1
|
Kim S, Kim S, Ko C, Lee W, Kim HD. A microfluidic electrochemical immunosensor for detection of CEA and Ki67 in 3D tumor spheroids. Mater Today Bio 2025; 32:101768. [PMID: 40290895 PMCID: PMC12022681 DOI: 10.1016/j.mtbio.2025.101768] [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: 02/04/2025] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Microfluidic chip-based electrochemical sensors have been developed to detect cancer biomarkers and monitor changes in the tumor microenvironment. However, the limitation of detecting only a single biomarker restricts their utility as accurate diagnostic tools. Simultaneous detection of multiple tumor biomarkers is important for early diagnosis of cancer. In this work, we report the development of a microfluidic-based electrochemical immunosensor platform capable of simultaneously observing multiple biomarkers expressed by three dimensions (3D) cell spheroids. The sensor platform employs a nanocomposite electrode material consisting of gold nanoparticles and carbon nanotubes, which enables sensitive and selective detection. The sensor was fabricated using 3D and printed circuit boards (PCB) printing techniques, demonstrating the feasibility of cost-effective manufacturing. The developed platform was able to quantitatively detect two key cancer biomarkers, carcinoembryonic antigen (CEA) and Ki67, with limits of detection of 0.97 ng/mL for each. Furthermore, the sensor was successfully utilized to observe the knockdown of these biomarkers, showcasing its potential for both diagnostic and therapeutic monitoring applications. These results suggest that the presented electrochemical sensor platform provides a promising lab-on-a-chip technology for comprehensive 3D cell spheroid-based cancer biomarker analysis, which could have significant implications for future clinical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Seonyeop Kim
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Chanjin Ko
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Wonseok Lee
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
- Department of Electrical Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Hwan Drew Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| |
Collapse
|
2
|
Feng J, Ponomareva P, Liu K, Nie C, Chen R, Haag R. Dual-Dynamic Covalently Cross-Linked Polyglycerol Hydrogels for Tumor Spheroid Culture. Biomacromolecules 2025. [PMID: 40347130 DOI: 10.1021/acs.biomac.4c01744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
Advancing cancer research depends significantly on developing accurate and reliable models that can replicate the complex tumor microenvironment. Tumor spheroids─three-dimensional clusters of cancer cells─have become crucial tools for this purpose. The overarching goal of tumor spheroid culture is to develop biomaterials that mimic the dynamic mechanical behavior of the native extracellular matrix, enabling high-fidelity culture models. In this study, we developed dynamic hydrogels based on dual-dynamic covalently cross-linked polyglycerol, using boronate bonds and Schiff-base interactions. In addition to good biocompatibility and long-term stability, the hydrogels showed tunable mechanical properties that enabled cells to actively remodel their surrounding microenvironment. This platform was used for successful 3D culture of various cancer cell lines, including HeLa, A549, HT-29, BT-474, and SK-BR-3, which were encapsulated in situ and formed 3D tumor spheroids. These results demonstrate the feasibility and versatility of our dynamic hydrogel system in supporting tumor spheroid culture.
Collapse
Affiliation(s)
- Jun Feng
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Polina Ponomareva
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Kunpeng Liu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Chuanxiong Nie
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Rui Chen
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| |
Collapse
|
3
|
Xu X, Dai X, Huang C, Guan X, Zhang C. 17beta-estradiol (E2) Regulates Malignancies and Stemness in Endometrial Carcinoma (EC) via Interacting with ESR1. Reprod Sci 2025:10.1007/s43032-025-01871-1. [PMID: 40342082 DOI: 10.1007/s43032-025-01871-1] [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: 02/20/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025]
Abstract
Endometrial cancer is one of the most common and fatal gynaecologic malignancies and is associated with the presence of estrogen. Endometrial cancer stem-like cells (ECSCs) are stem-like cell subpopulations endowed with self-renewal and differentiation capacities and are critical for EC progression. However, it is still unknown whether estrogen is involved in regulating stemness in EC and contributes to malignancies. Therefore, we investigated the regulatory effects of E2 treatment on oestrogen receptor-1 (ESR1).ESR1 expression was measured in EC tissues; After ESR1 overexpression of knockdown, the effects of E2 treatment was evaluated, including cell proliferation, sphere formation, invasion and colony formation. Our results indicate that ESR1 is critical for the regulatory effect of E2 on EC cells. After being cultured in serum-free medium, the ECSCs were found to be enriched in stemness markers, including CD133, CD44 and ALDH1. Interestingly, enriched ECSCs presented significantly lower expression levels of these factors than the parental cells. Overexpression of ESR1 slightly affected stemness and malignant potential in ECSCs, and the addition of 2 nM E2 markedly decreased stemness and inhibited malignant behaviours of ECSCs, including proliferation, invasion and tumour formation, in soft agar. Moreover, the addition of E2 significantly inhibited the expression of epithelial-mesenchymal transition (EMT) markers, including ZEB1, ZEB2, and E-cadherin.ESR1 is critical for regulating EC progression in the presence of E2, partially by regulating stemness in ECSCs, indicating that ESR1 might be a potential therapeutic target for EC.
Collapse
Affiliation(s)
- Xiaochao Xu
- College of Food and Biological Engineering, Chengdu University, Chengdu, 610000, Sichuan, China
| | - Xinzhi Dai
- The First Clinical lnstitute, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Cheng Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Precision Pathology Diagnosis for Serious Diseases Key Laboratory of LuZhou, Luzhou, 646000, Sichuan, China
| | - Xia Guan
- Department of Pathology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610000, Sichu, P.R. China.
| | - Cuiwei Zhang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Precision Pathology Diagnosis for Serious Diseases Key Laboratory of LuZhou, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
4
|
Vuille-Dit-Bille E, Fonta CL, Heub S, Boder-Pasche S, Sakar MS, Weder G. An acoustic levitation platform for high-content histological analysis of 3D tissue culture. LAB ON A CHIP 2025. [PMID: 40326244 PMCID: PMC12053053 DOI: 10.1039/d5lc00153f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Miniaturized three-dimensional (3D) cell culture systems, in particular organoids and spheroids, hold great potential for studying morphogenesis, disease modeling, and drug discovery. However, sub-cellular resolution 3D imaging of these biological samples remains a challenge. Histology, the gold standard for ex vivo microscopic interrogation of tissue anatomy, may address this challenge once the associated techniques are adapted. Due to their small size and delicate structure, organoids must be embedded in a supporting hydrogel. The histological sections have low information content because the distribution of the organoids within the gel is not controlled. To address this issue, we introduce an acoustic micromanipulation platform that concentrates and aligns organoids within a histology-compatible hydrogel block. Utilizing an array of micromachined lead zirconate titanate (PZT) transducers, the platform generates localised and precisely controlled acoustic standing waves to levitate organoids to a prescribed plane and fix their positions within a polyethylene glycol diacrylate (PEGDA)-gelatine hydrogel. Organoids from different culture conditions can be co-embedded in a traceable fashion with the use of a custom-design hydrogel grid. Our results demonstrate that more than 70% of spheroids can be positioned within a 150 μm-thick hydrogel block, substantially increasing the information content of histology sections. The platform's versatility, scalability, and ease of use will make histological assessment accessible to every life science laboratory.
Collapse
Affiliation(s)
- Emilie Vuille-Dit-Bille
- CSEM SA, Neuchâtel, Switzerland.
- Institute of Mechanical Engineering, EPFL, Lausanne, Switzerland.
| | | | | | | | | | | |
Collapse
|
5
|
Thomas G, Rahman R. Evolution of Preclinical Models for Glioblastoma Modelling and Drug Screening. Curr Oncol Rep 2025; 27:601-624. [PMID: 40183896 DOI: 10.1007/s11912-025-01672-4] [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] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW Isocitrate dehydrogenase wild-type glioblastoma is an extremely aggressive and fatal primary brain tumour, characterised by extensive heterogeneity and diffuse infiltration of brain parenchyma. Despite multimodal treatment and diverse research efforts to develop novel therapies, there has been limited success in improving patient outcomes. Constructing physiologically relevant preclinical models is essential to optimising drug screening processes and identifying more effective treatments. RECENT FINDINGS Traditional in-vitro models have provided critical insights into glioblastoma pathophysiology; however, they are limited in their ability to recapitulate the complex tumour microenvironment and its interactions with surrounding cells. In-vivo models offer a more physiologically relevant context, but often do not fully represent human pathology, are expensive, and time-consuming. These limitations have contributed to the low translational success of therapies from trials to clinic. Organoid and glioblastoma-on-a-chip technology represent significant advances in glioblastoma modelling and enable the replication of key features of the human tumour microenvironment, including its structural, mechanical, and biochemical properties. Organoids provide a 3D system that captures cellular heterogeneity and tumour architecture, while microfluidic chips offer dynamic systems capable of mimicking vascularisation and nutrient exchange. Together, these technologies hold tremendous potential for high throughput drug screening and personalised, precision medicine. This review explores the evolution of preclinical models in glioblastoma modelling and drug screening, emphasising the transition from traditional systems to more advanced organoid and microfluidic platforms. Furthermore, it aims to evaluate the advantages and limitations of both traditional and next-generation models, investigating their combined potential to address current challenges by integrating complementary aspects of specific models and techniques.
Collapse
Affiliation(s)
- Grace Thomas
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ruman Rahman
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
6
|
Oishi K, Matsumoto K, Hashimoto S, Uchida F, Hara R, Nishimuta M, Matsumoto T, Iwatake M, Tomoshige K, Doi R, Machino R, Obata T, Nagayasu T. Spheroid morphology of lung cancer cell lines correlates with oncological profiles. Discov Oncol 2025; 16:627. [PMID: 40293538 PMCID: PMC12037941 DOI: 10.1007/s12672-025-02132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
We assessed the correlation between Multicellular tumor spheroids (MCTS) morphology and the oncological profile of lung cancer cells. MCTS were generated in five lung cancer cell lines and classified into Type-A MCTS, which showed strong aggregation, and Type-B MCTS, which showed weak aggregation. Drug resistance was higher in Type-A MCTS, and invasive ability was higher in Type-B MCTS. The oncologic profile of lung cancer cell lines correlated with MCTS morphology. MCTS morphology could thus be used in basic oncology research and as a clinical prognostic tool.Registry and the Registration No. of the study/trial Not Applicable.
Collapse
Affiliation(s)
- Kaido Oishi
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan.
| | - Keitaro Matsumoto
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan.
