1
|
Duffield C, Rey Gomez LM, Hnit SST, Zhang W, Tsao SCH, Inglis DW, Wang Y. Multi-line lateral flow immunoassay for the detection and subtyping of breast cancer-derived small extracellular vesicles. Chem Commun (Camb) 2025. [PMID: 40395113 DOI: 10.1039/d5cc01038a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Cancer cell-derived small extracellular vesicles (sEVs) are new promising biomarkers for improved cancer diagnosis. A multi-line lateral flow immunoassay (LFIA) has been proposed for sensitive detection and subtyping of breast cancer-derived sEVs. The expression of cancer surface biomarkers (EpCAM) was confirmed through colorimetric analysis of cell line-derived sEVs (p = 0.076) and patient samples, highlighting the potential for their use as a clinical aid.
Collapse
Affiliation(s)
- Chloe Duffield
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Laura M Rey Gomez
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Su Su Thae Hnit
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Wei Zhang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - David W Inglis
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Australia
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| |
Collapse
|
2
|
Ngo CHL, Tukova A, Zhang W, Tsao SCH, Wang Y. Sensitive detection of small extracellular vesicles using a gold nanostar-based SERS assay. Analyst 2025; 150:2108-2117. [PMID: 40202797 DOI: 10.1039/d5an00110b] [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: 04/11/2025]
Abstract
Small extracellular vesicles (sEVs) are lipid bilayer-bound vesicles that carry critical biomarkers for disease detection. However, the inherent heterogeneity and complexity of sEV molecular characteristics pose significant challenges for accurate and comprehensive molecular profiling. Traditional analytical methods, including immunoblotting, enzyme-linked immunosorbent assay (ELISA), and flow cytometry, exhibit several limitations, such as being time-consuming, requiring large sample volumes, and demonstrating relatively low sensitivity. Therefore, there is an urgent need to develop a highly sensitive and specific assay for the reliable detection of sEVs. Surface-enhanced Raman scattering (SERS) assays have emerged as a promising approach for sEV detection, offering advantages including high sensitivity and specificity. In the proposed SERS assay, SERS nanotags - comprising nanoparticles coated with Raman-active molecules and conjugated with antibodies - are employed to label surface-bound molecules on sEVs. This approach facilitates the generation of a high-intensity signal from molecules present in low abundance. Recently, anisotropic nanoparticles, such as star-shaped nanostructures, have garnered interest due to their ability to significantly amplify generated SERS signals for ultra-sensitive biomarker detection. In this study, we explore the application of gold nanostars (AuNSs) as SERS nanotags for the detection of sEVs. In principle, AuNS-based SERS nanotags were used to label the EpCAM protein, which can be found on the surface of cancer cell derived sEVs, and then sEV labelled SERS nanotags were captured by CD9-conjugated magnetic beads to form an immunocomplex, which exhibits a SERS signal. Our results demonstrate that the proposed SERS assay utilizing AuNSs provides high specificity and sensitivity, with a detection limit as low as 2.47 × 103 particles per μL. Furthermore, the assay was tested with spiked plasma samples (cancer cell-derived sEVs spiked into healthy plasma), showing that its specificity remains unaffected by the presence of plasma. These findings suggest that the SERS assay incorporating AuNSs holds significant promise as an effective and reliable detection method for potential clinical applications.
Collapse
Affiliation(s)
| | - Anastasiia Tukova
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Macquarie University, Sydney, Australia.
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
3
|
Lee J, Kim I, Ryu J, Eling T, Baek SJ. NAG-1/GDF15 as a tumor suppressor in colorectal cancer: inhibition of β-catenin and NF-κB pathways via interaction with EpCAM. Cell Death Dis 2025; 16:355. [PMID: 40316530 PMCID: PMC12048721 DOI: 10.1038/s41419-025-07695-w] [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: 11/10/2024] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
NAG-1/GDF15, a tumor suppressor, is synthesized as a pro-form in colorectal cancer (CRC) cells and undergoes cleavage to generate its mature form. While the biological function of pro-NAG-1/GDF15 remains unclear, our study reveals its crucial role in suppressing oncogenic signaling. We demonstrate that pro-NAG-1/GDF15 is predominantly retained within cells, whereas its mature form is secreted into the media. The expression of NAG-1/GDF15, or uncleavable R193A mutant, inhibits β-catenin and NF-κB signaling, key pathways in CRC progression. Mechanistically, the pro-NAG-1/GDF15 interacts with EpCAM, preventing its cleavage and nuclear translocation, thereby reducing β-catenin and NF-κB activity. This inhibition correlates with decreased expression of oncogenic targets such as cyclin D1 and c-myc. In vivo, NAG-1/GDF15 expression significantly reduces tumor growth in cancer xenograft models, accompanied by decreased proliferation and increased apoptosis. Furthermore, analysis of public datasets suggests that high NAG-1/GDF15 expression is associated with improved CRC patient survival. These findings highlight NAG-1/GDF15 via the formation of pro-NAG-1/GDF15 as a promising therapeutic target for cancer, with potential applications in modulating tumorigenic signaling pathways.
Collapse
Affiliation(s)
- Jaehak Lee
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Ilju Kim
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Junsun Ryu
- Department of Otolaryngology-Head and Neck Surgery, Center for Thyroid Cancer, Research Institute and Hospital, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Thomas Eling
- Retired Scientist Emeritus, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Seung Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea.
| |
Collapse
|
4
|
Renata S, Verma N, Peddinti RK. Surface-enhanced Raman spectroscopy as effective tool for detection of sialic acid as cancer biomarker. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125631. [PMID: 39736186 DOI: 10.1016/j.saa.2024.125631] [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: 10/01/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025]
Abstract
Sialic acid, a negatively charged nine-carbon monosaccharide, is mainly located at the terminal end of glycan chains on glycoproteins and glycolipids of cell surface and most secreted proteins. Elevated levels of sialylated glycans have been known as a hallmark in numerous cancers. As a result, sialic acid acts as a useful and accessible cancer biomarker for early cancer detection and monitoring the disease development during cancer treatment which is crucial in elevating the survival rate. The detection of sialic acid has been done by many tools including surface-enhanced Raman spectroscopy (SERS) which gained incredible attention due to its high selectivity and sensitivity. However, currently, comprehensive reviews of sialic acid detection and imaging as a cancer biomarker using SERS are still lacking. Here, we present the significant breakthroughs in SERS-based detection of sialic acid levels on cells, tissues, and body fluids due to the presence of cancer, different cancer metastasis stages, and in response to the external stimuli. This review covers the SERS substrate and novel SERS strategies, using lectin, boronic acid, metabolic glycan labelling and label-free methods, for sialic acid detection as cancer biomarker. The remaining challenges to detect sialic acid and prospect of future development of SERS for other carbohydrate-based cancer biomarker, for instance fucose, are also discussed.
Collapse
Affiliation(s)
- Septila Renata
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| | - Nitish Verma
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Department of Chemistry, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Rama Krishna Peddinti
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
5
|
Qian X, Yi W, Yan W, Cai Y, Hu S, Yan D, Zhao Z, Li R, Wang L, Xu H, Li Y. Cryo-Shocked Tumor-Reprogrammed Sonosensitive Antigen-Presenting Cells Improving Sonoimmunotherapy via T Cells and NK Cells Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413289. [PMID: 39955715 DOI: 10.1002/adma.202413289] [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: 09/05/2024] [Revised: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Ultrasound therapy has turned up as a noninvasive multifunctional tool for cancer immunotherapy. However, the insufficient co-stimulating molecules and loss of peptide-major histocompatibility complex I (MHC-I) expression on tumor cells lead to poor therapy of sonoimmunotherapies. Herein, this work develops a sonosensitive system to augment MHC-I unrestricted natural killer (NK) cell-mediated innate immunity and T cell-mediated adaptive immunity by leveraging antigen presentation cell (APC)-like tumor cells. Genetically engineered tumor cells featuring sufficient co-stimulating molecules are cryo-shocked and conjugated with a sonosensitizer, hematoporphyrin monomethyl ether, using click chemistry. These cells (DPNLs) exhibit characteristics of tumor and draining lymph node homing. Under ultrasound, NK cell-mediated innate immunity within the tumor microenvironment could be activated, and T cells in the tumor-draining lymph nodes (TDLNs) are stimulated through co-stimulatory molecules. In combination with programmed cell death ligand 1 (PD-L1) antibody, DPNLs extend the survival time and inhibited lung metastasis in triple-negative breast cancer (TNBC) models. This study provides an alternative approach for sonoimmunotherapy with precise sonosensitizer delivery and enhanced NK cell and T cell activation.
