreira-Barbosa C, Marques-Magalhães Â, Dias S, Araújo M, Oliveira MJ, Sarmento B. Glioblastoma immuno-endothelial multicellular microtissue as a 3D in vitro evaluation tool of anti-cancer nano-therapeutics. J Control Release 2023; 353:77-95. [PMID: 36410614 DOI: 10.1016/j.jconrel.2022.11.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/27/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
Despite being the most prevalent and lethal type of adult brain cancer, glioblastoma (GBM) remains intractable. Promising anti-GBM nanoparticle (NP) systems have been developed to improve the anti-cancer performance of difficult-to-deliver therapeutics, with particular emphasis on tumor targeting strategies. However, current disease modeling toolboxes lack close-to-native in vitro models that emulate GBM microenvironment and bioarchitecture, thus partially hindering translation due to poorly predicted clinical responses. Herein, human GBM heterotypic multicellular tumor microtissues (MCTMs) are generated through high-throughput 3D modeling of U-251 MG tumor cells, tissue differentiated macrophages isolated from peripheral monocytes, and brain microvascular primary endothelial cells. GBM MCTMs mimicked tumor spatial organization, extracellular matrix production and necrosis areas. The bioactivity of a model drug, docetaxel (DTX), and of tumor-targeted DTX-loaded polymeric NPs with a surface L-Histidine moiety (H-NPs), were assessed in the MCTMs. MCTMs cell uptake and anti-proliferative effect was 8- and 3-times higher for H-NPs, respectively, compared to the non-targeted NPs and to free DTX. H-NPs provided a decrease of MCTMs anti-inflammatory M2-macrophages, while increasing their pro-inflammatory M1 counterparts. Moreover, H-NPs showed a particular biomolecular signature through reduced secretion of an array of medium cytokines (IFN-γ, IL-1β, IL-1Ra, IL-6, IL-8, TGF-β). Overall, MCTMs provide an in vitro biomimetic model to recapitulate key cellular and structural features of GBM and improve in vivo drug response predictability, fostering future clinical translation of anti-GBM nano-therapeutic strategies.
Collapse
Affiliation(s)
- Cláudia Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Catarina Pacheco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Moreira-Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Ângela Marques-Magalhães
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sofia Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Marco Araújo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal
| | - Maria J Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
17
|
Vinod D, Cherstvy AG, Metzler R, Sokolov IM. Time-averaging and nonergodicity of reset geometric Brownian motion with drift. Phys Rev E 2022; 106:034137. [PMID: 36266856 DOI: 10.1103/physreve.106.034137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/09/2022] [Indexed: 06/16/2023]
Abstract
How do near-bankruptcy events in the past affect the dynamics of stock-market prices in the future? Specifically, what are the long-time properties of a time-local exponential growth of stock-market prices under the influence of stochastically occurring economic crashes? Here, we derive the ensemble- and time-averaged properties of the respective "economic" or geometric Brownian motion (GBM) with a nonzero drift exposed to a Poissonian constant-rate price-restarting process of "resetting." We examine-based both on thorough analytical calculations and on findings from systematic stochastic computer simulations-the general situation of reset GBM with a nonzero [positive] drift and for all special cases emerging for varying parameters of drift, volatility, and reset rate in the model. We derive and summarize all short- and long-time dependencies for the mean-squared displacement (MSD), the variance, and the mean time-averaged MSD (TAMSD) of the process of Poisson-reset GBM under the conditions of both rare and frequent resetting. We consider three main regions of model parameters and categorize the crossovers between different functional behaviors of the statistical quantifiers of this process. The analytical relations are fully supported by the results of computer simulations. In particular, we obtain that Poisson-reset GBM is a nonergodic stochastic process, with generally MSD(Δ)≠TAMSD(Δ) and Variance(Δ)≠TAMSD(Δ) at short lag times Δ and for long trajectory lengths T. We investigate the behavior of the ergodicity-breaking parameter in each of the three regions of parameters and examine its dependence on the rate of reset at Δ/T≪1. Applications of these theoretical results to the analysis of prices of reset-containing options are pertinent.