| | - Shintaro Hashimoto
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Fumitake Uchida
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Ryosuke Hara
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Masato Nishimuta
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Takamune Matsumoto
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Mayumi Iwatake
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Koichi Tomoshige
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Ryoichiro Doi
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Ryusuke Machino
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Tomohiro Obata
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| | - Takeshi Nagayasu
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Medical-Engineering Hybrid Professional Development Program, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
7
|
Chen M, Huang L, Zhao S, Zhu M, Sun S, Li W, Cai J, Peng M, Wen Y, Wang Z. Ivosidenib enhances cisplatin sensitivity in ovarian cancer by reducing cancer cell stemness. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:20. [PMID: 40342735 PMCID: PMC12059478 DOI: 10.20517/cdr.2025.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Aim: Cancer stem cells (CSCs) are pivotal in mediating platinum resistance in ovarian cancer. This study aimed to screen compounds sensitizing CSCs to cisplatin by using a small molecule inhibitor library. Methods: A library of 105 common drugs was screened in ovarian CSC model SK-3rd and ovarian cancer platinum-resistant cell model SKDDP to identify those that could enhance sensitivity to cisplatin by MTT assay. The antitumor effect was assessed in ovarian cancer cells using the MTT assay, colony formation assay, and apoptosis assay. The impact on cancer cell stemness was evaluated using qPCR and Sphere-forming assays. Finally, the effect of the combination regimen was evaluated in patient-derived organoids (PDOs) under different treatments by the CellTiter-Glo Luminescence Assay. Results: The results of the initial screening on SK-3rd identified five candidate compounds. Rescreening on SKDDP showed that Ivosidenib was the most effective in sensitizing cisplatin. MTT, colony formation, and apoptosis assays demonstrated that Ivosidenib enhanced the sensitivity to cisplatin, inhibited proliferation, and induced apoptosis in ovarian cancer cells, including SK-3rd and SKDDP. Furthermore, Ivosidenib lowered stemness marker expression and countered CSC enrichment caused by platinum-based chemotherapy in ovarian cancer cells. Finally, the synergistic effect of this combination was also confirmed in three ovarian cancer PDOs. Conclusion: Ivosidenib may increase cisplatin sensitivity in ovarian cancer cells by decreasing their stemness, providing a potential therapeutic method for ovarian cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Minggang Peng
- Correspondence to: Minggang Peng, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China. E-mail: ; Yiping Wen, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China. E-mail: ; Zehua Wang, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, Hubei, China. E-mail:
| | - Yiping Wen
- Correspondence to: Minggang Peng, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China. E-mail: ; Yiping Wen, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China. E-mail: ; Zehua Wang, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, Hubei, China. E-mail:
| | - Zehua Wang
- Correspondence to: Minggang Peng, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China. E-mail: ; Yiping Wen, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China. E-mail: ; Zehua Wang, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan 430022, Hubei, China. E-mail:
| |
Collapse
|
8
|
Awale S, Maneenet J, Phan ND, Nguyen HH, Fujii T, Ihmels H, Soost D, Tajuddeen N, Feineis D, Bringmann G. Toyaburgine, a Synthetic N-Biphenyl-Dihydroisoquinoline Inspired by Related N, C-Coupled Naphthylisoquinoline Alkaloids, with High In Vivo Efficacy in Preclinical Pancreatic Cancer Models. ACS Chem Biol 2025; 20:917-929. [PMID: 40048237 DOI: 10.1021/acschembio.4c00870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Pancreatic cancer is a highly aggressive and lethal malignancy, with a 5-year survival rate below 10%. Traditional chemotherapy, including gemcitabine, has limited efficacy due to chemoresistance and a unique tumor microenvironment characterized by hypovascularity and nutrient deprivation. This study reports on the discovery of a new N-biphenyl-dihydroisoquinoline, named toyaburgine (4), inspired by naturally occurring N,C-coupled naphthylisoquinoline alkaloids. Developed through systematic structural optimization, toyaburgine is a potent anticancer agent, showing promise for pancreatic cancer treatment. It exhibits strong antiausterity activity with low nanomolar PC50 values, effectively inhibiting pancreatic cancer cell viability under nutrient-deprived conditions. In vitro, 4 causes significant morphological changes and cancer cell death in MIA PaCa-2 cells while also inhibiting cell migration and colony formation, which indicates its antimetastatic potential. Mechanistically, toyaburgine disrupts the PI3K/Akt/mTOR pathway, essential for pancreatic cancer cell survival in a stressful microenvironment, and inhibits MIA PaCa-2 spheroid formation. In vivo, toyaburgine, alone or combined with gemcitabine, shows effective tumor suppression in subcutaneous xenograft and clinically relevant orthotopic models, where it also reduces cachexia. These results highlight the potential of toyaburgine as a new therapeutic drug for pancreatic cancer. Its combination with gemcitabine presents a promising treatment approach by targeting both proliferating and gemcitabine-resistant cancer cells.
Collapse
Affiliation(s)
- Suresh Awale
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Juthamart Maneenet
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Nguyen Duy Phan
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hung Hong Nguyen
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Heiko Ihmels
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
| | - Denisa Soost
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
| | - Nasir Tajuddeen
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
- Department of Chemistry, Ahmadu Bello University, 810107 Zaria, Nigeria
| | - Doris Feineis
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
9
|
Pirota V, Iachettini S, Platella C, Zizza P, Fracchioni G, Di Vito S, Carachino A, Battistini F, Orozco M, Freccero M, Biroccio A, Montesarchio D, Doria F. Naphthalene diimide-naphthalimide dyads promote telomere damage by selectively targeting multimeric G-quadruplexes. Nucleic Acids Res 2025; 53:gkaf301. [PMID: 40239997 PMCID: PMC12000870 DOI: 10.1093/nar/gkaf301] [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: 06/17/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
G-quadruplex (G4) nucleic acid ligands have attracted significant attention as putative anticancer agents for selectively stabilizing telomeric structures. In our pursuit of targeting the most biologically relevant telomeric structures, we have investigated a new class of naphthalene diimide (NDI)-based ligands designed to bind multimeric G4s. The NDI unit covalently linked with one 1,8-naphthalimide (NI) moiety, results in ligands able to fold into a sandwich-like conformation fitting into the binding pockets of telomeric multimeric G4s, thus optimizing binding complementarity. Varying the NDI decorations, we synthesized a small library of NDI-NI dyads and then examined their capability of stabilizing G4s by biophysical assays. Given the relevance of G4 stabilizing agents in fighting cancer, the most promising NDI-NIs were evaluated for their antitumoral activity on a panel of human cell lines originating from different tumor histotypes. Obtained results evidenced that three of the selected ligands promoted an accumulation of telomere-localized damage leading to a robust impairment of cell viability, regardless of homologous recombination status. These data, then confirmed in advanced 3D models, paved the way for the advancement of NDI-NIs as a new class of clinically relevant antitumoral agents. Finally, computational analyses gained deeper insight into their binding modality.
Collapse
Affiliation(s)
- Valentina Pirota
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Sara Iachettini
- Translational Oncology Research Unit, IRCCS - Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Pasquale Zizza
- Translational Oncology Research Unit, IRCCS - Regina Elena National Cancer Institute, 00144 Rome, Italy
| | | | - Serena Di Vito
- Translational Oncology Research Unit, IRCCS - Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Alice Carachino
- Translational Oncology Research Unit, IRCCS - Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Federica Battistini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Department of Biochemistry and Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Department of Biochemistry and Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Mauro Freccero
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Annamaria Biroccio
- Translational Oncology Research Unit, IRCCS - Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Filippo Doria
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
10
|
Lu C, Jin A, Liu H, Gao C, Sun W, Zhang Y, Dai Q, Liu Y. Advancing tissue engineering through vascularized cell spheroids: building blocks of the future. Biomater Sci 2025; 13:1901-1922. [PMID: 40067332 DOI: 10.1039/d4bm01206b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Vascularization is a crucial aspect of biofabrication, as the development of vascular networks is essential for tissue survival and the optimization of cellular functions. Spheroids have emerged as versatile units for vascularization, demonstrating significant potential in angiogenesis and prevascularization for tissue engineering and regenerative medicine. However, a major challenge in creating customized vascularized spheroids is the construction of a biomimetic extracellular matrix (ECM) microenvironment. This process requires careful regulation of environmental factors, including the modulation of growth factors, the selection of culture media, and the co-culture of diverse cell types. Recent advancements in biofabrication have expanded the potential applications of vascularized spheroids. The integration of microfluidic technology with bioprinting offers promising solutions to existing challenges in regenerative medicine. Spheroids have been widely studied for their ability to promote vascularization in in vitro models. This review highlights the latest developments in vascularized biofabrication, and systematically explores strategies for constructing vascularized spheroids. We provide a comprehensive analysis of spheroid applications in specific tissues, including skin, liver, bone, cardiac, and tumor models. Finally, the review addresses the major challenges and future directions in the field.
Collapse
Affiliation(s)
- Chunxiang Lu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Aoxiang Jin
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Huazhen Liu
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Wenbin Sun
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Yi Zhang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Qiqi Dai
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai, 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| |
Collapse
|
11
|
Borrelli MJ, Kolendowski B, DiMattia GE, Shepherd TG. Spatiotemporal analysis of ratiometric biosensors in live multicellular spheroids using SPoRTS. CELL REPORTS METHODS 2025; 5:100987. [PMID: 39965566 PMCID: PMC11955269 DOI: 10.1016/j.crmeth.2025.100987] [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: 09/05/2024] [Revised: 11/06/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Here, we describe SPoRTS, an open-source workflow for high-throughput spatiotemporal image analysis of fluorescence-based ratiometric biosensors in living spheroids. To achieve this, we have implemented a fully automated algorithm for the acquisition of line intensity profile data, ultimately enabling semi-quantitative measurement of biosensor activity as a function of distance from the center of the spheroid. We demonstrate the functionality of SPoRTS via spatial analysis of live spheroids expressing a ratiometric biosensor based on the fluorescent, ubiquitin-based cell-cycle indicator (FUCCI) system, which identifies mitotic cells. We compare this FUCCI-based SPoRTS analysis with spatially quantified immunostaining for proliferation markers, finding that the results are strongly correlated.
Collapse
Affiliation(s)
- Matthew J Borrelli
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada; Department of Anatomy and Cell Biology, Western University, London, ON N6A 5C1, Canada
| | - Bart Kolendowski
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Gabriel E DiMattia
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada; Department of Oncology, Western University, London, ON N6A 5W9, Canada; Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
| | - Trevor G Shepherd
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada; Department of Anatomy and Cell Biology, Western University, London, ON N6A 5C1, Canada; Department of Oncology, Western University, London, ON N6A 5W9, Canada; Department of Obstetrics and Gynecology, Western University, London, ON N6A 5W9, Canada.
| |
Collapse
|
12
|
Pyrczak-Felczykowska A, Herman-Antosiewicz A. Modification in Structures of Active Compounds in Anticancer Mitochondria-Targeted Therapy. Int J Mol Sci 2025; 26:1376. [PMID: 39941144 PMCID: PMC11818413 DOI: 10.3390/ijms26031376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer is a multifaceted disease characterised by uncontrolled cellular proliferation and metastasis, resulting in significant global mortality. Current therapeutic strategies, including surgery, chemotherapy, and radiation therapy, face challenges such as systemic toxicity and tumour resistance. Recent advancements have shifted towards targeted therapies that act selectively on molecular structures within cancer cells, reducing off-target effects. Mitochondria have emerged as pivotal targets in this approach, given their roles in metabolic reprogramming, retrograde signalling, and oxidative stress, all of which drive the malignant phenotype. Targeting mitochondria offers a promising strategy to address these mechanisms at their origin. Synthetic derivatives of natural compounds hold particular promise in mitochondrial-targeted therapies. Innovations in drug design, including the use of conjugates and nanotechnology, focus on optimizing these compounds for mitochondrial specificity. Such advancements enhance therapeutic efficacy while minimizing systemic toxicity, presenting a significant step forward in modern anticancer strategies.
Collapse
Affiliation(s)
| | - Anna Herman-Antosiewicz
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, 80-308 Gdańsk, Poland;
| |
Collapse
|
13
|
Świerczewska M, Nowacka M, Stasiak P, Iżycki D, Sterzyńska K, Płóciennik A, Nowicki M, Januchowski R. Doxorubicin and topotecan resistance in ovarian cancer: Gene expression and microenvironment analysis in 2D and 3D models. Biomed Pharmacother 2025; 183:117804. [PMID: 39787968 DOI: 10.1016/j.biopha.2024.117804] [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: 09/10/2024] [Revised: 12/19/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
This study explores the mechanisms underlying chemotherapy resistance in ovarian cancer (OC) using doxorubicin (DOX) and topotecan (TOP)-resistant cell lines derived from the drug-sensitive A2780 ovarian cancer cell line. Both two-dimensional (2D) monolayer cell cultures and three-dimensional (3D) spheroid models were employed to examine the differential drug responses in these environments. The results revealed that 3D spheroids demonstrated significantly higher resistance to DOX and TOP than 2D cultures, suggesting a closer mimicry of in vivo tumour conditions. Molecular analyses identified overexpression of essential drug resistance-related genes, including MDR1 and BCRP, and extracellular matrix (ECM) components, such as MYOT and SPP1, which were more pronounced in resistant cell lines. MDR1 and BCRP overexpression contribute to chemotherapy resistance in OC by expelling drugs like DOX and TOP. Targeting these transporters with inhibitors or gene silencing could improve drug efficacy, making them key therapeutic targets to enhance treatment outcomes for drug-resistant OC. The study further showed that EMT-associated markers, including VIM, SNAIL1, and SNAIL2, were upregulated in the 3D spheroids, reflecting a more mesenchymal phenotype. These findings suggest that factors beyond gene expression, such as spheroid architecture, cell-cell interactions, and drug penetration, contribute to the enhanced resistance observed in 3D cultures. These results highlight the importance of 3D cell culture models for a more accurate representation of tumour drug resistance mechanisms in ovarian cancer, providing valuable insights for therapeutic development.
Collapse
Affiliation(s)
- Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., Poznan 61-781, Poland; Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, Zyty 28 St., Zielona Góra 65-046, Poland.
| | - Marta Nowacka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., Poznan 61-781, Poland.
| | - Piotr Stasiak
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, Zyty 28 St., Zielona Góra 65-046, Poland.
| | - Dariusz Iżycki
- Department of Cancer Immunology, Poznan University of Medical Sciences, Garbary 15 St., Poznan 61-866, Poland.
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., Poznan 61-781, Poland.
| | - Artur Płóciennik
- Department of Plant Ecophysiology, Adam Mickiewicz University, Wieniawskiego 1 St., Poznan 61-712, Poland.