Collapse
Affiliation(s)
- Xindi Qian
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, School of Medicine, Tongji University, Shanghai, 200072, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Wenzhe Yi
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations Yantai Institute of Materia Medica Shandong, Shanghai, 264000, China
| | - Shuangshuang Hu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dan Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhiwen Zhao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rongzhang Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liying Wang
- Department of Medical Ultrasound and Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations Yantai Institute of Materia Medica Shandong, Shanghai, 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264000, China
| |
Collapse
|
6
|
Lim S, Chung HJ, Oh YJ, Hinterdorfer P, Myung SC, Seo Y, Ko K. Modification of Fc-fusion protein structures to enhance efficacy of cancer vaccine in plant expression system. PLANT BIOTECHNOLOGY JOURNAL 2025; 23:960-982. [PMID: 39724301 PMCID: PMC11869200 DOI: 10.1111/pbi.14552] [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: 10/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
Epithelial cell adhesion molecule (EpCAM) fused to IgG, IgA and IgM Fc domains was expressed to create IgG, IgA and IgM-like structures as anti-cancer vaccines in Nicotiana tabacum. High-mannose glycan structures were generated by adding a C-terminal endoplasmic reticulum (ER) retention motif (KDEL) to the Fc domain (FcK) to produce EpCAM-Fc and EpCAM-FcK proteins in transgenic plants via Agrobacterium-mediated transformation. Cross-fertilization of EpCAM-Fc (FcK) transgenic plants with Joining chain (J-chain, J and JK) transgenic plants led to stable expression of large quaternary EpCAM-IgA Fc (EpCAM-A) and IgM-like (EpCAM-M) proteins. Immunoblotting, SDS-PAGE and ELISA analyses demonstrated that proteins with KDEL had higher expression levels and binding activity to anti-EpCAM IgGs. IgM showed the strongest binding among the fusion proteins, followed by IgA and IgG. Sera from BALB/c mice immunized with these vaccines produced anti-EpCAM IgGs. Flow cytometry indicated that the EpCAM-Fc fusion proteins significantly activated CD8+ cytotoxic T cells, CD4+ helper T cells and B cells, particularly with EpCAM-FcKP and EpCAM-FcP (FcKP) × JP (JKP). The induced anti-EpCAM IgGs captured human prostate cancer PC-3 and colorectal cancer SW620 cells. Sera from immunized mice inhibited cancer cell proliferation, migration and invasion; down-regulated proliferation markers (PCNA, Ki-67) and epithelial-mesenchymal transition markers (Vimentin); and up-regulated E-cadherin. These findings suggest that N. tabacum can produce effective vaccine candidates to induce anti-cancer immune responses.
Collapse
Affiliation(s)
- Sohee Lim
- BioSystems Design Lab, Department of Medicine, College of MedicineChung‐Ang UniversitySeoulKorea
| | - Hyun Joo Chung
- Department of Urology, College of MedicineChung‐Ang UniversitySeoulKorea
| | - Yoo Jin Oh
- Department of Applied Experimental BiophysicsJohannes Kepler UniversityLinzAustria
| | - Peter Hinterdorfer
- Department of Applied Experimental BiophysicsJohannes Kepler UniversityLinzAustria
| | - Soon Chul Myung
- Department of Urology, College of MedicineChung‐Ang UniversitySeoulKorea
| | - Young‐Jin Seo
- Department of Life ScienceChung‐Ang UniversitySeoulKorea
| | - Kisung Ko
- BioSystems Design Lab, Department of Medicine, College of MedicineChung‐Ang UniversitySeoulKorea
| |
Collapse
|
7
|
Aslemarz A, Fagotto-Kaufmann M, Ruppel A, Fagotto-Kaufmann C, Balland M, Lasko P, Fagotto F. An EpCAM/Trop2 mechanostat differentially regulates collective behaviour of human carcinoma cells. EMBO J 2025; 44:75-106. [PMID: 39572744 PMCID: PMC11696905 DOI: 10.1038/s44318-024-00309-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 01/04/2025] Open
Abstract
EpCAM and its close relative Trop2 are well-known cell surface markers of carcinoma, but their potential role in cancer metastasis remains unclear. They are known, however, to downregulate myosin-dependent contractility, a key parameter involved in adhesion and migration. We investigate here the morphogenetic impact of the high EpCAM and Trop2 levels typically found in epithelial breast cancer cells, using spheroids of MCF7 cells as an in vitro model. Intriguingly, EpCAM depletion stimulated spheroid cohesive spreading, while Trop2 depletion had the opposite effect. Combining cell biological and biophysical approaches, we demonstrate that while EpCAM and Trop2 both contribute to moderate cell contractility, their depletions differentially impact on the process of "wetting" a substrate, here both matrix and neighboring cells, by affecting the balance of cortical tension at cell and tissue interfaces. These distinct phenotypes can be explained by partial enrichment at specific interfaces. Our data are consistent with the EpCAM-Trop2 pair acting as a mechanostat that tunes adhesive and migratory behaviours.
Collapse
Affiliation(s)
- Azam Aslemarz
- CRBM, University of Montpellier and CNRS, Montpellier, 34293, France
- Dept. of Biology, McGill University, Montreal, QC, H3A1B1, Canada
- SGS, Mississauga, ON, L5T 1W8, Canada
| | - Marie Fagotto-Kaufmann
- CRBM, University of Montpellier and CNRS, Montpellier, 34293, France
- Department of Neurobiology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Artur Ruppel
- LIPHY, UMR5588, University of Grenoble, 38400, Grenoble, France
- CRBM, University of Montpellier and CNRS, Montpellier, 34293, France
| | | | - Martial Balland
- LIPHY, UMR5588, University of Grenoble, 38400, Grenoble, France
| | - Paul Lasko
- Dept. of Biology, McGill University, Montreal, QC, H3A1B1, Canada
| | - François Fagotto
- CRBM, University of Montpellier and CNRS, Montpellier, 34293, France.
| |
Collapse
|
8
|
Yun Y, Kim S, Lee SN, Cho HY, Choi JW. Nanomaterial-based detection of circulating tumor cells and circulating cancer stem cells for cancer immunotherapy. NANO CONVERGENCE 2024; 11:56. [PMID: 39671082 PMCID: PMC11645384 DOI: 10.1186/s40580-024-00466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Nanomaterials have emerged as transformative tools for detecting circulating tumor cells (CTCs) and circulating cancer stem cells (CCSCs), significantly enhancing cancer diagnostics and immunotherapy. Nanomaterials, including those composed of gold, magnetic materials, and silica, have enhanced the sensitivity, specificity, and efficiency of isolating these rare cells from blood. These developments are of paramount importance for the early detection of cancer and for providing real-time insights into metastasis and treatment resistance, which are essential for the development of personalized immunotherapies. The combination of nanomaterial-based platforms with phenotyping techniques, such as Raman spectroscopy and microfluidics, enables researchers to enhance immunotherapy protocols targeting specific CTC and CCSC markers. Nanomaterials also facilitate the targeted delivery of immunotherapeutic agents, including immune checkpoint inhibitors and therapeutic antibodies, directly to tumor cells. This synergistic approach has the potential to enhance therapeutic efficacy and mitigate the risk of metastasis and relapse. In conclusion, this review critically examines the use of nanomaterial-driven detection systems for detecting CTCs and CCSCs, their application in immunotherapy, and suggests future directions, highlighting their potential to transform the integration of diagnostics and treatment, thereby paving the way for more precise and personalized cancer therapies.
Collapse
Affiliation(s)
- Yeochan Yun
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Seewoo Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Sang-Nam Lee
- Uniance Gene Inc., 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea.
| | - Hyeon-Yeol Cho
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea.
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
9
|
Ridnour LA, Heinz WF, Cheng RY, Wink AL, Kedei N, Pore M, Imtiaz F, Femino EL, Gonzalez AL, Coutinho LL, Moffat RL, Butcher D, Edmondson EF, Li X, Rangel MC, Kinders RJ, Rittscher J, Lipkowitz S, Wong ST, Anderson SK, McVicar DW, Glynn SA, Billiar TR, Chang JC, Hewitt SM, Ambs S, Lockett SJ, Wink DA. Tumor NOS2 and COX2 Spatial Juxtaposition with CD8+ T Cells Promote Metastatic and Cancer Stem Cell Niches that Lead to Poor Outcome in ER- Breast Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2766-2782. [PMID: 39356141 PMCID: PMC11497117 DOI: 10.1158/2767-9764.crc-24-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/08/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024]
Abstract
SIGNIFICANCE This work identifies CD8-NOS2+COX2+ and CD8-NOS2-COX2+ unique cellular neighborhoods that drive the tumor immune spatial architecture of CD8+ T cells predictive of clinical outcome and can be targeted with clinically available NOS inhibitors and NSAIDs.