Collapse
Affiliation(s)
- Deepak Vinod
- Institute for Physics & Astronomy, University of Potsdam, Karl-Liebknecht-Straße 24/25, 14476 Potsdam-Golm, Germany
| | - Andrey G Cherstvy
- Institute for Physics & Astronomy, University of Potsdam, Karl-Liebknecht-Straße 24/25, 14476 Potsdam-Golm, Germany
- Institut für Physik, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Ralf Metzler
- Institute for Physics & Astronomy, University of Potsdam, Karl-Liebknecht-Straße 24/25, 14476 Potsdam-Golm, Germany
| | - Igor M Sokolov
- Institut für Physik, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
- IRIS Adlershof, Zum Großen Windkanal 6, 12489 Berlin, Germany
| |
Collapse
|
18
|
Roy SM, Barman S, Basu A, Ghatak T, Pore SK, Ghosh SK, Mukherjee R, Maity AR. Amine as a bottom-line functionality on DDS surface for efficient endosomal escape and further subcellular targets. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
Lotsberg ML, Røsland GV, Rayford AJ, Dyrstad SE, Ekanger CT, Lu N, Frantz K, Stuhr LEB, Ditzel HJ, Thiery JP, Akslen LA, Lorens JB, Engelsen AST. Intrinsic Differences in Spatiotemporal Organization and Stromal Cell Interactions Between Isogenic Lung Cancer Cells of Epithelial and Mesenchymal Phenotypes Revealed by High-Dimensional Single-Cell Analysis of Heterotypic 3D Spheroid Models. Front Oncol 2022; 12:818437. [PMID: 35530312 PMCID: PMC9076321 DOI: 10.3389/fonc.2022.818437] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/22/2022] [Indexed: 11/30/2022] Open
Abstract
The lack of inadequate preclinical models remains a limitation for cancer drug development and is a primary contributor to anti-cancer drug failures in clinical trials. Heterotypic multicellular spheroids are three-dimensional (3D) spherical structures generated by self-assembly from aggregates of two or more cell types. Compared to traditional monolayer cell culture models, the organization of cells into a 3D tissue-like structure favors relevant physiological conditions with chemical and physical gradients as well as cell-cell and cell-extracellular matrix (ECM) interactions that recapitulate many of the hallmarks of cancer in situ. Epidermal growth factor receptor (EGFR) mutations are prevalent in non-small cell lung cancer (NSCLC), yet various mechanisms of acquired resistance, including epithelial-to-mesenchymal transition (EMT), limit the clinical benefit of EGFR tyrosine kinase inhibitors (EGFRi). Improved preclinical models that incorporate the complexity induced by epithelial-to-mesenchymal plasticity (EMP) are urgently needed to advance new therapeutics for clinical NSCLC management. This study was designed to provide a thorough characterization of multicellular spheroids of isogenic cancer cells of various phenotypes and demonstrate proof-of-principle for the applicability of the presented spheroid model to evaluate the impact of cancer cell phenotype in drug screening experiments through high-dimensional and spatially resolved imaging mass cytometry (IMC) analyses. First, we developed and characterized 3D homotypic and heterotypic spheroid models comprising EGFRi-sensitive or EGFRi-resistant NSCLC cells. We observed that the degree of EMT correlated with the spheroid generation efficiency in monocultures. In-depth characterization of the multicellular heterotypic spheroids using immunohistochemistry and high-dimensional single-cell analyses by IMC revealed intrinsic differences between epithelial and mesenchymal-like cancer cells with respect to self-sorting, spatiotemporal organization, and stromal cell interactions when co-cultured with fibroblasts. While the carcinoma cells harboring an epithelial phenotype self-organized into a barrier sheet surrounding the fibroblasts, mesenchymal-like carcinoma cells localized to the central hypoxic and collagen-rich areas of the compact heterotypic spheroids. Further, deep-learning-based single-cell segmentation of IMC images and application of dimensionality reduction algorithms allowed a detailed visualization and multiparametric analysis of marker expression across the different cell subsets. We observed a high level of heterogeneity in the expression of EMT markers in both the carcinoma cell populations and the fibroblasts. Our study supports further application of these models in pre-clinical drug testing combined with complementary high-dimensional single-cell analyses, which in turn can advance our understanding of the impact of cancer-stroma interactions and epithelial phenotypic plasticity on innate and acquired therapy resistance in NSCLC.
Collapse
Affiliation(s)
- Maria L. Lotsberg
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Gro V. Røsland
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Austin J. Rayford
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- BerGenBio, Bergen, Norway
| | - Sissel E. Dyrstad
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Camilla T. Ekanger
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Ning Lu
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kirstine Frantz
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Linda E. B. Stuhr
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Henrik J. Ditzel
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Jean Paul Thiery
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Guangzhou Laboratory, Guangzhou, China
- Gustave Roussy Cancer Campus, UMR 1186, Inserm, Université Paris-Saclay, Villejuif, France
| | - Lars A. Akslen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, Section for Pathology, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - James B. Lorens
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Agnete S. T. Engelsen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|