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., Poznan 61-781, Poland.
| | - Radosław Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, Zyty 28 St., Zielona Góra 65-046, Poland.
| |
Collapse
|
14
|
Song B, Wu M, Qin L, Liang W, Wang X. Smart Design of Targeted Drug Delivery System for Precise Drug Delivery and Visual Treatment of Brain Gliomas. Adv Healthc Mater 2025; 14:e2402967. [PMID: 39707642 DOI: 10.1002/adhm.202402967] [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: 08/09/2024] [Revised: 12/04/2024] [Indexed: 12/23/2024]
Abstract
In the treatment of glioma, which is one of the malignant tumors, although chemotherapy is used as the most common treatment method, it often suffers from low bioavailability. Therefore, improving the precision and efficiency of drugs is crucial in treating gliomas and a great challenge. Here, an advanced drug delivery system is reported for gliomas (CZQD@HA@DOX), which aggregates multiple features such as the susceptible imaging tracer property due to the use of CZQD and the targeting of HA to the receptor cluster 44 (CD44) of glioma cells, which provides the system with the functions of targeted enrichment and precise drug delivery at the tumor site. The pH-responsive drug delivery system has not only an excellent encapsulation rate but also a high drug loading capacity, and the doxorubicin loaded on it can be released centrally at the tumor microenvironment site and causes an increase of reactive oxygen species in the mitochondria and trigger oxidative stress, which leads to high expression of Bax apoptotic proteins, ultimately activating the mitochondrial pathway-mediated apoptotic process in glioma cells. Overall, this drug delivery system has great potential for application in precision targeted therapy and visual tracer imaging of gliomas.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Mengru Wu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Lijing Qin
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| |
Collapse
|
15
|
Joseph S, Zhang X, Droby GN, Wu D, Bae-Jump V, Lyons S, Mordant A, Mills A, Herring L, Rushing B, Bowser JL, Vaziri C. MAPK14/p38α shapes the molecular landscape of endometrial cancer and promotes tumorigenic characteristics. Cell Rep 2025; 44:115104. [PMID: 39708320 DOI: 10.1016/j.celrep.2024.115104] [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: 06/18/2024] [Revised: 10/25/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The molecular underpinnings of high-grade endometrial carcinoma (HGEC) metastatic growth and survival are poorly understood. Here, we show that ascites-derived and primary tumor HGEC cell lines in 3D spheroid culture faithfully recapitulate key features of malignant peritoneal effusion and exhibit fundamentally distinct transcriptomic, proteomic, and metabolomic landscapes compared with conventional 2D monolayers. Using a genetic screening platform, we identify MAPK14 (which encodes the protein kinase p38α) as a specific requirement for HGEC in spheroid culture. MAPK14/p38α has broad roles in programming the phosphoproteome, transcriptome, and metabolome of HGEC spheroids, yet has negligible impact on monolayer cultures. MAPK14 promotes tumorigenicity in vivo and is specifically required to sustain a sub-population of spheroid cells that is enriched in cancer stemness markers. Therefore, spheroid growth of HGEC activates unique biological programs, including p38α signaling, that cannot be captured using 2D culture models and are highly relevant to malignant disease pathology.
Collapse
Affiliation(s)
- Sayali Joseph
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gaith N Droby
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Di Wu
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victoria Bae-Jump
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Scott Lyons
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Angie Mordant
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Allie Mills
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Laura Herring
- Department of Pharmacology, UNC Proteomics Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Blake Rushing
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
16
|
Jin X, Dong H, Wang J, Ou G, Lai X, Tian X, Wang L, Zhuang H, Li T, Xiang K. HBx Facilitates Drug Resistance in Hepatocellular Carcinoma via CD133-regulated Self-renewal of Liver Cancer Stem Cells. J Clin Transl Hepatol 2025; 13:15-24. [PMID: 39801781 PMCID: PMC11712087 DOI: 10.14218/jcth.2024.00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 01/16/2025] Open
Abstract
Background and Aims Hepatitis B virus (HBV) infection contributes to hepatocellular carcinoma (HCC) tumorigenesis, drug resistance, and recurrence, although the underlying molecular mechanisms remain unclear. Recent studies suggest that HBV infection may be associated with liver cancer stem cells (LCSCs), but the exact mechanisms are yet to be resolved. In this study, we aimed to analyze the role of HBV infection in regulating the stemness of HCCs, which is closely linked to drug resistance. Methods Sphere formation assay and real-time Polymerase Chain Reaction quantification were used to isolate and confirm liver cancer stem cells. The inhibitory concentration values of sorafenib and regorafenib were calculated and compared using the Cell Counting Kit-8 assay. HBV infection was used to assess the effect of HBV replication on LCSC markers. Co-immunoprecipitation assay was performed to detect the interaction between CD133 and SRC. Furthermore, we utilized the CRISPR-Cas9 system to knockout CD133 expression in HepG2.2.15 cells. Results LCSCs derived from HCCs exhibited high expression of stem cell markers and demonstrated reduced sensitivity to sorafenib and regorafenib. HBV replication promoted both drug resistance and stemness in hepatoma cells and clinical samples. Overexpression of HBx protein in HepG2 cells upregulated the expression of CD133, EpCAM, and CD24, enhancing resistance to sorafenib and regorafenib. Knockout of CD133 expression using the CRISPR-Cas9 system significantly inhibited drug resistance to both sorafenib and regorafenib in HepG2.2.15 cells. Mechanistically, HBV replication promoted CD133 expression, which in turn interacted with the SRC/STAT3 signaling pathway. Conclusions Our data suggest that HBV replication enhances the stemness and drug resistance of HCC, providing a strong theoretical foundation for the development of targeted and efficient treatments for HBV-infected HCCs.
Collapse
Affiliation(s)
- Xiangshu Jin
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Department of Obstetrics and Gynecology, the Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Huijun Dong
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University–YHLO Joint Laboratory for Molecular Diagnostics of Infectious Diseases, Peking University, Beijing, China
| | - Juan Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guomin Ou
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University–YHLO Joint Laboratory for Molecular Diagnostics of Infectious Diseases, Peking University, Beijing, China
| | - Xinyuan Lai
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University–YHLO Joint Laboratory for Molecular Diagnostics of Infectious Diseases, Peking University, Beijing, China
| | - Xing Tian
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Department of Physiology, Shenyang Medical College, Shenyang, Liaoning, China
| | - Lei Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hui Zhuang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University–YHLO Joint Laboratory for Molecular Diagnostics of Infectious Diseases, Peking University, Beijing, China
| | - Tong Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University–YHLO Joint Laboratory for Molecular Diagnostics of Infectious Diseases, Peking University, Beijing, China
| | - Kuanhui Xiang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Peking University–YHLO Joint Laboratory for Molecular Diagnostics of Infectious Diseases, Peking University, Beijing, China
| |
Collapse
|
17
|
Xu J, Fang W, Zhou H, Jiang R, Chen Z, Wang X. Application and progress of 3D tumor models in breast cancer. Biotechnol Bioeng 2025; 122:30-43. [PMID: 39402769 DOI: 10.1002/bit.28860] [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: 08/13/2023] [Revised: 07/09/2024] [Accepted: 10/01/2024] [Indexed: 12/12/2024]
Abstract
Due to its high heterogeneity and significant impact on women's health globally, breast cancer necessitates robust preclinical models to understand tumor biology and guide personalized treatment strategies. Three-dimensional (3D) in vitro tumor models hold immense promise in this regard. These tumor models not only mimic the spatial structure and growth environment of tumors in vivo, but also retain the pathological and genetic characteristics of solid tumors. This fidelity makes them powerful tools for accelerating advancements in fundamental research and translational medicine. The diversity, modularity, and efficacy of 3D tumor models are driving a biotechnological revolution. As these technologies become increasingly sophisticated, 3D tumor models are poised to become powerful weapons in the fight against breast cancer. This article expounds on the progress made in utilizing 3D tumor models for breast cancer research.
Collapse
Affiliation(s)
- Jiaojiao Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wanxia Fang
- The Department of Colorectal Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Huanhuan Zhou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruiyuan Jiang
- The Department of Breast Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhanhong Chen
- The Department of Breast Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiaojia Wang
- The Department of Breast Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
18
|
Sekeroglu ZA, Sekeroglu V. A Review on Patient-derived 3D Micro Cancer Approach for Drug Screen in Personalized Cancer Medicine. Curr Cancer Drug Targets 2025; 25:118-130. [PMID: 38445692 DOI: 10.2174/0115680096285910240206044830] [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: 11/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 03/07/2024]
Abstract
Precision medicine in oncology aims to identify an individualized treatment plan based on genomic alterations in a patient's tumor. It helps to select the most beneficial therapy for an individual patient. As it is now known that no patient's cancer is the same, and therefore, different patients may respond differently to conventional treatments, precision medicine, which replaces the one-size-fits-all approach, supports the development of tailored treatments for specific cancers of different patients. Patient-specific organoid or spheroid models as 3D cell culture models are very promising for predicting resistance to anti-cancer drugs and for identifying the most effective cancer therapy for high-throughput drug screening combined with genomic analysis in personalized medicine. Because tumor spheroids incorporate many features of solid tumors and reflect resistance to drugs and radiation, as in human cancers, they are widely used in drug screening studies. Testing patient-derived 3D cancer spheroids with some anticancer drugs based on information from molecular profiling can reveal the sensitivity of tumor cells to drugs and provide the right compounds to be effective against resistant cells. Given that many patients do not respond to standard treatments, patient-specific treatments will be more effective, less toxic. They will affect survival better compared to the standard approach used for all patients.
Collapse
Affiliation(s)
- Zulal Atlı Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Vedat Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| |
Collapse
|
19
|
Du Y, Ouyang B, Liu Y, Yin Y, Wu Y, Guo H. A Hydrogel for Nitric Oxide Sensitization Chemotherapy Mediated by Tumor Microenvironment Changes in 3D Spheroids and Breast Tumor Models. Curr Pharm Des 2025; 31:1227-1238. [PMID: 39819416 DOI: 10.2174/0113816128348357241209050425] [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: 08/13/2024] [Revised: 10/20/2024] [Accepted: 11/15/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Nitric oxide (NO) is a low-toxicity and high-efficiency anticancer treatment that can augment the cytotoxicity of doxorubicin (DOX) towards breast cancer cells, thereby exhibiting a favorable effect on chemotherapy sensitization. OBJECTIVE The study aimed to establish a hydrogel that sensitizes chemotherapy by inducing local inflammatory stimulation to change the tumor microenvironment and promote NO production. The purpose of the study was to examine the anti-tumor effect in vivo and in vitro. METHODS The functional properties of the composite hydrogels were tested by UV spectrophotometry and NO detection kit. CCK8, DCFH-DA fluorescent probe, Calcein-AM/PI detection kit, and confocal detection methods were used for the cytocompatibility and cytotoxicity of the composite hydrogels. The subcutaneous tumor volume, weight, and tumor inhibition rate of 4T1 breast cancer cells were evaluated for pharmacodynamic study in vivo. RESULTS Each component of hydrogel has good biocompatibility. The combination of gas therapy and chemotherapy can significantly enhance the effect of inhibiting tumor cell growth. The tumor growth of tumor- bearing mice in the hydrogel administration group was slow, and the tumor inhibition rate was 85.10%. The body weight grew steadily, and no significant pathological changes were observed in the H&E staining of major organs. CONCLUSION A composite hydrogel with alginate as the carrier was successfully established, which was based on improving the tumor microenvironment to trigger gas therapy combined with chemotherapy for tumor treatment.
Collapse
Affiliation(s)
- Yang Du
- Central Laboratory, First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- The Institute of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
| | - Boshu Ouyang
- Department of Integrative Medicine, Institute of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yao Liu
- The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200092, China
- Center for Medical Research and Innovation, Pudong Medical Center, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Yuzhen Yin
- Central Laboratory, First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- The Institute of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
| | - Yining Wu
- Central Laboratory, First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- The Institute of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- The Institute of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
| |
Collapse
|
20
|
Li A, Pan W, Zhang Z, Yang F, Gou Y, Zhang Y, Ma L. Hydrazone copper(II) complexes suppressed lung adenocarcinoma by activating multiple anticancer pathway. Bioorg Chem 2025; 154:107994. [PMID: 39603071 DOI: 10.1016/j.bioorg.2024.107994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Activating multiple anti-cancer pathways has great potential for tumor treatment. Herein, we synthesized two binuclear Cu(II) hydrazone complexes ([Cu2(HL1)2Cl2] 1 and [Cu2(HL1)2Br2] 2) and two mononuclear hydrazone-Cu(II) complexes ([Cu(HL2)Cl]·CH3OH 3 and [Cu(HL2)(H2O)Br]·2H2O 4), to evaluate their anti-lung cancer activities. MTT assays revealed that the Cu(II) complexes demonstrate superior anticancer activity compared to cisplatin. Among them, complex 3 exhibited selective toxicity towards A549 cancer cells in comparison to normal cells and demonstrated hemolytic activity comparable to cisplatin. The low toxicity and effective antitumor capabilities of complex 3 have been confirmed in xenograft experiments using A549 tumor-bearing mice. Interestingly, complex 3 eradicates lung tumor cells both in vivo and in vitro by initiating multiple anticancer pathways, including cuproptosis. Our research extends the study of hydrazone copper complexes and provides strategies for the treatment of lung cancer.