Collapse
Affiliation(s)
- Lisa A. Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - William F. Heinz
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert Y.S. Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Adelaide L. Wink
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Noemi Kedei
- Collaborative Protein Technology Resource (CPTR) Nanoscale Protein Analysis, Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Milind Pore
- Imaging Mass Cytometry, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Fatima Imtiaz
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Elise L. Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Ana L. Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Leandro L. Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Rebecca L. Moffat
- Optical Microscopy and Analysis Laboratory, Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Elijah F. Edmondson
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Maria Cristina Rangel
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Robert J. Kinders
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Jens Rittscher
- Institute of Biomedical Engineering, Big Data Institute, Ludwig Oxford Branch, University of Oxford, Oxford, United Kingdom
| | - Stanley Lipkowitz
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen T.C. Wong
- Houston Methodist Neal Cancer Center, Weill Cornell Medical College, Houston Methodist Hospital, Houston, Texas
| | - Stephen K. Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Daniel W. McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Sharon A. Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jenny C. Chang
- Houston Methodist Neal Cancer Center, Weill Cornell Medical College, Houston Methodist Hospital, Houston, Texas
| | - Stephen M. Hewitt
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen J. Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - David A. Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| |
Collapse
|
10
|
Wang P, Wang B, Chen Y, Lin N, Zheng Z, Chen H, Wang W, He Y. Highly selective detection of breast cancer cells mediated by multi-aptamer and dye-loaded mesoporous silica nanoparticles. Mikrochim Acta 2024; 191:577. [PMID: 39240334 DOI: 10.1007/s00604-024-06664-y] [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: 05/24/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
Multi-aptamer recognition of breast cancer cells (MCF-7) is utilized to achieve high specificity. The method comprises two parts, aptamer-functionalized mesoporous silica nanoparticles (MSNs) loaded with dissimilar dyes (thymolphthalein or curcumin) as signal transducers and aptamer-modified magnetic beads (MBs) as capture agents, which worked together to detect MCF-7 cells sensitively and accurately. The results indicated that the aptasensor has a linear detection range of 100 to 4000 cells and a detection threshold of 10 cells/mL. The method had been successfully employed to detect breast cancer cells in real blood samples to distinguish between breast cancer patients and healthy individuals. In conclusion, the development of the multi-aptamer-based colorimetric sensor offered a novel method for the highly selective detection of MCF-7 cells, contributing to the accurate identification of breast cancer.
Collapse
Affiliation(s)
- Panlin Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Bingbing Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yating Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Nan Lin
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zixin Zheng
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Haoting Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenxiang Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| | - Ye He
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
11
|
Zhang J, Wang Y, Zhang J, Wang X, Liu J, Huo M, Hu T, Ma T, Zhang D, Li Y, Guo C, Yang Y, Zhang M, Yuan B, Qin H, Teng X, Gao T, Hao X, Yu H, Huang W, Xu B, Wang Y. The feedback loop between MTA1 and MTA3/TRIM21 modulates stemness of breast cancer in response to estrogen. Cell Death Dis 2024; 15:597. [PMID: 39154024 PMCID: PMC11330498 DOI: 10.1038/s41419-024-06942-w] [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: 02/27/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024]
Abstract
The metastasis-associated protein (MTA) family plays a crucial role in the development of breast cancer, a common malignancy with a high incidence rate among women. However, the mechanism by which each member of the MTA family contributes to breast cancer progression is poorly understood. In this study, we aimed to investigate the roles of MTA1, MTA3, and tripartite motif-containing 21 (TRIM21) in the proliferation, invasion, epithelial-mesenchymal transition (EMT), and stem cell-like properties of breast cancer cells in vivo and in vitro. The molecular mechanisms of the feedback loop between MTA1 and MTA3/TRIM21 regulated by estrogen were explored using Chromatin immunoprecipitation (ChIP), luciferase reporter, immunoprecipitation (IP), and ubiquitination assays. These findings demonstrated that MTA1 acts as a driver to promote the progression of breast cancer by repressing the transcription of tumor suppressor genes, including TRIM21 and MTA3. Conversely, MTA3 inhibited MTA1 transcription and TRIM21 regulated MTA1 protein stability in breast cancer. Estrogen disrupted the balance between MTA1 and MTA3, as well as between MTA1 and TRIM21, thereby affecting stemness and the EMT processes in breast cancer. These findings suggest that MTA1 plays a vital role in stem cell fate and the hierarchical regulatory network of EMT through negative feedback loops with MTA3 or TRIM21 in response to estrogen, supporting MTA1, MTA3, and TRIM21 as potential prognostic biomarkers and MTA1 as a treatment target for future breast cancer therapies.
Collapse
Affiliation(s)
- Jingyao Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinuo Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miaomiao Huo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Hu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyu Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Die Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang Guo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunkai Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baowen Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Qin
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Teng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Tianyang Gao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xinhui Hao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hefen Yu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yan Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
12
|
Hu T, Han W, Zhou Y, Tu W, Li X, Ni Z. Flow-electricity coupling fields enhance microfluidic platforms for efficient exosome isolation. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5335-5344. [PMID: 39034856 DOI: 10.1039/d4ay00740a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Recently, exosomes have emerged as important biomarkers for cancer diagnosis, playing a significant role in disease diagnosis. Consequently, efficient isolation of exosomes from complex body fluids is now a critical focus in clinical research. We have designed and fabricated an exosome separation chip, leveraging the synergies of flow and electric fields through 3D printing technology. This approach harnesses the combined strengths of both fields, substantially enhancing separation efficiency and purity. This also effectively reduced the voltage required to form an electric field (from 120 V down to 10 V), minimizing the risk of Joule heating, thereby preserving the structural integrity and biological activity of the exosomes. Compared with the standard exosome separation method of ultracentrifugation (UC), our chip offers numerous benefits: it is cost-effective (under 50 RMB), boasts a high recovery rate (64.8%) and high purity (almost 100%), achieves remarkable separation efficiency (within 30 minutes), and is straightforward to operate. Moreover, since an unmarked separation method is used, the separated exosomes can be directly used for downstream detection and analysis, which has certain practicality for future clinical research and application.
Collapse
Affiliation(s)
- Tao Hu
- Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, Jiangsu, 211189, China.
- Southeast Univ, Sch Mech Engn, Nanjing 211189, People's Republic of China
| | - Wenhu Han
- Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, Jiangsu, 211189, China.
- Southeast Univ, Sch Mech Engn, Nanjing 211189, People's Republic of China
| | - Yuxuan Zhou
- Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, Jiangsu, 211189, China.
- Southeast Univ, Sch Mech Engn, Nanjing 211189, People's Republic of China
| | - Weilong Tu
- Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, Jiangsu, 211189, China.
- Southeast Univ, Sch Mech Engn, Nanjing 211189, People's Republic of China
| | - Xiao Li
- Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, Jiangsu, 211189, China.
- Southeast Univ, Sch Mech Engn, Nanjing 211189, People's Republic of China
| | - Zhonghua Ni
- Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, Jiangsu, 211189, China.
- Southeast Univ, Sch Mech Engn, Nanjing 211189, People's Republic of China
| |
Collapse
|
13
|
Guo Y, Qian R, Li Z, Lv T, Yang C, Li W, Pan T, Hou X, Wang Z. Tumor-derived nanovesicles enhance cancer synergistic chemo-immunotherapy by promoting cGAS/STING pathway activation and immunogenetic cell death. Life Sci 2024; 348:122687. [PMID: 38718856 DOI: 10.1016/j.lfs.2024.122687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
AIMS Checkpoint blockade immunotherapy is a promising therapeutic modality that has revolutionized cancer treatment; however, the therapy is only effective on a fraction of patients due to the tumor environment. In tumor immunotherapy, the cGAS-STING pathway is a crucial intracellular immune response pathway. Therefore, this study aimed to develop an immunotherapy strategy based on the cGAS-STING pathway. MATERIALS AND METHODS The physicochemical properties of the nanoparticles EM@REV@DOX were characterized by TEM, DLS, and WB. Subcutaneous LLC xenograft tumors were used to determine the biodistribution, antitumor efficacy, and immune response. Blood samples and tissues of interest were harvested for hematological analysis and H&E staining. SIGNIFICANCE Overall, our designed nanovesicles provide a new perspective on tumor immunotherapy by ICD and cGAS-STING pathway, promoting DCs maturation, macrophage polarization, and activating T cells, offering a meaningful strategy for accelerating the clinical development of immunotherapy. KEY FINDINGS EM@REV@DOX accumulated in the tumor site through EPR and homing targeting effect to release REV and DOX, resulting in DNA damage and finally activating the cGAS-STING pathway, thereby promoting DCs maturation, macrophage polarization, and activating T cells. Additionally, EM@REV@DOX increased the production of pro-inflammatory cytokines (e.g., TNF-α and IFN-β). As a result, EM@REV@DOX was effective in treating tumor-bearing mice and prolonged their lifespans. When combined with αPD-L1, EM@REV@DOX significantly inhibited distant tumor growth, extended the survival of mice, and prevented long-term postoperative tumor metastasis, exhibiting great potential in antitumor immunotherapy.
Collapse
Affiliation(s)
- Yawen Guo
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Ruijie Qian
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, PR China
| | - Zijie Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Tingting Lv
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Chunwang Yang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Wen Li
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Teng Pan
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xiaoming Hou
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Zhiyu Wang
- Department of Immuno-Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China.
| |
Collapse
|
14
|
Qian R, Guo Y, Wang R, Wang S, Gao X, Zhu Z, Wang K, Zhu K, Jia B, Chen Y, Wang Z, Ren J, Duan X, Han X. Cell Membrane Hybrid Lipid Nanovesicles Enhance Innate Immunity for Synergistic Immunotherapy by Promoting Immunogenic Cell Death and cGAS Activation. Biomater Res 2024; 28:0038. [PMID: 38868091 PMCID: PMC11168305 DOI: 10.34133/bmr.0038] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/08/2024] [Indexed: 06/14/2024] Open
Abstract
Immunotherapy shows great therapeutic potential for long-term protection against tumor relapse and metastasis. Innate immune sensors, such as cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING), dissolve DNA and induce type I interferon. Through activation of the cGAS/STING pathway, chemotherapy drugs and reversine (REV) may provide synergetic anti-tumor effects. Here, we prepared drug-loaded cell membrane hybrid lipid nanovesicles (LEVs) (designated LEV@DOX@REV) by fusion of cell membranes, phospholipids, doxorubicin (DOX), and REV, to realize accurate delivery to tumors and chemo-immunotherapy. The cell membranes of LEVs confer "homing" abilities. DOX can induce immunogenic cell death as a result of its specific immunomodulatory effects, which promotes the maturation of immune cells and improves the microenvironment of the immune system. REV is proven to efficiently activate cGAS/STING signaling, thereby enhancing the immune system. The antitumor efficacy of LEV@DOX@REV was evaluated in a 4T1 subcutaneous tumor xenograft model, a distant metastatic tumor model, and a liver metastatic tumor model. LEV@DOX@REV facilitated the infiltration of cytotoxic T lymphocytes within tumors, increased the secretion of proinflammatory cytokines, and modified the tumor microenvironment. In conclusion, LEV@DOX@REV displayed favorable antitumor effects and extended the survival of tumor-bearing mice. We therefore successfully developed nanoparticles capable of enhancing immune activation that have potential therapeutic applications for cancer immunotherapy.
Collapse
Affiliation(s)
- Ruijie Qian
- Department of Interventional Radiology,
The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yawen Guo
- Department of Immuno-Oncology,
The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ruihua Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, College of Medicine, Henan Medical Key Laboratory of Molecular Imaging,
Zhengzhou University, Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Shuai Wang
- Department of Medical Technology,
Nanyang Medical College, Nanyang 473000, Henan, China
| | - Xuemei Gao
- Department of Nuclear Medicine, The First Affiliated Hospital, College of Medicine, Henan Medical Key Laboratory of Molecular Imaging,
Zhengzhou University, Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Sichuan Provincial People’s Hospital, Chengdu, Sichuan 610072, China
| | - Kun Wang
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine,
Tongji University, Shanghai 200120, China
| | - Ke Zhu
- Department of Cardiology, Shanghai East Hospital, School of Medicine,
Tongji University, Shanghai 200120, China
| | - Baosong Jia
- Department of Breast and Thyroid Surgery,
The Second People’s Hospital of Lianyungang, Lianyungang, China
| | - Yijian Chen
- Department of Radiology,
Beijing Jingmei Group General Hospital, Beijing, China
| | - Zhiyu Wang
- Department of Immuno-Oncology,
The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianzhuang Ren
- Department of Interventional Radiology,
The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuhua Duan
- Department of Interventional Radiology,
The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinwei Han
- Department of Interventional Radiology,
The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Xiao D, Xiong M, Wang X, Lyu M, Sun H, Cui Y, Chen C, Jiang Z, Sun F. Regulation of the Function and Expression of EpCAM. Biomedicines 2024; 12:1129. [PMID: 38791091 PMCID: PMC11117676 DOI: 10.3390/biomedicines12051129] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is a single transmembrane protein on the cell surface. Given its strong expression on epithelial cells and epithelial cell-derived tumors, EpCAM has been identified as a biomarker for circulating tumor cells (CTCs) and exosomes and a target for cancer therapy. As a cell adhesion molecule, EpCAM has a crystal structure that indicates that it forms a cis-dimer first and then probably a trans-tetramer to mediate intercellular adhesion. Through regulated intramembrane proteolysis (RIP), EpCAM and its proteolytic fragments are also able to regulate multiple signaling pathways, Wnt signaling in particular. Although great progress has been made, increasingly more findings have revealed the context-specific expression and function patterns of EpCAM and their regulation processes, which necessitates further studies to determine the structure, function, and expression of EpCAM under both physiological and pathological conditions, broadening its application in basic and translational cancer research.
Collapse
Affiliation(s)
- Di Xiao
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Mingrui Xiong
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xin Wang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Mengqing Lyu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Hanxiang Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yeting Cui
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Chen Chen
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Ziyu Jiang
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Fan Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
16
|
Zhang Y, Yang Q, Zhu L, Lu X, Xin W, Ding J, Wang S, Tang Z, Fan GC, Cen Y, Song ZL, Luo X. Intelligent Cell Profiling and Precision Release: Multimolecular Marker-Activated Transmembrane DNA Computing Nanosystem. Anal Chem 2024; 96:7747-7755. [PMID: 38691774 DOI: 10.1021/acs.analchem.4c01122] [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: 05/03/2024]
Abstract
Accurate classification of tumor cells is of importance for cancer diagnosis and further therapy. In this study, we develop multimolecular marker-activated transmembrane DNA computing systems (MTD). Employing the cell membrane as a native gate, the MTD system enables direct signal output following simple spatial events of "transmembrane" and "in-cell target encounter", bypassing the need of multistep signal conversion. The MTD system comprises two intelligent nanorobots capable of independently sensing three molecular markers (MUC1, EpCAM, and miR-21), resulting in comprehensive analysis. Our AND-AND logic-gated system (MTDAND-AND) demonstrates exceptional specificity, allowing targeted release of drug-DNA specifically in MCF-7 cells. Furthermore, the transformed OR-AND logic-gated system (MTDOR-AND) exhibits broader adaptability, facilitating the release of drug-DNA in three positive cancer cell lines (MCF-7, HeLa, and HepG2). Importantly, MTDAND-AND and MTDOR-AND, while possessing distinct personalized therapeutic potential, share the ability of outputting three imaging signals without any intermediate conversion steps. This feature ensures precise classification cross diverse cells (MCF-7, HeLa, HepG2, and MCF-10A), even in mixed populations. This study provides a straightforward yet effective solution to augment the versatility and precision of DNA computing systems, advancing their potential applications in biomedical diagnostic and therapeutic research.
Collapse
Affiliation(s)
- Yuxi Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qian Yang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Lina Zhu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xinyi Lu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Wenjuan Xin
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jiani Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shumin Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Zijie Tang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Gao-Chao Fan
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yao Cen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhi-Ling Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
17
|
Zhang M, Wang Y, Song Z, Lu Y, Zhao H, Wang Y, Lu P, Liu Y. Recent Progress of Bioinspired Cell Membrane in Cancer Immunotherapy. Clin Med Insights Oncol 2024; 18:11795549241236896. [PMID: 38645894 PMCID: PMC11032066 DOI: 10.1177/11795549241236896] [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: 11/26/2023] [Accepted: 01/20/2024] [Indexed: 04/23/2024] Open
Abstract
By modifying immune cells, immunotherapy can activate immune response to establish long-term immune memory and prevent tumor recurrence. However, their effectiveness is largely constricted by the poor immunogenicity, immune escape, and immune tolerance of the tumor. This is related to the characteristics of the tumor itself, such as genome instability and mutation. The combination of various nanocarriers with tumor immunotherapy is beneficial for overcoming the shortcomings of traditional immunotherapy. Nanocarriers coated by cell membranes can extend blood circulation time, improve ability to evade immune clearance, and enhance targeting, thus significantly enhancing the efficacy of immunotherapy and showing great potential in tumor immunotherapy. This article reviews the application research progress of different types of cell membrane-modified nanocarriers in tumor immunotherapy, immunotherapy combination therapy, and tumor vaccines, and provides prospects for future research.