Collapse
Affiliation(s)
- Aili Li
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Weiping Pan
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - ZhenLei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, China
| | - Yi Gou
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China.
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, China.
| | - Libing Ma
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China.
| |
Collapse
|
21
|
Moreno-Sanchez R, Vargas-Navarro JL, Padilla-Flores JA, Robledo-Cadena DX, Granados-Rivas JC, Taba R, Terasmaa A, Auditano GL, Kaambre T, Rodriguez-Enriquez S. Energy Metabolism Behavior and Response to Microenvironmental Factors of the Experimental Cancer Cell Models Differ from that of Actual Human Tumors. Mini Rev Med Chem 2025; 25:319-339. [PMID: 39411957 DOI: 10.2174/0113895575322436240924101642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 04/09/2025]
Abstract
Analysis of the biochemical differences in the energy metabolism among bi-dimensional (2D) and tri-dimensional (3D) cultured cancer cell models and actual human tumors was undertaken. In 2D cancer cells, the oxidative phosphorylation (OxPhos) fluxes range is 2.5-19 nmol O2/min/mg cellular protein. Hypoxia drastically decreased OxPhos flux by 2-3 times in 2D models, similar to what occurs in mature multicellular tumor spheroids (MCTS), a representative 3D cancer cell model. However, mitochondrial protein contents and enzyme activities were significantly different between both models. Moreover, glycolytic fluxes were also significantly different between 2D and MCTS. The glycolytic flux range in 2D models is 1-34 nmol lactate/min/mg cellular protein, whereas in MCTS the range of glycolysis fluxes is 60-80 nmol lactate/min/mg cellular. In addition, sensitivity to anticancer canonical and metabolic drugs was greater in MCTS than in 2D. Actual solid human tumor samples show lower (1.6-4.5 times) OxPhos fluxes compared to normoxic 2D cancer cell cultures. These observations indicate that tridimensional organization provides a unique microenvironment affecting tumor physiology, which has not been so far faithfully reproduced by the 2D environment. Thus, the analysis of the resemblances and differences among cancer cell models undertaken in the present study raises caution on the interpretation of results derived from 2D cultured cancer cells when they are extended to clinical settings. It also raises awareness about detecting which biological and environmental factors are missing in 2D and 3D cancer cell models to be able to reproduce the actual human tumor behavior.
Collapse
Affiliation(s)
- Rafael Moreno-Sanchez
- Laboratorio de Control Metabólico, Carrera de Biología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Estado de México, México
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Jorge Luis Vargas-Navarro
- Laboratorio de Control Metabólico, Carrera de Biología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Estado de México, México
| | - Joaquin Alberto Padilla-Flores
- Laboratorio de Control Metabólico, Carrera de Biología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Estado de México, México
| | - Diana Xochiquetzal Robledo-Cadena
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Colonia Sección XVI, Tlalpan, México
| | - Juan Carlos Granados-Rivas
- Laboratorio de Control Metabólico, Carrera de Biología, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Estado de México, México
| | - Rutt Taba
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Anton Terasmaa
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | | | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Sara Rodriguez-Enriquez
- Laboratorio de Control Metabólico, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Estado de México, México
| |
Collapse
|
22
|
Lu C, Jin A, Gao C, Qiao H, Liu H, Zhang Y, Sun W, Yang SM, Liu Y. Synergistic Approach of High-Precision 3D Printing and Low Cell Adhesion for Enhanced Self-Assembled Spheroid Formation. BIOSENSORS 2024; 15:7. [PMID: 39852058 PMCID: PMC11764235 DOI: 10.3390/bios15010007] [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: 11/07/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025]
Abstract
Spheroids, as three-dimensional (3D) cell aggregates, can be prepared using various methods, including hanging drops, microwells, microfluidics, magnetic manipulation, and bioreactors. However, current spheroid manufacturing techniques face challenges such as complex workflows, the need for specialized personnel, and poor batch reproducibility. In this study, we designed a support-free, 3D-printed microwell chip and developed a compatible low-cell-adhesion process. Through simulation and experimental validation, we rapidly optimized microwell size and the coating process. We successfully formed three types of spheroids-human immortalized epidermal cells (HaCaTs), umbilical cord mesenchymal stem cells (UC-MSCs), and human osteosarcoma cells (MG63s)-on the chip. Fluorescent viability staining confirmed the biocompatibility and reliability of the chip. Finally, drug response experiments were conducted using the chip. Compared to traditional methods, our proposed strategy enables high-throughput production of size-controlled spheroids with excellent shape retention, while enhanced gas exchange during culture improves differentiation marker expression. This platform provides an efficient and cost-effective solution for biosensing applications, such as drug screening, disease modeling, and personalized therapy monitoring. Furthermore, the chip shows significant potential for real-time in vitro monitoring of cellular viability, reaction kinetics, and drug sensitivity, offering valuable advancements in biosensor technology for life sciences and medical applications.
Collapse
Affiliation(s)
- Chunxiang Lu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Aoxiang Jin
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Hao Qiao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Huazhen Liu
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Yi Zhang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Wenbin Sun
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Shih-Mo Yang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China; (C.L.); (A.J.); (C.G.); (H.Q.); (Y.Z.); (W.S.); (S.-M.Y.)
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, China
| |
Collapse
|
23
|
Patel T, Jain N. Multicellular tumor spheroids: A convenient in vitro model for translational cancer research. Life Sci 2024; 358:123184. [PMID: 39490437 DOI: 10.1016/j.lfs.2024.123184] [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: 08/23/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
In the attempts to mitigate uncertainties in the results of monolayer culture for the identification of cancer therapeutic targets and compounds, there has been a growing interest in using 3D cancer spheroid models, which include tumorospheres (TSs), tissue-derived tumor spheres (TDTSs), organotypic multicellular tumor spheroids (OMSs), and multicellular tumor spheroids (MCTSs). The MCTSs, either Mono-MCTSs or Hetero-MCTSs, with or without scaffold, in particular, offer numerous advantages over other spheroid models, including easy cultivation, high reproducibility, accessibility, high throughput, controllable size, well-rounded shape, simplicity of genetic manipulation, economical and availability of various biological methods for their development. In this review, we have attempted to discuss the role of MCTSs concerning various aspects of translational cancer research, such as drug response and penetration, cell-cell interaction, and invasion and metastasis. However, the Mono-MCTSs, either scaffold-free or scaffold-based, may not adequately represent the cellular heterogeneity and complexity of clinical tumors, limiting their utility in translational cancer research. Conversely, Hetero-MCTS models, both scaffold-free and scaffold-based, show better suitability due to the presence of a similar in vivo type tumor microenvironment. Nonetheless, scaffold-based Hetero-MCTS models show batch variability and challenges in performing quantitative assays due to difficulties extracting spheroids and cells from scaffolds. Further, incorporating stromal cells with cancer cells in a more precise ratio to develop Hetero-MCTSs can enhance the model's relevance, yielding more clinically reliable outcomes for drug candidates and improving insights into tumor biology.
Collapse
Affiliation(s)
- Tushar Patel
- P D Patel Institute of Applied Sciences, Charotar University of Science and Technology (CHARUSAT), Changa 388 421, India
| | - Neeraj Jain
- Dr. K C Patel Research and Development Centre, University Research Centre(s), Charotar University of Science and Technology (CHARUSAT), Changa 388 421, India.
| |
Collapse
|
24
|
El Mokbel N, Goyeneche AA, Prakash R, Forgie BN, Abdalbari FH, Zeng X, Tessier-Cloutier B, Annie Leung SO, Telleria CM. Comparison of two-dimensional and three-dimensional culture systems and their responses to chemotherapy in cells representing disease progression of high-grade serous ovarian cancer. Biochem Biophys Rep 2024; 40:101838. [PMID: 39469046 PMCID: PMC11513490 DOI: 10.1016/j.bbrep.2024.101838] [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: 07/12/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024] Open
Abstract
High-grade serous cancer is the most common type of ovarian cancer and is usually diagnosed at advanced stages with high mortality due to recurrence and eventual resistance to standard platinum therapy. The aim of this study was to compare two-dimensional (2D) versus tridimensional (3D) cell culture as a preclinical model of response to carboplatin, paclitaxel and niraparib using PEO1, PEO4 and PEO6 cell lines, which were generated from the same patient along disease progression. Morphologically, cells formed flat adherent layers versus spheroidal structures with different compaction patterns in 2D and 3D respectively. In 2D, apoptosis was rare whereas in 3D cells formed a multilayered structure with an outer layer of live proliferating cells and an inner core of apoptotic cells. Furthermore, a differential capacity to produce ATP was observed among the cell lines in 3D but not in 2D. While response to carboplatin, paclitaxel and niraparib in both settings followed a similar trend, a lower sensitivity was observed in 3D with respect to 2D. Overall, 3D cell culture is likely more reflective of the in vivo cellular tumor behavior and more suitable of therapeutic evaluation given its added complexity absent in 2D.
Collapse
Affiliation(s)
- Naya El Mokbel
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
| | - Alicia A. Goyeneche
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
| | - Rewati Prakash
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
| | - Benjamin N. Forgie
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
| | - Farah H. Abdalbari
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
| | - Xing Zeng
- McGill University Health Centre, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Montreal, QC, Canada
| | - Basile Tessier-Cloutier
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
| | - Shuk On Annie Leung
- McGill University Health Centre, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Cancer Research Program, Montreal, QC, Canada
| | - Carlos M. Telleria
- McGill University, Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, Cancer Research Program, Montreal, QC, Canada
| |
Collapse
|
25
|
Son KH, Kim DH, Park S, Kim HJ, Park M, Kim SJ, Lee SJ, Ahn K, Lee JW. Spherical Shell Bioprinting to Produce Uniform Spheroids with Controlled Sizes. J Funct Biomater 2024; 15:350. [PMID: 39590553 PMCID: PMC11595458 DOI: 10.3390/jfb15110350] [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: 11/01/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Conventional cell spheroid production methods are largely manual, leading to variations in size and shape that compromise consistency and reliability for use in cell-based therapeutic applications. To enhance spheroid production, a spherical shell bioprinting system was implemented, enabling the high-throughput generation of uniform cell spheroids with precisely controlled sizes. The system encapsulates cells within thin alginate hydrogel shells formed through bioprinting and ion crosslinking reactions. Alginate-calcium ion crosslinking created alginate shells that contained gelatin-based bioinks with embedded cells, facilitating spontaneous cell aggregation within the shells and eliminating the need for plastic wells. By adjusting cell concentrations in the alginate-gelatin bioink, we achieved precise control over spheroid size, maintaining a sphericity above 0.94 and size deviations within ±10 µm. This method has been successfully applied to various cell types including cancer cells, fibroblasts, chondrocytes, and epithelial cells, demonstrating its versatility. This scalable approach enhances the reliability of cell therapy and drug screening, offering a robust platform for future biomedical applications.