Collapse
Affiliation(s)
- Min Zhang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yuanhang Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Zhiyuan Song
- Department of Ultrasound Medicine, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Yimeng Lu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Houyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yihan Wang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Ping Lu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| |
Collapse
|
18
|
Giunashvili N, Thomas JM, Schvarcz CA, Viana PHL, Aloss K, Bokhari SMZ, Koós Z, Bócsi D, Major E, Balogh A, Benyó Z, Hamar P. Enhancing therapeutic efficacy in triple-negative breast cancer and melanoma: synergistic effects of modulated electro-hyperthermia (mEHT) with NSAIDs especially COX-2 inhibition in in vivo models. Mol Oncol 2024; 18:1012-1030. [PMID: 38217262 PMCID: PMC10994232 DOI: 10.1002/1878-0261.13585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a leading cause of cancer mortality and lacks modern therapy options. Modulated electro-hyperthermia (mEHT) is an adjuvant therapy with demonstrated clinical efficacy for the treatment of various cancer types. In this study, we report that mEHT monotherapy stimulated interleukin-1 beta (IL-1β) and interleukin-6 (IL-6) expression, and consequently cyclooxygenase 2 (COX-2), which may favor a cancer-promoting tumor microenvironment. Thus, we combined mEHT with nonsteroid anti-inflammatory drugs (NSAIDs): a nonselective aspirin, or the selective COX-2 inhibitor SC236, in vivo. We demonstrate that NSAIDs synergistically increased the effect of mEHT in the 4T1 TNBC model. Moreover, the strongest tumor destruction ratio was observed in the combination SC236 + mEHT groups. Tumor damage was accompanied by a significant increase in cleaved caspase-3, suggesting that apoptosis played an important role. IL-1β and COX-2 expression were significantly reduced by the combination therapies. In addition, a custom-made nanostring panel demonstrated significant upregulation of genes participating in the formation of the extracellular matrix. Similarly, in the B16F10 melanoma model, mEHT and aspirin synergistically reduced the number of melanoma nodules in the lungs. In conclusion, mEHT combined with a selective COX-2 inhibitor may offer a new therapeutic option in TNBC.
Collapse
Grants
- STIA-OTKA-2022 Semmelweis Science and Innovation Fund
- OTKA_ANN 110810 National Research, Development, and Innovation Office
- OTKA_SNN 114619 National Research, Development, and Innovation Office
- ÚNKP-23-3-II-SE-45 National Research, Development, and Innovation Office
- ÚNKP-23-4-I-SE-22 National Research, Development, and Innovation Office
- OTKA_K 145998 National Research, Development, and Innovation Office
- Tempus Foundation
- EFOP-3.6.3-VEKOP-16-2017-00009 Semmelweis Excellence 250+ Scholarship
Collapse
Affiliation(s)
- Nino Giunashvili
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | | | - Csaba András Schvarcz
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
- HUN‐REN‐SU Cerebrovascular and Neurocognitive Diseases Research GroupBudapestHungary
| | | | - Kenan Aloss
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | | | - Zoltán Koós
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | - Dániel Bócsi
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | - Enikő Major
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
- HUN‐REN‐SU Cerebrovascular and Neurocognitive Diseases Research GroupBudapestHungary
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
- HUN‐REN‐SU Cerebrovascular and Neurocognitive Diseases Research GroupBudapestHungary
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| |
Collapse
|
19
|
Sun L, Li C, Zhao N, Wang B, Li H, Wang H, Zhang X, Zhao X. Host protein EPCAM interacting with EtMIC8-EGF is essential for attachment and invasion of Eimeria tenella in chickens. Microb Pathog 2024; 188:106549. [PMID: 38281605 DOI: 10.1016/j.micpath.2024.106549] [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: 10/10/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/30/2024]
Abstract
The five epidermal growth factor-like domains (EGF) of Eimeria tenella microneme protein 8 (EtMIC8) (EtMIC8-EGF) plays a vital role in host cell attachment and invasion. These processes require interactions between parasite proteins and receptors on the surface of host cells. In this study, five chicken membrane proteins potentially interacting with EtMIC8-EGF were identified using the GST pull-down assay and mass spectrometry analysis, and only chicken (Gallus gallus) epithelial cell adhesion molecule (EPCAM) could bind to EtMIC8-EGF. EPCAM-specific antibody and recombinant EPCAM protein (rEPCAM) inhibited the EtMIC8-EGF binding to host cells in a concentration-dependent manner. Furthermore, the rEPCAM protein showed a binding activity to sporozoites in vitro, and a significant reduction of E. tenella invasion in DF-1 cells was further observed after pre-incubation of sporozoites with rEPCAM. The specific anti-EPCAM antibody further significantly decreased weight loss, lesion score and oocyst output during E. tenella infection, displaying partial inhibition of E. tenella infection. These results indicate that chicken EPCAM is an important EtMIC8-interacting host protein involved in E. tenella-host cell adhesion and invasion. The findings will contribute to a better understanding of the role of adhesion-associated microneme proteins in E. tenella.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
| | - Chao Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding City, 071000, Hebei Province, China
| | - Ningning Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
| | - Bingxiang Wang
- Shandong Vocational Animal Science and Veterinary College, Weifang City, Shandong Province, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
| | - Hairong Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, China.
| |
Collapse
|
20
|
Cheng S, Zhang C, Hu X, Zhu Y, Shi H, Tan W, Luo X, Xian Y. Ultrasensitive determination of surface proteins on tumor-derived small extracellular vesicles for breast cancer identification based on lanthanide-activated signal amplification strategy. Talanta 2024; 267:125189. [PMID: 37714039 DOI: 10.1016/j.talanta.2023.125189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Small extracellular vesicles (sEVs) carrying multiple tumor-associated proteins inherited from parental cells play crucial roles in noninvasive breast cancer (BC) diagnosis. However, it is challenging to assess the subtle variations of surface proteins on sEV membranes due to the highly heterogeneous BC. Therefore, a simple and ultrasensitive assay based on lanthanide (Ln3+)-activated luminescence signal amplification was developed to detect multiple surface proteins on BC-derived sEVs. Multiple protein biomarkers on sEVs can be well identified with high sensitivity and specificity through dissolution-amplified luminescence of the NaEuF4 nanoparticle-based nanoprobe. We employ linear discriminant analysis to successfully discriminate triple negative BC cell (MDA-MB-231 cell) derived sEVs from other breast cell lines (MCF-7, SK-BR-3, BT474 and MCF-10A cell). Furthermore, the strategy enables high accuracy for districting the progression stages of BC patients and healthy donors. The simple and sensitive signal amplification strategy exhibits great potential for early clinic diagnosis by precise protein profiling of sEVs.
Collapse
Affiliation(s)
- Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| | - Xinyu Hu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yingxin Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenqiao Tan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Xianzhu Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
21
|
Hachey SJ, Hatch CJ, Gaebler D, Mocherla A, Nee K, Kessenbrock K, Hughes CCW. Targeting tumor-stromal interactions in triple-negative breast cancer using a human vascularized micro-tumor model. Breast Cancer Res 2024; 26:5. [PMID: 38183074 PMCID: PMC10768273 DOI: 10.1186/s13058-023-01760-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive with limited available treatments. Stromal cells in the tumor microenvironment (TME) are crucial in TNBC progression; however, understanding the molecular basis of stromal cell activation and tumor-stromal crosstalk in TNBC is limited. To investigate therapeutic targets in the TNBC stromal niche, we used an advanced human in vitro microphysiological system called the vascularized micro-tumor (VMT). Using single-cell RNA sequencing, we revealed that normal breast tissue stromal cells activate neoplastic signaling pathways in the TNBC TME. By comparing interactions in VMTs with clinical data, we identified therapeutic targets at the tumor-stromal interface with potential clinical significance. Combining treatments targeting Tie2 signaling with paclitaxel resulted in vessel normalization and increased efficacy of paclitaxel in the TNBC VMT. Dual inhibition of HER3 and Akt also showed efficacy against TNBC. These data demonstrate the potential of inducing a favorable TME as a targeted therapeutic approach in TNBC.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
| | | | - Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Aneela Mocherla
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Kevin Nee
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Christopher C W Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
22
|
Hasan A, Khan NA, Uddin S, Khan AQ, Steinhoff M. Deregulated transcription factors in the emerging cancer hallmarks. Semin Cancer Biol 2024; 98:31-50. [PMID: 38123029 DOI: 10.1016/j.semcancer.2023.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/25/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Cancer progression is a multifaceted process that entails several stages and demands the persistent expression or activation of transcription factors (TFs) to facilitate growth and survival. TFs are a cluster of proteins with DNA-binding domains that attach to promoter or enhancer DNA strands to start the transcription of genes by collaborating with RNA polymerase and other supporting proteins. They are generally acknowledged as the major regulatory molecules that coordinate biological homeostasis and the appropriate functioning of cellular components, subsequently contributing to human physiology. TFs proteins are crucial for controlling transcription during the embryonic stage and development, and the stability of different cell types depends on how they function in different cell types. The development and progression of cancer cells and tumors might be triggered by any anomaly in transcription factor function. It has long been acknowledged that cancer development is accompanied by the dysregulated activity of TF alterations which might result in faulty gene expression. Recent studies have suggested that dysregulated transcription factors play a major role in developing various human malignancies by altering and rewiring metabolic processes, modifying the immune response, and triggering oncogenic signaling cascades. This review emphasizes the interplay between TFs involved in metabolic and epigenetic reprogramming, evading immune attacks, cellular senescence, and the maintenance of cancer stemness in cancerous cells. The insights presented herein will facilitate the development of innovative therapeutic modalities to tackle the dysregulated transcription factors underlying cancer.
Collapse
Affiliation(s)
- Adria Hasan
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Lucknow 226026, India; Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow 226026, India
| | - Naushad Ahmad Khan
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad General Hospital, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Biosciences, Integral University, Lucknow 226026, India; Animal Research Center, Qatar University, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar.
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Animal Research Center, Qatar University, Doha, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; College of Medicine, Qatar University, Doha 2713, Qatar
| |
Collapse
|
23
|
Yue Y, Ouyang H, Ma M, Yang Y, Zhang H, He A, Liu R. Nucleic acid aptasensor with magnetically induced self-assembly for the detection of EpCAM glycoprotein. Mikrochim Acta 2023; 191:64. [PMID: 38157059 DOI: 10.1007/s00604-023-06117-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024]
Abstract
A "turn-on" aptasensor for label-free and cell-free EpCAM detection was constructed by employing magnetic α-Fe2O3/Fe3O4@Au nanocomposites as a matrix for signal amplification and double-stranded complex (SH-DNA/Apt probes) immobilization through Au-S binding. α-Fe2O3/Fe3O4@Au could be efficiently assembled into uniform and stable self-assembly films via magnetic-induced self-assembly technique on a magnetic glassy carbon electrode (MGCE). The effectiveness of the platform for EpCAM detection was confirmed through differential pulse voltammetry (DPV). Under optimized conditions, the platform exhibited excellent specificity for EpCAM, and a strong linear correlation was observed between the current and the logarithm of EpCAM protein concentration in the range 1 pg/mL-1000 pg/mL (R2 = 0.9964), with a limit of detection (LOD) of 0.27 pg/mL. Furthermore, the developed platform demonstrated good stability during a 14-day storage test, with fluctuations remaining below 93.33% of the initial current value. Promising results were obtained when detecting EpCAM in spiked serum samples, suggesting its potential as a point-of-care (POC) testing.
Collapse
Affiliation(s)
- Yao Yue
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Hezhong Ouyang
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212300, People's Republic of China
| | - Mingyi Ma
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Yaping Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Haoda Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Aolin He
- Affiliated Kunshan Hospital, Jiangsu University, Suzhou, 215300, People's Republic of China.
| | - Ruijiang Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| |
Collapse
|
24
|
Lin Y, Guan X, Su J, Chen S, Fu X, Xu X, Deng X, Chang J, Qin A, Shen A, Zhang L. Cell Membrane-Camouflaged Nanoparticles Mediated Nucleic Acids Delivery. Int J Nanomedicine 2023; 18:8001-8021. [PMID: 38164266 PMCID: PMC10758188 DOI: 10.2147/ijn.s433737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024] Open
Abstract
Nucleic acids have emerged as promising therapeutic agents for many diseases because of their potential in modulating gene expression. However, the delivery of nucleic acids remains a significant challenge in gene therapy. Although viral vectors have shown high transfection efficiency, concerns regarding teratogenicity or carcinogenicity have been raised. Non-viral vehicles, including cationic polymers, liposomes, and inorganic materials possess advantages in terms of safety, ease of preparation, and low cost. Nevertheless, they also face limitations related to immunogenicity, quick clearance in vivo, and lack of targeting specificity. On the other hand, bioinspired strategies have shown increasing potential in the field of drug delivery, yet there is a lack of comprehensive reviews summarizing the rapid development of bioinspired nanoparticles based on the cell membrane camouflage to construct the nucleic acids vehicles. Herein, we enumerated the current difficulties in nucleic acid delivery with various non-viral vehicles and provided an overview of bioinspired strategies for nucleic acid delivery.
Collapse
Affiliation(s)
- Yinshan Lin
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xiaoling Guan
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Jianfen Su
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Sheng Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xihua Fu
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
| | - Xiaowei Xu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Xiaohua Deng
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Jishuo Chang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Aiping Qin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Ao Shen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| | - Lingmin Zhang
- Pharmacy Department & Panyu Institute of Infectious Diseases, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, 511400, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, People’s Republic of China
| |
Collapse
|
25
|
Ridnour LA, Heinz WF, Cheng RY, Wink AL, Kedei N, Pore M, Imtiaz F, Femino EL, Gonzalez AL, Coutinho L, Butcher D, Edmondson EF, Rangel MC, Kinders RJ, Lipkowitz S, Wong ST, Anderson SK, McVicar DW, Li X, Glynn SA, Billiar TR, Chang JC, Hewitt SM, Ambs S, Lockett SJ, Wink DA. NOS2 and COX2 Provide Key Spatial Targets that Determine Outcome in ER- Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572859. [PMID: 38187532 PMCID: PMC10769386 DOI: 10.1101/2023.12.21.572859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Estrogen receptor-negative (ER-) breast cancer is an aggressive breast cancer subtype with limited therapeutic options. Upregulated expression of both inducible nitric oxide synthase (NOS2) and cyclo-oxygenase (COX2) in breast tumors predicts poor clinical outcomes. Signaling molecules released by these enzymes activate oncogenic pathways, driving cancer stemness, metastasis, and immune suppression. The influence of tumor NOS2/COX2 expression on the landscape of immune markers using multiplex fluorescence imaging of 21 ER- breast tumors were stratified for survival. A powerful relationship between tumor NOS2/COX2 expression and distinct CD8+ T cell phenotypes was observed at 5 years post-diagnosis. These results were confirmed in a validation cohort using gene expression data showing that ratios of NOS2 to CD8 and COX2 to CD8 are strongly associated with poor outcomes in high NOS2/COX2-expressing tumors. Importantly, multiplex imaging identified distinct CD8+ T cell phenotypes relative to tumor NOS2/COX2 expression in Deceased vs Alive patient tumors at 5-year survival. CD8+NOS2-COX2- phenotypes defined fully inflamed tumors with significantly elevated CD8+ T cell infiltration in Alive tumors expressing low NOS2/COX2. In contrast, two distinct phenotypes including inflamed CD8+NOS2+COX2+ regions with stroma-restricted CD8+ T cells and CD8-NOS2-COX2+ immune desert regions with abated CD8+ T cell penetration, were significantly elevated in Deceased tumors with high NOS2/COX2 expression. These results were supported by applying an unsupervised nonlinear dimensionality-reduction technique, UMAP, correlating specific spatial CD8/NOS2/COX2 expression patterns with patient survival. Moreover, spatial analysis of the CD44v6 and EpCAM cancer stem cell (CSC) markers within the CD8/NOS2/COX2 expression landscape revealed positive correlations between EpCAM and inflamed stroma-restricted CD8+NOS2+COX2+ phenotypes at the tumor/stroma interface in deceased patients. Also, positive correlations between CD44v6 and COX2 were identified in immune desert regions in deceased patients. Furthermore, migrating tumor cells were shown to occur only in the CD8-NOS2+COX2+ regions, identifying a metastatic hot spot. Taken together, this study shows the strength of spatial localization analyses of the CD8/NOS2/COX2 landscape, how it shapes the tumor immune microenvironment and the selection of aggressive tumor phenotypes in distinct regions that lead to poor clinical outcomes. This technique could be beneficial for describing tumor niches with increased aggressiveness that may respond to clinically available NOS2/COX2 inhibitors or immune-modulatory agents.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - William F Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Robert Ys Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Noemi Kedei
- Collaborative Protein Technology Resource (CPTR) Nanoscale Protein Analysis, OSTR, CCR, NCI, NIH
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
| | - Fatima Imtiaz
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Elise L Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Ana L Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Leandro Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - M Cristina Rangel
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Robert J Kinders
- Office of the Director, Division of Cancer Treatment and Diagnosis, NCI, Frederick, MD
| | | | | | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Danial W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Jenny C Chang
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | | | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| |
Collapse
|
26
|
Shi L, Zhang D, Han H, Zhang L, Li S, Yang F, He C. HOTAIR knockdown impairs metastasis of cervical cancer cells by down-regulating metastasis-related genes. J OBSTET GYNAECOL 2023; 43:2181060. [PMID: 36972141 DOI: 10.1080/01443615.2023.2181060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
This study investigated the role of LncRNA HOTAIR knockdown in the biological impacts on cervical cancer cells. The HOTAIR gene in two human cervical cancer cell lines was silenced with small interfering (si) RNA siHOTAIR. Proliferation, apoptosis, migration and invasion of cells were assessed following the knockdown. The expressions of Notch1, EpCAM, E-cadherin, vimentin and STAT3 were assessed using qRT-PCR and Western blotting analysis. Compared with controls, HOTAIR levels were reduced significantly, the OD values of cells were significantly decreased in proliferation assays, cell apoptosis was significantly increased, cell migration and invasion were significantly reduced after HOTAIR knockdown. Molecular analysis showed that Notch1, EpCAM, vimentin and STAT3 expressions were decreased significantly, while the expression of E-cadherin was significantly increased after HOTAIR knockdown. Rescue experiments further confirmed that Notch1 and STAT3 were involved in siHOTAIR-mediated reduction of migration and invasion of cervical cancer cells.IMPACT STATEMENTWhat is already known on this subject? Long non-coding RNAs including HOTAIR, is implicated in occurrence and development of cancer and have been explored to develop new therapeutic options for cancer.What do the results of this study add? HOTAIR silencing significantly reduces the viability and migration ability of cells and induces cell apoptosis, adding experimental data supporting the potential use of HOTAIR specific-siRNA as a therapeutic avenue for the cancer.What are the implications of these findings for clinical practice and/or further research? The finding from this study would help develop clinically applicable therapeutic avenues for the cancer and identify new treatment targets in the relevant pathways leading to new drugs or treatments.