Collapse
Affiliation(s)
- Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, College of Medicine, Gachon University, 21, Namdong-daero 774 Beon-gil, Namdong-gu, Incheon 21565, Republic of Korea;
| | - Dong-Ha Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seunghye Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
| | - Hyun Jae Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Mira Park
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seung-Jin Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Keunsun Ahn
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| |
Collapse
|
26
|
de Andrade-da-Costa J, de-Souza-Ferreira M, Dos Santos Touça NC, Sousa-Squiavinato ACM, Soares-Lima SC, Morgado-Díaz JA, de-Freitas-Junior JCM. Enrichment of cancer stem cell subpopulation alters the glycogene expression profile of colorectal cancer cells. Discov Oncol 2024; 15:647. [PMID: 39532788 PMCID: PMC11557779 DOI: 10.1007/s12672-024-01536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Colorectal cancer (CRC) has a high mortality rate, resulting from the processes of metastasis and disease recurrence. Cancer stem cells (CSCs) are believed to be crucial for both processes, as they ensure the maintenance of the tumor bulk, in addition to being intrinsically resistant to conventional therapies. Thus, the present study aimed to investigate glycobiomarkers in colorectal cancer stem cell subpopulations. For this purpose, a sphere formation assay was standardized for CACO-2 and HT-29 cell lines, which were monitored through gene expression analysis of five known CSC markers (CD24, CD44, ALDH1, LGR5, and PROM1). Compared to the parental condition (2D), a reduction in CD24 expression was seen in CACO-2, while in HT-29 an increase in the expression levels of ALDH1, LGR5, and PROM1 was observed. Regarding glycogenes, eight of them (ST3GAL1, OGT, OGA, MGAT5, GFAT1, GFAT2, B4GALT1 e B3GNT2) have had their expression monitored. An increase in B3GNT2, OGT, and OGA was observed in the HT-29 sphere condition. On the other hand, no change in the glycogenes expression was observed in CACO-2. In silico correlation analyses (CSCs markers versus glycogenes) using TCGA data from colon and rectum carcinoma samples showed a weak positive correlation between LGR5 vs OGA expression regardless of the sample location. In addition, an increase in the expression of LGR5, OGA, and OGT as well as a decrease in the expression of ALDH1 were observed in colon carcinoma samples when compared to the adjacent normal tissue. Interestingly, greater OGA expression resulted in both lower overall survival of colon carcinoma patients and lower disease-free survival of rectum carcinoma patients. Therefore, our data indicates that OGA expression correlates with CSC markers and directly impacts the survival of colorectal carcinoma patients.
Collapse
Affiliation(s)
- Jéssica de Andrade-da-Costa
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Michelle de-Souza-Ferreira
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
- Cellular and Molecular Pharmacology Laboratory, Rio de Janeiro State University (UERJ), Rio de Janeiro, RJ, Brazil
| | - Nathália Campos Dos Santos Touça
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
- Cellular and Molecular Pharmacology Laboratory, Rio de Janeiro State University (UERJ), Rio de Janeiro, RJ, Brazil
| | | | - Sheila Coelho Soares-Lima
- Molecular Carcinogenesis Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - José Andrés Morgado-Díaz
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Julio Cesar Madureira de-Freitas-Junior
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil.
- Cellular and Molecular Pharmacology Laboratory, Rio de Janeiro State University (UERJ), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
27
|
Bhat N, Al-Mathkour M, Maacha S, Lu H, El-Rifai W, Ballout F. Esophageal adenocarcinoma models: a closer look. Front Mol Biosci 2024; 11:1440670. [PMID: 39600303 PMCID: PMC11589788 DOI: 10.3389/fmolb.2024.1440670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Esophageal adenocarcinoma (EAC) is a subtype of esophageal cancer with significant morbidity and mortality rates worldwide. Despite advancements in tumor models, the underlying cellular and molecular mechanisms driving EAC pathogenesis are still poorly understood. Therefore, gaining insights into these mechanisms is crucial for improving patient outcomes. Researchers have developed various models to better understand EAC and evaluate clinical management strategies. However, no single model fully recapitulates the complexity of EAC. Emerging technologies, such as patient-derived organoids and immune-competent mouse models, hold promise for personalized EAC research and drug development. In this review, we shed light on the various models for studying EAC and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Nadeem Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Marwah Al-Mathkour
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Selma Maacha
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Farah Ballout
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
28
|
Ebrahimi A, Ak G, Özel C, İzgördü H, Ghorbanpoor H, Hassan S, Avci H, Metintaş M. Clinical Perspectives and Novel Preclinical Models of Malignant Pleural Mesothelioma: A Critical Review. ACS Pharmacol Transl Sci 2024; 7:3299-3333. [PMID: 39539262 PMCID: PMC11555512 DOI: 10.1021/acsptsci.4c00324] [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: 05/31/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Pleural mesothelioma (PM), a rare malignant tumor explicitly associated with asbestos and erionite exposures, has become a global health problem due to limited treatment options and a poor prognosis, in which the median life expectancy varies depending on the method of treatment. However, the importance of early diagnosis is emphasized, and the practical methods have not matured yet. This study provides a critical overview of PM, addressing various aspects like epidemiology, etiology, diagnosis, treatment options, and the potential use of advanced technologies like microfluidic chip-based models for research and diagnosis. It initially begins with fundamentals of clinical aspects and then discusses the identification of disease-specific biomarkers in patients' serum or plasma samples, which could potentially be used for early diagnosis. A detailed investigation of the sophisticated preclinical models is highlighted. Recent three-dimensional (3D) model accomplishments, including microarchitecture modeling by transwell coculture, spheroids, organoids, 3D bioprinting constructs, and ex vivo tumor slices, are discussed comprehensively. On-chip models that imitate physiological processes, such as detection chips and therapeutic screening chips, are assessed as potential techniques. The review concludes with a critical and constructive discussion of the growing interest in the topic and its limitations and suggestions.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Güntülü Ak
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Ceren Özel
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Hüseyin İzgördü
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
| | - Hamed Ghorbanpoor
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Biomedical Engineering, Eskişehir
Osmangazi University, Eskişehir 26040, Turkey
| | - Shabir Hassan
- Department
of Biological Sciences, Khalifa University
of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Huseyin Avci
- Cellular
Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Stem Cell, Institute of Health Sciences, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Department
of Metallurgical and Materials Engineering, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| | - Muzaffer Metintaş
- Eskisehir
Osmangazi University, Faculty of Medicine, Department of Pulmonary
Diseases, Lung and Pleural Cancers Research
and Clinical Center, Eskisehir 26040, Turkey
- Translational
Medicine Research and Clinical Center (TATUM), Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| |
Collapse
|
29
|
Gómez-Mercader A, Monzón-Atienza L, Montero D, Bravo J, Acosta F. Fish Cell Spheroids, a Promising In Vitro Model to Mimic In Vivo Research: A Review. Cells 2024; 13:1818. [PMID: 39513924 PMCID: PMC11544930 DOI: 10.3390/cells13211818] [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: 09/30/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
In vitro cell culture systems serve as instrumental platforms for probing biological phenomena and elucidating intricate cellular mechanisms. These systems afford researchers the opportunity to scrutinize cellular responses within a regulated environment, thereby circumventing the ethical and logistical challenges associated with in vivo experimentation. Three-dimensional (3D) cell cultures have emerged as a viable alternative to mimic in vivo environments. Within this context, spheroids are recognized as one of the most straightforward and efficacious models, presenting a promising substitute for conventional monolayer cultures. The application of 3D cultures of fish cells remains limited, focusing mainly on physiological and morphological characterization studies. However, given the capacity of spheroids to emulate in vivo conditions, researchers are exploring diverse applications of these 3D cultures. These include eco-toxicology, immunology, drug screening, endocrinology, and metabolism studies, employing a variety of cell types such as fibroblasts, hepatocytes, embryonic cells, gonadal cells, gastrointestinal cells, and pituitary cells. This review provides a succinct overview, concentrating on the most frequently employed methods for generating fish cell spheroids and their applications to date. The aim is to compile and highlight the significant contributions of these methods to the field and their potential for future research.
Collapse
|
30
|
Jiang Y, Hao M, Chen S, Xie Y, Liu K. Exploring the impact of microfluidic chip structure on the efficacy of three-dimensional tumor microspheres cultivation. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:7005-7014. [PMID: 39282915 DOI: 10.1039/d4ay01343c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Three-dimensional (3D) tumor microspheres can simulate the interaction and growth dynamics of tumor cells, and have been used as a new in vitro model for drug screening and tumor biology related research. The scaffold-free culture of 3D tumor microspheres on microfluidic chips has many advantages, including low cost, high throughput, convenience and flexibility. However, it is still unclear how various factors, such as chip structure, influence the culture effect of tumor microspheres. The lack of standardized evaluation and characterization of the culture effect hinders the further optimization and development of chip function. This study presents numerical simulations of multiple parts or processes of the proposed 3D culture chips with two different structural parameters based on computational fluid dynamics (CFD) methods. An evaluation system for tumor microspheres was established. The prediction of the CFD simulation was consistent with the culture results of the chips, reflecting the important role of the structural parameters of the microtrap in the formation of uniform tumor microspheres. Furthermore, the velocity of cell suspension also had a significant impact on the retention of tumor cells. Additionally, the drug screening results of tumor microspheres indicated that tumor microspheres exhibit greater drug resistance, which may be attributed to their size. These results offer valuable insights into the factors that influence the characteristics of tumor microspheres. This research provides a reference and direction for the optimal design and functional evaluation of scaffold-free 3D culture chips, and holds promise for promoting the development of novel drug research platforms.
Collapse
Affiliation(s)
- Yue Jiang
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Ming Hao
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Shulei Chen
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Yuanhua Xie
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
| | - Kun Liu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China.
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, China
| |
Collapse
|
31
|
Anjum MR, Subramaniam V, Higgins BR, Abrahan C, Chisolm SJ, Krishnaprasad KA, Azie O, Palmer GD, Angelini TE, Sarntinoranont M. Determining Rates of Molecular Secretion from Supernatant Concentration Measurements in a 3D-Bioprinted Human Liver Tissue Model. ACS Biomater Sci Eng 2024; 10:6711-6720. [PMID: 39259932 DOI: 10.1021/acsbiomaterials.4c01086] [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: 09/13/2024]
Abstract
The secretion rate of albumin is a key indicator of function in liver tissue models used for hepatotoxicity and pharmacokinetic testing. However, it is not generally clear how to determine molecular secretion rates from measurements of the molecular concentration in supernatant media. Here, we develop computational and analytical models of molecular transport in an experimental system that enable determination of albumin secretion rates based on measurements of albumin concentration in supernatant media. The experimental system is a 3D-bioprinted human liver tissue construct embedded in a 3D culture environment made from packed microgel particles swollen in liquid growth media. The mathematical models reveal that the range of albumin synthesis rates necessary to match experimentally measured albumin concentrations corresponds to reaction-limited conditions, where a steady state of albumin spatial distribution is rapidly reached between media exchanges. Our results show that temporally resolved synthesis rates can be inferred from serial concentration measurements of collected supernatant media. This link is critical to confidently assessing in vitro tissue performance in applications where critical quality attributes must be quantified, like in drug development and screening.
Collapse
Affiliation(s)
- M Rasheed Anjum
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Brett R Higgins
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Carolina Abrahan
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Steven J Chisolm
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - K A Krishnaprasad
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Obiora Azie
- Otomagnetics, Inc., Bethesda, Maryland 20852, United States
| | - Glyn D Palmer
- Department of Orthopaedic Surgery and Sports Medicine, College of Medicine, University of Florida, Gainesville, Florida 32611, United States
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department of Materials Science and Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Malisa Sarntinoranont
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
32
|
Ye Y, Wang J, Izban MG, Ballard BR, Barsky SH. Initiation of tumor dormancy by the lymphovascular embolus. Oncotarget 2024; 15:726-740. [PMID: 39392391 PMCID: PMC11468568 DOI: 10.18632/oncotarget.28658] [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: 06/13/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Cancer dormancy followed by recurrence remains an enigma in cancer biology. Since both local and systemic recurrences are thought to emanate from dormant micrometastasis which take origin from lymphovascular tumor emboli we wondered whether the process of dormancy might initiate within lymphovascular emboli. This study combines experimental studies with a patient-derived xenograft (PDX) of inflammatory breast cancer (Mary-X) that spontaneously forms spheroids in vitro and budding lymphovascular tumor emboli in vivo with observational studies utilizing tissue microarrays (TMAs) of human breast cancers. In the experimental studies, Mary-X during both lymphovascular emboli formation in vivo and spheroidgenesis in vitro exhibited decreased proliferation, a G0/G1 cell cycle arrest and decreased mTOR signaling. This induction of dormancy required calpain-mediated E-cadherin proteolysis and was mediated by decreased P13K signaling, resulting in decreased mTOR activity. In observational human breast cancer studies, increased E-cadherin immunoreactivity due to increased E-cad/NTF-1 but both decreased Ki-67 and mTOR activity was observed selectively and differentially within the lymphovascular tumor emboli. Both our experimental as well as observational studies indicate that in vivo lymphovascular tumor emboli and their in vitro spheroid equivalent initiate dormancy through these pathways.