Collapse
Affiliation(s)
- Lei Shi
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Dehui Zhang
- Department of Oncology, Daqing Oilfield General Hospital and Huiren Cancer Hospital, Daqing, P.R. China
| | - Huijuan Han
- Department of Obstetrics and Gynecology, the 962 Hospital, Joint Logistics Support of the Chinese People's Liberation Army, Harbin, P.R. China
| | - Liangyu Zhang
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Sirui Li
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Fang Yang
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, P.R. China
| | - Caijun He
- Department of Obstetrics and Gynecology, the 962 Hospital, Joint Logistics Support of the Chinese People's Liberation Army, Harbin, P.R. China
| |
Collapse
|
27
|
Jiang W, Lei Y, Peng C, Wu D, Wu J, Xu Y, Xia X. Recent advances in cancer cell bionic nanoparticles for tumour therapy. J Drug Target 2023; 31:1065-1080. [PMID: 37962304 DOI: 10.1080/1061186x.2023.2283838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
Nanoparticle-based drug delivery systems have found extensive use in delivering oncology therapeutics; however, some delivery vehicles still exhibit rapid immune clearance, lack of biocompatibility and insufficient targeting. In recent years, bionanoparticles constructed from tumour cell membranes have gained momentum as tumour-targeting therapeutic agents. Cancer cell membrane-coated nanoparticles (CCMCNPs) typically consist of a drug-loaded nanoparticle core coated with cancer cell membrane. CCMCNPs retain homologous tumour cell surface antigens, receptors and proteins, and it has been shown that the modified nanoparticles exhibit better homologous targeting, immune escape and biocompatibility. CCMCNPs are now widely used in a variety of cancer treatments, including photothermal, photodynamic and sonodynamic therapies, chemotherapy, immunotherapy, chemodynamical therapy or other combination therapies. This article presents different therapeutic approaches using multimodal antitumour therapy-combination of two or more therapies that treat tumours synergistically-based on tumour cell membrane systems. The advantages of CCMCNPs in different cancer treatments in recent years are summarised, thus, providing new strategies for cancer treatment research.
Collapse
Affiliation(s)
- Wanting Jiang
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yujing Lei
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Cheng Peng
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Donghai Wu
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Wu
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yiling Xu
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Xinhua Xia
- Laboratory of Key Technologies of Targeted and Compound Preparations of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
28
|
Guo Q, Pan K, Qiu P, Liu Z, Chen J, Lin J. Identification of an exosome-related signature associated with prognosis and immune infiltration in breast cancer. Sci Rep 2023; 13:18198. [PMID: 37875600 PMCID: PMC10598067 DOI: 10.1038/s41598-023-45325-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/18/2023] [Indexed: 10/26/2023] Open
Abstract
Exosomes, nanosized vesicles, play a vital role in breast cancer (BC) occurrence, development, and drug resistance. Hence, we proceeded to study the potential prognostic value of exosome-related genes and their relationship to the immune microenvironment in BC. 121 exosome-related genes were provided by the ExoBCD database, and 7 final genes were selected to construct the prognostic signature. Besides, the expression levels of the 7 exosome-related genes were validated by the experiment in BC cell lines. Based on the signature, BC patients from the training and validation cohorts were separated into low- and high-risk groups. Subsequently, the R clusterProfiler package was applied to identify the distinct enrichment pathways between high-risk groups and low-risk groups. The relevance of the tumor immune microenvironment and exosome-related gene risk score were analyzed in BC. Eventually, the different expression levels of immune checkpoint-related genes were compared between the two risk groups. Based on the risk model, the low-risk groups were identified with a higher survival rate both in the training and validation cohorts. A better overall survival was revealed in patients with higher scores evaluated by the estimation of stromal and immune cells in malignant tumor tissues using expression (ESTIMATE) algorithm. Subsequently, BC patients with lower risk scores were indicated by higher expression levels of some immune checkpoint-related genes and immune cell infiltration. Exosomes are closely associated with the prognosis and immune cell infiltration of BC. These findings may contribute to improving immunotherapy and provide a new vision for BC treatment strategies.
Collapse
Affiliation(s)
- Qiaonan Guo
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Kelun Pan
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Pengjun Qiu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zundong Liu
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jianpeng Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jianqing Lin
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
29
|
Ma Z, Xu H, Ye BC. Recent progress in quantitative technologies for the analysis of cancer-related exosome proteins. Analyst 2023; 148:4954-4966. [PMID: 37721099 DOI: 10.1039/d3an01228j] [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: 09/19/2023]
Abstract
Exosomes are a kind of extracellular vesicles, which play a significant role in intercellular communication and molecular exchange. Cancer-derived exosomes are potential and ideal biomarkers for the early diagnosis and treatment monitoring of cancers because of their abundant biological information and contribution to the interaction between cancer cells and the tumor microenvironment. However, there are a number of drawbacks, such as low sensitivity and tedious steps, in conventional detection techniques. Furthermore, exosome quantification is not enough to accurately distinguish cancer patients from healthy individuals. Therefore, developing efficient, accurate, and inexpensive exosome surface protein analysis techniques is necessary and critical. In recent years, a considerable number of researchers have presented novel detection strategies in this field. This review summarizes the recent progress in quantitative technologies for the analysis of cancer-related exosome proteins, mainly including the detection methods based on aptamers, nanomaterials, and antibodies, discusses a roadmap for future developments, and aims to offer an innovative perspective of exosome research.
Collapse
Affiliation(s)
- Zhongwen Ma
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Huiying Xu
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| | - Bang-Ce Ye
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
30
|
Maurya SB, Shetty DC, Rathore AS, Juneja S, Jain A, Banga A. Immunolocalization of epithelial cell adhesion molecule and matrix metalloproteinase-9 in oral epithelial dysplasia and oral squamous cell carcinoma. J Cancer Res Ther 2023; 19:1775-1780. [PMID: 38376277 DOI: 10.4103/jcrt.jcrt_1012_21] [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: 06/24/2021] [Accepted: 12/20/2021] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Cancers are complex tissues composed of multiple distinct cell types that participate in heterotypic interactions with one another. Physiologically cell-to-cell contacts formed by dense populations of normal cells operate to suppress further cell proliferation. OBJECTIVES The objective of the study is to evaluate and compare the immunoexpression of matrix metalloproteinase-9 (MMP-9) and epithelial cell adhesion molecule (EpCAM) in oral epithelial dysplasia (OED) and oral squamous cell carcinoma (OSCC) and to hypothesize their role in the progression in varying grades of these lesions. MATERIALS AND METHODS A total of 60 samples comprising of 30 cases each of OED and OSCC. Three micrometers thin sections were taken and subjected for hematoxylin and eosin stain and immunohistochemical procedure. The sections were incubated with monoclonal anti-EpCAM anti-MMP-9 antibody. The data were analyzed using SPSS software version 19. RESULTS The results of the study show EpCAM immunoexpression decreased in OSCC when compared to OED. MMP-9 immunoexpression increased in OSCC when compared to OED (statistically significant, P ≤ 0.05). CONCLUSION Correlation between EpCAM and MMP-9 may help to unravel the signaling cascades involved in the carcinomatous changes, tumor cell invasion, and progression of OSCCs.