Collapse
Affiliation(s)
- Yin Ye
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| | - Justin Wang
- Department of Graduate Medical Education, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Michael G. Izban
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| | - Billy R. Ballard
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| | - Sanford H. Barsky
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
33
|
Xu M, Hong JJ, Zhang X, Sun M, Liu X, Kang J, Stack H, Fang W, Lei H, Lacoste X, Okada R, Jung R, Nguyen R, Shern JF, Thiele CJ, Liu Z. Targeting SWI/SNF ATPases reduces neuroblastoma cell plasticity. EMBO J 2024; 43:4522-4541. [PMID: 39174852 PMCID: PMC11480351 DOI: 10.1038/s44318-024-00206-1] [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: 09/26/2023] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Tumor cell heterogeneity defines therapy responsiveness in neuroblastoma (NB), a cancer derived from neural crest cells. NB consists of two primary subtypes: adrenergic and mesenchymal. Adrenergic traits predominate in NB tumors, while mesenchymal features becomes enriched post-chemotherapy or after relapse. The interconversion between these subtypes contributes to NB lineage plasticity, but the underlying mechanisms driving this phenotypic switching remain unclear. Here, we demonstrate that SWI/SNF chromatin remodeling complex ATPases are essential in establishing an mesenchymal gene-permissive chromatin state in adrenergic-type NB, facilitating lineage plasticity. Targeting SWI/SNF ATPases with SMARCA2/4 dual degraders effectively inhibits NB cell proliferation, invasion, and notably, cellular plasticity, thereby preventing chemotherapy resistance. Mechanistically, depletion of SWI/SNF ATPases compacts cis-regulatory elements, diminishes enhancer activity, and displaces core transcription factors (MYCN, HAND2, PHOX2B, and GATA3) from DNA, thereby suppressing transcriptional programs associated with plasticity. These findings underscore the pivotal role of SWI/SNF ATPases in driving intrinsic plasticity and therapy resistance in neuroblastoma, highlighting an epigenetic target for combinational treatments in this cancer.
Collapse
Affiliation(s)
- Man Xu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason J Hong
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xiyuan Zhang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ming Sun
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xingyu Liu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jeeyoun Kang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Hannah Stack
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wendy Fang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xavier Lacoste
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Reona Okada
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Raina Jung
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jack F Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Carol J Thiele
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Zhihui Liu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
34
|
Zhang Q, Zhang M. Recent advances in lung cancer organoid (tumoroid) research (Review). Exp Ther Med 2024; 28:383. [PMID: 39161616 PMCID: PMC11332118 DOI: 10.3892/etm.2024.12672] [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: 07/13/2023] [Accepted: 02/01/2024] [Indexed: 08/21/2024] Open
Abstract
Lung cancer is the most critical type of malignant tumor that threatens human health. Traditional preclinical models have certain defects; for example, they cannot accurately reflect the characteristics of lung cancer and their development is costly and time-consuming. Through self-organization, cancer stem cells (CSCs) generate cancer organoids that have a structure similar to that of lung cancer tissues, overcoming to some extent the aforementioned challenges, thus enabling them to have broader application prospects. Lung cancer organoid (LCO) development methods can be divided into three broad categories based on the source of cells, which include cell lines, patient-derived xenografts and patient tumor tissue/pleural effusion. There are 17 different methods that have been described for the development of LCOs. These methods can be further merged into six categories based on the source of cells, the pre-treatment method used, the composition of the medium and the culture scaffold. These categories are: i) CSCs induced by defined transcription factors; ii) suspension culture; iii) relative optimal culture medium; iv) suboptimal culture medium; v) mechanical digestion and suboptimal culture medium; and vi) hydrogel scaffold. In the current review, the advantages and disadvantages of each of the aforementioned methods are summarized, and references for supporting studies are cited.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Clinical Laboratory, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Mingyang Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
35
|
Dakal TC, Bhushan R, Xu C, Gadi BR, Cameotra SS, Yadav V, Maciaczyk J, Schmidt‐Wolf IGH, Kumar A, Sharma A. Intricate relationship between cancer stemness, metastasis, and drug resistance. MedComm (Beijing) 2024; 5:e710. [PMID: 39309691 PMCID: PMC11416093 DOI: 10.1002/mco2.710] [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: 03/13/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cancer stem cells (CSCs) are widely acknowledged as the drivers of tumor initiation, epithelial-mesenchymal transition (EMT) progression, and metastasis. Originating from both hematologic and solid malignancies, CSCs exhibit quiescence, pluripotency, and self-renewal akin to normal stem cells, thus orchestrating tumor heterogeneity and growth. Through a dynamic interplay with the tumor microenvironment (TME) and intricate signaling cascades, CSCs undergo transitions from differentiated cancer cells, culminating in therapy resistance and disease recurrence. This review undertakes an in-depth analysis of the multifaceted mechanisms underlying cancer stemness and CSC-mediated resistance to therapy. Intrinsic factors encompassing the TME, hypoxic conditions, and oxidative stress, alongside extrinsic processes such as drug efflux mechanisms, collectively contribute to therapeutic resistance. An exploration into key signaling pathways, including JAK/STAT, WNT, NOTCH, and HEDGEHOG, sheds light on their pivotal roles in sustaining CSCs phenotypes. Insights gleaned from preclinical and clinical studies hold promise in refining drug discovery efforts and optimizing therapeutic interventions, especially chimeric antigen receptor (CAR)-T cell therapy, cytokine-induced killer (CIK) cell therapy, natural killer (NK) cell-mediated CSC-targeting and others. Ultimately use of cell sorting and single cell sequencing approaches for elucidating the fundamental characteristics and resistance mechanisms inherent in CSCs will enhance our comprehension of CSC and intratumor heterogeneity, which ultimately would inform about tailored and personalized interventions.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology LabDepartment of BiotechnologyMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Ravi Bhushan
- Department of ZoologyM.S. CollegeMotihariBiharIndia
| | - Caiming Xu
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research InstituteCity of HopeMonroviaCaliforniaUSA
| | - Bhana Ram Gadi
- Stress Physiology and Molecular Biology LaboratoryDepartment of BotanyJai Narain Vyas UniversityJodhpurRajasthanIndia
| | | | - Vikas Yadav
- School of Life SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Jarek Maciaczyk
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
| | - Ingo G. H. Schmidt‐Wolf
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| | - Abhishek Kumar
- Manipal Academy of Higher EducationManipalKarnatakaIndia
- Institute of BioinformaticsInternational Technology ParkBangaloreIndia
| | - Amit Sharma
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
36
|
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
|
37
|
Karlsson JW, Sah VR, Olofsson Bagge R, Kuznetsova I, Iqba M, Alsen S, Stenqvist S, Saxena A, Ny L, Nilsson LM, Nilsson JA. Patient-derived xenografts and single-cell sequencing identifies three subtypes of tumor-reactive lymphocytes in uveal melanoma metastases. eLife 2024; 12:RP91705. [PMID: 39312285 PMCID: PMC11419671 DOI: 10.7554/elife.91705] [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] [Indexed: 09/25/2024] Open
Abstract
Uveal melanoma (UM) is a rare melanoma originating in the eye's uvea, with 50% of patients experiencing metastasis predominantly in the liver. In contrast to cutaneous melanoma, there is only a limited effectiveness of combined immune checkpoint therapies, and half of patients with uveal melanoma metastases succumb to disease within 2 years. This study aimed to provide a path toward enhancing immunotherapy efficacy by identifying and functionally validating tumor-reactive T cells in liver metastases of patients with UM. We employed single-cell RNA-seq of biopsies and tumor-infiltrating lymphocytes (TILs) to identify potential tumor-reactive T cells. Patient-derived xenograft (PDX) models of UM metastases were created from patients, and tumor sphere cultures were generated from these models for co-culture with autologous or MART1-specific HLA-matched allogenic TILs. Activated T cells were subjected to TCR-seq, and the TCRs were matched to those found in single-cell sequencing data from biopsies, expanded TILs, and in livers or spleens of PDX models injected with TILs. Our findings revealed that tumor-reactive T cells resided not only among activated and exhausted subsets of T cells, but also in a subset of cytotoxic effector cells. In conclusion, combining single-cell sequencing and functional analysis provides valuable insights into which T cells in UM may be useful for cell therapy amplification and marker selection.
Collapse
Affiliation(s)
- Joakim W Karlsson
- Harry Perkins Institute of Medical Research and University of Western AustraliaPerthAustralia
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Vasu R Sah
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Surgery, Sahlgrenska University HospitalGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, University of GothenburgGothenburgSweden
| | - Irina Kuznetsova
- Harry Perkins Institute of Medical Research and University of Western AustraliaPerthAustralia
| | - Munir Iqba
- Genomics WA, Telethon Kids Institute, Harry Perkins Institute of Medical Research and University of Western AustraliaNedlandsAustralia
| | - Samuel Alsen
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Sofia Stenqvist
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Alka Saxena
- Genomics WA, Telethon Kids Institute, Harry Perkins Institute of Medical Research and University of Western AustraliaNedlandsAustralia
| | - Lars Ny
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Oncology, Sahlgrenska University HospitalGothenburgSweden
| | - Lisa M Nilsson
- Harry Perkins Institute of Medical Research and University of Western AustraliaPerthAustralia
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Jonas A Nilsson
- Harry Perkins Institute of Medical Research and University of Western AustraliaPerthAustralia
- Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
38
|
Kraski A, Migdał P, Klopfleisch R, Räckel C, Sharbati J, Heimesaat MM, Alter T, Hanisch C, Gölz G, Einspanier R, Sharbati S. Structured multicellular intestinal spheroids (SMIS) as a standardized model for infection biology. Gut Pathog 2024; 16:47. [PMID: 39289703 PMCID: PMC11406839 DOI: 10.1186/s13099-024-00644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND 3D cell culture models have recently garnered increasing attention for replicating organ microarchitecture and eliciting in vivo-like responses, holding significant promise across various biological disciplines. Broadly, 3D cell culture encompasses organoids as well as single- and multicellular spheroids. While the latter have found successful applications in tumor research, there is a notable scarcity of standardized intestinal models for infection biology that mimic the microarchitecture of the intestine. Hence, this study aimed to develop structured multicellular intestinal spheroids (SMIS) specifically tailored for studying molecular basis of infection by intestinal pathogens. RESULTS We have successfully engineered human SMIS comprising four relevant cell types, featuring a fibroblast core enveloped by an outer monolayer of enterocytes and goblet cells along with monocytic cells. These SMIS effectively emulate the in vivo architecture of the intestinal mucosal surface and manifest differentiated morphological characteristics, including the presence of microvilli, within a mere two days of culture. Through analysis of various differentiation factors, we have illustrated that these spheroids attain heightened levels of differentiation compared to 2D monolayers. Moreover, SMIS serve as an optimized intestinal infection model, surpassing the capabilities of traditional 2D cultures, and exhibit a regulatory pattern of immunological markers similar to in vivo infections after Campylobacter jejuni infection. Notably, our protocol extends beyond human spheroids, demonstrating adaptability to other species such as mice and pigs. CONCLUSION Based on the rapid attainment of enhanced differentiation states, coupled with the emergence of functional brush border features, increased cellular complexity, and replication of the intestinal mucosal microarchitecture, which allows for exposure studies via the medium, we are confident that our innovative SMIS model surpasses conventional cell culture methods as a superior model. Moreover, it offers advantages over stem cell-derived organoids due to scalability and standardization capabilities of the protocol. By showcasing differentiated morphological attributes, our model provides an optimal platform for diverse applications. Furthermore, the investigated differences of several immunological factors compared to monotypic monolayers after Campylobacter jejuni infection underline the refinement of our spheroid model, which closely mimics important features of in vivo infections.
Collapse
Affiliation(s)
- Angelina Kraski
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Paweł Migdał
- Institute of Animal Husbandry and Breeding, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Clara Räckel
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Alter
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | | | - Greta Gölz
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
39
|
Martinez-Bernabe T, Morla-Barcelo PM, Melguizo-Salom L, Munar-Gelabert M, Maroto-Blasco A, Torrens-Mas M, Oliver J, Roca P, Nadal-Serrano M, Pons DG, Sastre-Serra J. Tumorspheres as In Vitro Model for Identifying Predictive Chemoresistance and Tumor Aggressiveness Biomarkers in Breast and Colorectal Cancer. BIOLOGY 2024; 13:724. [PMID: 39336151 PMCID: PMC11429065 DOI: 10.3390/biology13090724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
Chemoresistance remains a major challenge in the treatment of breast and colorectal cancer. For this reason, finding reliable predictive biomarkers of response to chemotherapy has become a significant research focus in recent years. However, validating in vitro results may be problematic due to the outcome heterogeneity. In this study, we evaluate the use of tumorspheres as an in vitro model for validating biomarkers of chemoresistance in breast and colorectal cancer. Our investigation highlights the crucial role of inflammation-related pathways in modulating the response to chemotherapy. Using in silico approaches, we identified specific markers elevated in responders versus non-responders patients. These markers were consistently higher in three-dimensional (3D) tumorsphere models compared to traditional adherent cell culture models. Furthermore, the number of tumorspheres from breast and colorectal cancer cells increased in response to cisplatin and oxaliplatin treatment, respectively, whereas cell viability decreased in adherent cell culture. This differential response underscores the importance of the 3D tumorsphere model in mimicking the tumor microenvironment more accurately than adherent cell culture. The enhanced chemoresistance observed in the 3D tumorspheres model and their correlation of data with the in silico study suggest that 3D culture models are a better option to approach the in vivo model and also to validate in silico data. Our findings indicate that tumorspheres are an ideal model for validating chemoresistance biomarkers and exploring the interplay between inflammation and chemoresistance in breast and colon cancer.
Collapse
Affiliation(s)
- Toni Martinez-Bernabe
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Pere Miquel Morla-Barcelo
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Lucas Melguizo-Salom
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Margalida Munar-Gelabert
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
| | - Alba Maroto-Blasco
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
| | - Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Jordi Oliver
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Mercedes Nadal-Serrano
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma de Mallorca, Spain; (T.M.-B.); (P.M.M.-B.); (L.M.-S.); (M.M.-G.); (A.M.-B.); (M.T.-M.); (J.O.); (P.R.); (M.N.-S.); (J.S.-S.)
- Instituto de Investigación Sanitaria de las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120 Palma de Mallorca, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
40
|
Lim K, Han SH, Han S, Lee JY, Choi HS, Choi D, Ryu CJ. A monoclonal antibody recognizing CD98 on human embryonic stem cells shows anti-tumor activity in hepatocellular carcinoma xenografts. Cancer Immunol Immunother 2024; 73:231. [PMID: 39261363 PMCID: PMC11390997 DOI: 10.1007/s00262-024-03827-x] [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: 06/15/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
CD98, also known as SLC3A2, is a multifunctional cell surface molecule consisting of amino acid transporters. CD98 is ubiquitously expressed in many types of tissues, but expressed at higher levels in cancerous tissues than in normal tissues. CD98 is also upregulated in most hepatocellular carcinoma (HCC) patients; however, the function of CD98 in HCC cells has been little studied. In this study, we generated a panel of monoclonal antibodies (MAbs) against surface proteins on human embryonic stem cells (hESCs). NPB15, one of the MAbs, bound to hESCs and various cancer cells, including HCC cells and non-small cell lung carcinoma (NSCLC) cells, but not to peripheral blood mononuclear cells (PBMCs) and primary hepatocytes. Immunoprecipitation and mass spectrometry identified the target antigen of NPB15 as CD98. CD98 depletion decreased cell proliferation, clonogenic survival, and migration and induced apoptosis in HCC cells. In addition, CD98 depletion decreased the expression of cancer stem cell (CSC) markers in HCC cells. In tumorsphere cultures, the expression of CD98 interacting with NPB15 was significantly increased, as were known CSC markers. After cell sorting by NPB15, cells with high expression of CD98 (CD98-high) showed higher clonogenic survival than cells with low expression of CD98 (CD98-low) in HCC cells, suggesting CD98 as a potential CSC marker on HCC cells. The chimeric version of NPB15 was able to induce antibody-dependent cellular cytotoxicity (ADCC) against HCC cells in vitro. NPB15 injection showed antitumor activity in an HCC xenograft mouse model. The results suggest that NPB15 may be developed as a therapeutic antibody for HCC patients.
Collapse
Affiliation(s)
- Keunpyo Lim
- Department of Integrative Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 05006, Republic of Korea
| | - San Ha Han
- Department of Integrative Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 05006, Republic of Korea
| | - Sein Han
- Department of Integrative Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 05006, Republic of Korea
| | - Ji Yoon Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 05006, Republic of Korea
| | - Hong Seo Choi
- Department of Integrative Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 05006, Republic of Korea
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea
| | - Chun Jeih Ryu
- Department of Integrative Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 05006, Republic of Korea.
| |
Collapse
|
41
|
Xu FX, Sun R, Owens R, Hu K, Fu D. Assessing Drug Uptake and Response Differences in 2D and 3D Cellular Environments Using Stimulated Raman Scattering Microscopy. Anal Chem 2024; 96:14480-14489. [PMID: 39186736 DOI: 10.1021/acs.analchem.4c02592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The architecture of cell culture, two-dimensional (2D) versus three-dimensional (3D), significantly impacts various cellular factors, including cell-cell interactions, nutrient and oxygen gradients, metabolic activity, and gene expression profiles. This can result in different cellular responses during cancer drug treatment, with 3D-cultured cells often exhibiting higher resistance to chemotherapeutic drugs. While various genetic and proteomic analyses have been employed to investigate the underlying mechanisms of this increased resistance, complementary techniques that provide experimental evidence of spatial molecular profiling data are limited. Stimulated Raman scattering (SRS) microscopy has demonstrated its capability to measure both intracellular drug uptake and growth inhibition. In this work, we applied three-band (C-D, C-H, and fingerprint regions) SRS imaging to 2D and 3D cell cultures and performed a comparative analysis of drug uptake and response with the goal of understanding whether the difference in drug uptake explains the drug resistance in 3D culture compared to 2D. Our investigations revealed that despite similar intracellular drug levels in 2D and 3D A549 cells during lapatinib treatment, the growth of 3D spheroids was less impacted, supporting an enhanced drug tolerance in the 3D microenvironment. We further elucidated drug penetration patterns and the resulting heterogeneous cellular responses across different spheroid layers. Additionally, we investigated the role of the extracellular matrix in modulating drug delivery and cell response and discovered that limited drug penetration in 3D could also contribute to lower drug response. Our study provides valuable insights into the intricate mechanisms of increased drug resistance in 3D tumor models during cancer drug treatments.
Collapse
Affiliation(s)
- Fiona Xi Xu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Rui Sun
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Ryan Owens
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Kailun Hu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
42
|
Nguyen TNH, Horowitz LF, Krilov T, Lockhart E, Kenerson HL, Gujral TS, Yeung RS, Arroyo-Currás N, Folch A. Label-free, real-time monitoring of cytochrome C drug responses in microdissected tumor biopsies with a multi-well aptasensor platform. SCIENCE ADVANCES 2024; 10:eadn5875. [PMID: 39241078 PMCID: PMC11378948 DOI: 10.1126/sciadv.adn5875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/31/2024] [Indexed: 09/08/2024]
Abstract
Functional assays on intact tumor biopsies can complement genomics-based approaches for precision oncology, drug testing, and organs-on-chips cancer disease models by capturing key therapeutic response determinants, such as tissue architecture, tumor heterogeneity, and the tumor microenvironment. Most of these assays rely on fluorescent labeling, a semiquantitative method best suited for single-time-point assays or labor-intensive immunostaining analysis. Here, we report integrated aptamer electrochemical sensors for on-chip, real-time monitoring of cytochrome C, a cell death indicator, from intact microdissected tissues with high affinity and specificity. The platform features a multi-well sensor layout and a multiplexed electronic setup. The aptasensors measure increases in cytochrome C in the supernatant of mouse or human microdissected tumors after exposure to various drug treatments. Because of the sensor's high affinity, it primarily tracks rising concentrations of cytochrome C, capturing dynamic changes during apoptosis. This approach could help develop more advanced cancer disease models and apply to other complex in vitro disease models, such as organs-on-chips and organoids.
Collapse
Affiliation(s)
- Tran N. H. Nguyen
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Lisa F. Horowitz
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Timothy Krilov
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Ethan Lockhart
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Heidi L. Kenerson
- Department of Surgery, University of Washington, Seattle, WA 98105, USA
| | - Taranjit S. Gujral
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98105, USA
| | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, WA 98105, USA
| | | | - Albert Folch
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
43
|
Hu L, Cohen RI, Barroso M, Boustany NN. Comparison of vinculin tension in cellular monolayers and three-dimensional multicellular aggregates. BIOMEDICAL OPTICS EXPRESS 2024; 15:5199-5214. [PMID: 39296399 PMCID: PMC11407257 DOI: 10.1364/boe.529156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/21/2024]
Abstract
Confocal frequency-domain fluorescence lifetime and Förster resonance energy transfer (FRET) microscopy of Chinese hamster ovary (CHO-K1) cells expressing the vinculin tension sensor (VinTS) is used to compare vinculin tension in three-dimensional (3D) multicellular aggregates and 2D cellular monolayers. In both 2D and 3D cultures, the FRET efficiency of VinTS is 5-6% lower than that of VinTL (p < 0.05), a tail-less control which cannot bind actin or paxillin. The difference between VinTS and VinTL FRET efficiency can be mitigated by treatment with the Rho-associated kinase inhibitor Y-27632, demonstrating that VinTS is under tension in both 2D and 3D cultures. However, there is an overall decrease in FRET efficiency of both VinTS and VinTL in the 3D multicellular aggregates compared with the 2D monolayers. Expression of VinTS in 2D and 3D cultures exhibits puncta consistent with cellular adhesions. While paxillin is present at the sites of VinTS expression in the 2D monolayers, it is generally absent from VinTS puncta in the 3D aggregates. The results suggest that VinTS experiences a modified environment in 3D aggregates compared with 2D monolayers and provide a basis for further investigation of molecular tension sensors in 3D tissue models.
Collapse
Affiliation(s)
- Luni Hu
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Nada N Boustany
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
44
|
Chen C, Boché A, Wang Z, Lopez E, Peng J, Carreiras F, Schanne-Klein MC, Chen Y, Lambert A, Aimé C. The Balance Between Shear Flow and Extracellular Matrix in Ovarian Cancer-on-Chip. Adv Healthc Mater 2024; 13:e2400938. [PMID: 38829702 DOI: 10.1002/adhm.202400938] [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: 03/12/2024] [Revised: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Ovarian cancer is the most lethal gynecologic cancer in developed countries. In the tumor microenvironment, the extracellular matrix (ECM) and flow shear stress are key players in directing ovarian cancer cells invasion. Artificial ECM models based only on ECM proteins are used to build an ovarian tumor-on-chip to decipher the crosstalk between ECM and shear stress on the migratory behavior and cellular heterogeneity of ovarian tumor cells. This work shows that in the shear stress regime of the peritoneal cavity, the ECM plays a major role in driving individual or collective ovarian tumor cells migration. In the presence of basement membrane proteins, migration is more collective than on type I collagen regardless of shear stress. With increasing shear stress, individual cell migration is enhanced; while, no significant impact on collective migration is measured. This highlights the central position that ECM and flow shear stress should hold in in vitro ovarian cancer models to deepen understanding of cellular responses and improve development of ovarian cancer therapeutic platforms. In this frame, adding flow provides significant improvement in biological relevance over the authors' previous work. Further steps for enhanced clinical relevance require not only multiple cell lines but also patient-derived cells and sera.
Collapse
Affiliation(s)
- Changchong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Alphonse Boché
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Zixu Wang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Elliot Lopez
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Juan Peng
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Marie-Claire Schanne-Klein
- Laboratoire d'Optique et Biosciences (LOB), École polytechnique, CNRS, Inserm, Institut Polytechnique de Paris, Palaiseau, F-91128, France
| | - Yong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Carole Aimé
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| |
Collapse
|
45
|
Kot M, Simiczyjew A, Wądzyńska J, Ziętek M, Matkowski R, Nowak D. Characterization of two melanoma cell lines resistant to BRAF/MEK inhibitors (vemurafenib and cobimetinib). Cell Commun Signal 2024; 22:410. [PMID: 39175042 PMCID: PMC11342534 DOI: 10.1186/s12964-024-01788-3] [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: 02/22/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND BRAF (v-raf murine sarcoma viral oncogene homolog B1)/MEK (mitogen-activated protein kinase kinase) inhibitors are used for melanoma treatment. Unfortunately, patients treated with this combined therapy develop resistance to treatment quite quickly, but the mechanisms underlying this phenomenon are not yet fully understood. Here, we report and characterize two melanoma cell lines (WM9 and Hs294T) resistant to BRAF (vemurafenib) and MEK (cobimetinib) inhibitors. METHODS Cell viability was assessed via the XTT test. The level of selected proteins as well as activation of signaling pathways were evaluated using Western blotting. The expression of the chosen genes was assessed by RT-PCR. The distribution of cell cycle phases was analyzed by flow cytometry, and confocal microscopy was used to take photos of spheroids. The composition of cytokines secreted by cells was determined using a human cytokine array. RESULTS The resistant cells had increased survival and activation of ERK kinase in the presence of BRAF/MEK inhibitors. The IC50 values for these cells were over 1000 times higher than for controls. Resistant cells also exhibited elevated activation of AKT, p38, and JNK signaling pathways with increased expression of EGFR, ErbB2, MET, and PDGFRβ receptors as well as reduced expression of ErbB3 receptor. Furthermore, these cells demonstrated increased expression of genes encoding proteins involved in drug transport and metabolism. Resistant cells also exhibited features of epithelial-mesenchymal transition and cancer stem cells as well as reduced proliferation rate and elevated cytokine secretion. CONCLUSIONS In summary, this work describes BRAF/MEK-inhibitor-resistant melanoma cells, allowing for better understanding the underlying mechanisms of resistance. The results may thus contribute to the development of new, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Magdalena Kot
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland.
| | - Justyna Wądzyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Marcin Ziętek
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Rafał Matkowski
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| |
Collapse
|
46
|
Brooks A, Zhang Y, Chen J, Zhao CX. Cancer Metastasis-on-a-Chip for Modeling Metastatic Cascade and Drug Screening. Adv Healthc Mater 2024; 13:e2302436. [PMID: 38224141 DOI: 10.1002/adhm.202302436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Microfluidic chips are valuable tools for studying intricate cellular and cell-microenvironment interactions. Traditional in vitro cancer models lack accuracy in mimicking the complexities of in vivo tumor microenvironment. However, cancer-metastasis-on-a-chip (CMoC) models combine the advantages of 3D cultures and microfluidic technology, serving as powerful platforms for exploring cancer mechanisms and facilitating drug screening. These chips are able to compartmentalize the metastatic cascade, deepening the understanding of its underlying mechanisms. This article provides an overview of current CMoC models, focusing on distinctive models that simulate invasion, intravasation, circulation, extravasation, and colonization, and their applications in drug screening. Furthermore, challenges faced by CMoC and microfluidic technologies are discussed, while exploring promising future directions in cancer research. The ongoing development and integration of these models into cancer studies are expected to drive transformative advancements in the field.
Collapse
Affiliation(s)
- Anastasia Brooks
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Jiezhong Chen
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
47
|
Rahman MM, Wells G, Rantala JK, Helleday T, Muthana M, Danson SJ. In-vitro assays for immuno-oncology drug efficacy assessment and screening for personalized cancer therapy: scopes and challenges. Expert Rev Clin Immunol 2024; 20:821-838. [PMID: 38546609 DOI: 10.1080/1744666x.2024.2336583] [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: 11/30/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Immunotherapies have revolutionized cancer treatment, but often fail to produce desirable therapeutic outcomes in all patients. Due to the inter-patient heterogeneity and complexity of the tumor microenvironment, personalized treatment approaches are gaining demand. Researchers have long been using a range of in-vitro assays including 2D models, organoid co-cultures, and cancer-on-a-chip platforms for cancer drug screening. A comparative analysis of these assays with their suitability, high-throughput capacity, and clinical translatability is required for optimal translational use. AREAS COVERED The review summarized in-vitro platforms with their comparative advantages and limitations including construction strategies, and translational potential for immuno-oncology drug efficacy assessment. We also discussed end-point analysis strategies so that researchers can contextualize their usefulness and optimally design experiments for personalized immunotherapy efficacy prediction. EXPERT OPINION Researchers developed several in-vitro platforms that can provide information on personalized immunotherapy efficacy from different angles. Image-based assays are undoubtedly more suitable to gather a wide range of information including cellular morphology and phenotypical behaviors but need significant improvement to overcome issues including background noise, sample preparation difficulty, and long duration of experiment. More studies and clinical trials are needed to resolve these issues and validate the assays before they can be used in real-life scenarios.
Collapse
Affiliation(s)
- Md Marufur Rahman
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Directorate General of Health Services, Dhaka, Bangladesh
| | - Greg Wells
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Juha K Rantala
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Misvik Biology Ltd, Turku, Finland
| | - Thomas Helleday
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Department of Oncology-Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Munitta Muthana
- Nanobug Oncology Sheffield, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Sarah J Danson
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
48
|
Young KA, Wojdyla K, Lai T, Mulholland KE, Aldaz Casanova S, Antrobus R, Andrews SR, Biggins L, Mahler-Araujo B, Barton PR, Anderson KR, Fearnley GW, Sharpe HJ. The receptor protein tyrosine phosphatase PTPRK promotes intestinal repair and catalysis-independent tumour suppression. J Cell Sci 2024; 137:jcs261914. [PMID: 38904097 PMCID: PMC11298714 DOI: 10.1242/jcs.261914] [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: 12/18/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
PTPRK is a receptor tyrosine phosphatase that is linked to the regulation of growth factor signalling and tumour suppression. It is stabilized at the plasma membrane by trans homophilic interactions upon cell-cell contact. PTPRK regulates cell-cell adhesion but is also reported to regulate numerous cancer-associated signalling pathways. However, the signalling mechanism of PTPRK remains to be determined. Here, we find that PTPRK regulates cell adhesion signalling, suppresses invasion and promotes collective, directed migration in colorectal cancer cells. In vivo, PTPRK supports recovery from inflammation-induced colitis. In addition, we confirm that PTPRK functions as a tumour suppressor in the mouse colon and in colorectal cancer xenografts. PTPRK regulates growth factor and adhesion signalling, and suppresses epithelial to mesenchymal transition (EMT). Contrary to the prevailing notion that PTPRK directly dephosphorylates EGFR, we find that PTPRK regulation of both EGFR and EMT is independent of its catalytic function. This suggests that additional adaptor and scaffold functions are important features of PTPRK signalling.
Collapse
Affiliation(s)
| | | | - Tiffany Lai
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| | | | | | - Robin Antrobus
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | | | - Laura Biggins
- Bioinformatics, Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Philippa R. Barton
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Keith R. Anderson
- Molecular biology department, Genentech, South San Francisco, CA 94080, USA
| | | | - Hayley J. Sharpe
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| |
Collapse
|
49
|
Nielsen BS, Madsen NH, Larsen J, Skandorff I, Gad M, Holmstrøm K. Architectural organization and molecular profiling of 3D cancer heterospheroids and their application in drug testing. Front Oncol 2024; 14:1386097. [PMID: 39011470 PMCID: PMC11246882 DOI: 10.3389/fonc.2024.1386097] [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: 02/14/2024] [Accepted: 05/31/2024] [Indexed: 07/17/2024] Open
Abstract
3D cancer cell cultures have enabled new opportunities for replacing compound testing in experimental animals. However, most solid tumors are composed of multiple cell types, including fibroblasts. In this study we developed multicellular tumor heterospheroids composed of cancer and fibroblasts cell lines. We developed heterospheroids by combining HT-29, MCF-7, PANC-1 or SW480 with 1BR.3.G fibroblasts, which we have previously reported support spheroid formation. We also tested fibroblast cell lines, MRC-5, GM00498 and HIF, but 1BR.3.G was found to best form heterospheroids with morphological similarity to in vivo tumor tissue. The architectural organization of heterospheroids was based on histological examination using immunohistochemistry. We found that HT-29 and MCF-7 cells developed spheroids with the cancer cells surrounding the fibroblasts, whereas PANC-1 cells interspersed with the fibroblasts and SW480 cells were surrounded by fibroblasts. The fibroblasts also expressed collagen-1 and FAP-α, and whole transcriptomic analysis (WTA) showed abundant ECM- and EMT-related expression in heterospheroids, thus reflecting a representative tumor-like microenvironment. The WTA showed that PANC-1 heterospheroids possess a strong EMT profile with abundant Vimentin and CDH2 expression. Drug testing was evaluated by measuring cytotoxicity of 5FU and cisplatin using cell viability and apoptosis assays. We found no major impact on the cytotoxicity when fibroblasts were added to the spheroids. We conclude that the cancer cell lines together with fibroblasts shape the architectural organization of heterospheroids to form tumor-like morphology, and we propose that the various 3D tumor structures can be used for drug testing directed against the cancer cells as well as the fibroblasts.
Collapse
Affiliation(s)
- Boye Schnack Nielsen
- Department of Cellular Engineering & Disease Modeling, Bioneer A/S, Hørsholm, Denmark
| | | | | | | | | | | |
Collapse
|
50
|
Thorel L, Perréard M, Florent R, Divoux J, Coffy S, Vincent A, Gaggioli C, Guasch G, Gidrol X, Weiswald LB, Poulain L. Patient-derived tumor organoids: a new avenue for preclinical research and precision medicine in oncology. Exp Mol Med 2024; 56:1531-1551. [PMID: 38945959 PMCID: PMC11297165 DOI: 10.1038/s12276-024-01272-5] [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: 11/24/2023] [Revised: 03/18/2024] [Accepted: 04/14/2024] [Indexed: 07/02/2024] Open
Abstract
Over the past decade, the emergence of patient-derived tumor organoids (PDTOs) has broadened the repertoire of preclinical models and progressively revolutionized three-dimensional cell culture in oncology. PDTO can be grown from patient tumor samples with high efficiency and faithfully recapitulates the histological and molecular characteristics of the original tumor. Therefore, PDTOs can serve as invaluable tools in oncology research, and their translation to clinical practice is exciting for the future of precision medicine in oncology. In this review, we provide an overview of methods for establishing PDTOs and their various applications in cancer research, starting with basic research and ending with the identification of new targets and preclinical validation of new anticancer compounds and precision medicine. Finally, we highlight the challenges associated with the clinical implementation of PDTO, such as its representativeness, success rate, assay speed, and lack of a tumor microenvironment. Technological developments and autologous cocultures of PDTOs and stromal cells are currently ongoing to meet these challenges and optimally exploit the full potential of these models. The use of PDTOs as standard tools in clinical oncology could lead to a new era of precision oncology in the coming decade.
Collapse
Grants
- AP-RM-19-020 Fondation de l'Avenir pour la Recherche Médicale Appliquée (Fondation de l'Avenir)
- PJA20191209649 Fondation ARC pour la Recherche sur le Cancer (ARC Foundation for Cancer Research)
- TRANSPARANCE Fondation ARC pour la Recherche sur le Cancer (ARC Foundation for Cancer Research)
- TRANSPARANCE Ligue Contre le Cancer
- ORGAPRED Ligue Contre le Cancer
- 3D-Hub Canceropôle PACA (Canceropole PACA)
- Pré-néo 2019-188 Institut National Du Cancer (French National Cancer Institute)
- Conseil Régional de Haute Normandie (Upper Normandy Regional Council)
- GIS IBiSA, Cancéropôle Nord-Ouest (ORGRAFT project), the Groupement des Entreprises Françaises dans la Lutte contre le Cancer (ORGAVADS project), the Fonds de dotation Patrick de Brou de Laurière (ORGAVADS project),and Normandy County Council (ORGATHEREX project).
- GIS IBiSA, Cancéropôle Nord-Ouest (OrgaNO project), Etat-région
- GIS IBiSA, Region Sud
- GIS IBiSA, Cancéropôle Nord-Ouest (OrgaNO project), and Normandy County Council (ORGAPRED, PLATONUS ONE, POLARIS, and EQUIP’INNOV projects).
Collapse
Affiliation(s)
- Lucie Thorel
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Marion Perréard
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Department of Head and Neck Surgery, Caen University Hospital, Caen, France
| | - Romane Florent
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Jordane Divoux
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France
| | - Sophia Coffy
- Biomics, CEA, Inserm, IRIG, UA13 BGE, Univ. Grenoble Alpes, Grenoble, France
| | - Audrey Vincent
- CNRS UMR9020, INSERM U1277, CANTHER Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, CNRS, Inserm, CHU Lille, Lille, France
| | - Cédric Gaggioli
- CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), 3D-Hub-S Facility, CNRS University Côte d'Azur, Nice, France
| | - Géraldine Guasch
- CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Epithelial Stem Cells and Cancer Team, Aix-Marseille University, Marseille, France
| | - Xavier Gidrol
- Biomics, CEA, Inserm, IRIG, UA13 BGE, Univ. Grenoble Alpes, Grenoble, France
| | - Louis-Bastien Weiswald
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France.
| | - Laurent Poulain
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- ORGAPRED core facility, US PLATON, Université de Caen Normandie, Caen, France.
| |
Collapse
|