Collapse
Affiliation(s)
- Shashi Bhal Maurya
- Department of Oral Pathology and Microbiology, ITS Dental College, Greater Noida, Uttar Pradesh, India
| | - Devi Charan Shetty
- Department of Oral Pathology and Microbiology, ITS Dental College, Ghaziabad, Uttar Pradesh, India
| | - Ajit Singh Rathore
- Department of Oral Pathology and Microbiology, ITS Dental College, Ghaziabad, Uttar Pradesh, India
| | - Saurabh Juneja
- Department of Oral Pathology and Microbiology, ITS Dental College, Ghaziabad, Uttar Pradesh, India
| | - Anshi Jain
- Department of Oral Pathology and Microbiology, ITS Dental College, Ghaziabad, Uttar Pradesh, India
| | - Akanksha Banga
- Department of Oral Pathology and Microbiology, ITS Dental College, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
31
|
Kalaitsidou I, Pasteli N, Venetis G, Poulopoulos A, Antoniades K. Immunohistochemical Expression of Epithelial Cell Adhesion Molecule (EpCAM) in Salivary Gland Cancer: Correlation with the Biological Behavior. Diagnostics (Basel) 2023; 13:2652. [PMID: 37627911 PMCID: PMC10453306 DOI: 10.3390/diagnostics13162652] [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: 07/19/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Salivary gland neoplasms comprise a diverse group of tumors with different biological behaviors and clinical outcomes. Understanding the underlying molecular alterations associated with these malignancies is critical for accurate diagnosis, prognosis, and treatment strategies. Among the many biomarkers under investigation, epithelial cell adhesion molecule (EpCAM) has emerged as a promising candidate in salivary gland cancer research. This article aims to provide a comprehensive overview of the differential expression of EpCAM in salivary gland cancer and its potential correlation with the biological behavior of these tumors. The clinical characteristics of 65 patients with salivary gland malignancy of different histopathological subtypes were included. We report the differential expression of EpCAM and the relationship between the clinical and histopathologic features of these tumors. Regarding the evaluation of the effect of EpCAM expression on survival, in our study, we showed that tumors with high EpCAM expression had reduced disease-free survival (DFS) and overall survival (OS) (p < 0.001) compared to patients with cancers with low EpCAM expression. In addition, the concurrent presence of perineural invasion and positive EpCAM expression appeared to be associated with shorter disease-free survival and overall survival. In conclusion, our study confirmed the prognostic value of detecting perineural invasion and EpCAM expression.
Collapse
Affiliation(s)
- Ioanna Kalaitsidou
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.V.)
- Department of Cranio-Maxillofacial Surgery, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Nikoleta Pasteli
- Pathology Department, G. Papanikolaou Hospital, 57010 Thessaloniki, Greece
| | - Gregory Venetis
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.V.)
| | - Athanasios Poulopoulos
- Department of Oral Medicine and Maxillofacial Pathology, Aristotle University, 54124 Thessaloniki, Greece;
| | - Konstantinos Antoniades
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.V.)
| |
Collapse
|
32
|
Dong X, Wu W, Pan P, Zhang XZ. Engineered Living Materials for Advanced Diseases Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2304963. [PMID: 37436776 DOI: 10.1002/adma.202304963] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/13/2023]
Abstract
Natural living materials serving as biotherapeutics exhibit great potential for treating various diseases owing to their immunoactivity, tissue targeting, and other biological activities. In this review, the recent developments in engineered living materials, including mammalian cells, bacteria, viruses, fungi, microalgae, plants, and their active derivatives that are used for treating various diseases are summarized. Further, the future perspectives and challenges of such engineered living material-based biotherapeutics are discussed to provide considerations for future advances in biomedical applications.
Collapse
Affiliation(s)
- Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Wei Wu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Pei Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
33
|
Geng P, Chi Y, Yuan Y, Yang M, Zhao X, Liu Z, Liu G, Liu Y, Zhu L, Wang S. Novel chimeric antigen receptor T cell-based immunotherapy: a perspective for triple-negative breast cancer. Front Cell Dev Biol 2023; 11:1158539. [PMID: 37457288 PMCID: PMC10339351 DOI: 10.3389/fcell.2023.1158539] [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: 02/04/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and does not express estrogen receptor (ER), progesterone (PR), or human epidermal growth factor receptor 2 (HER2). It has a poor prognosis, and traditional endocrine and anti-HER2 targeted therapies have low efficacy against it. In contrast, surgery, radiotherapy, and/or systemic chemotherapy are relatively effective at controlling TNBC. The resistance of TNBC to currently available clinical therapies has had a significantly negative impact on its treatment outcomes. Hence, new therapeutic options are urgently required. Chimeric antigen receptor T cell (CAR-T) therapy is a type of immunotherapy that integrates the antigen specificity of antibodies and the tumor-killing effect of T cells. CAR-T therapy has demonstrated excellent clinical efficacy against hematological cancers. However, its efficacy against solid tumors such as TNBC is inadequate. The present review aimed to investigate various aspects of CAR-T administration as TNBC therapy. We summarized the potential therapeutic targets of CAR-T that were identified in preclinical studies and clinical trials on TNBC. We addressed the limitations of using CAR-T in the treatment of TNBC in particular and solid tumors in general and explored key strategies to overcome these impediments. Finally, we comprehensively examined the advancement of CAR-T immunotherapy as well as countermeasures that could improve its efficacy as a TNBC treatment and the prognosis of patients with this type of cancer.
Collapse
Affiliation(s)
- Peizhen Geng
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| | - Yuhua Chi
- Department of General Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yuan Yuan
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| | - Maoquan Yang
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| | - Xiaohua Zhao
- Department of Thoracic Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Zhengchun Liu
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| | - Guangwei Liu
- Key Laboratory of Precision Radiation Therapy for Tumors in Weifang City, Department of Radiotherapy, School of Medical Imaging, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| | - Yihui Liu
- Key Laboratory of Precision Radiation Therapy for Tumors in Weifang City, Department of Radiotherapy, School of Medical Imaging, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| | - Liang Zhu
- Clinical Research Center, Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Shuai Wang
- Key Laboratory of Precision Radiation Therapy for Tumors in Weifang City, Department of Radiotherapy, School of Medical Imaging, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
34
|
Hu T, Huang Y, Liu J, Shen C, Wu F, He Z. Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy. Pharmaceutics 2023; 15:1821. [PMID: 37514008 PMCID: PMC10383408 DOI: 10.3390/pharmaceutics15071821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy can significantly prevent tumor growth and metastasis by activating the autoimmune system without destroying normal cells. Although cancer immunotherapy has made some achievements in clinical cancer treatment, it is still restricted by systemic immunotoxicity, immune cell dysfunction, cancer heterogeneity, and the immunosuppressive tumor microenvironment (ITME). Biomimetic cell-derived nanoparticles are attracting considerable interest due to their better biocompatibility and lower immunogenicity. Moreover, biomimetic cell-derived nanoparticles can achieve different preferred biological effects due to their inherent abundant source cell-relevant functions. This review summarizes the latest developments in biomimetic cell-derived nanoparticles for cancer immunotherapy, discusses the applications of each biomimetic system in cancer immunotherapy, and analyzes the challenges for clinical transformation.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuezhou Huang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chao Shen
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fengbo Wu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
35
|
Varzandeh M, Sabouri L, Mansouri V, Gharibshahian M, Beheshtizadeh N, Hamblin MR, Rezaei N. Application of nano-radiosensitizers in combination cancer therapy. Bioeng Transl Med 2023; 8:e10498. [PMID: 37206240 PMCID: PMC10189501 DOI: 10.1002/btm2.10498] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 11/08/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Radiosensitizers are compounds or nanostructures, which can improve the efficiency of ionizing radiation to kill cells. Radiosensitization increases the susceptibility of cancer cells to radiation-induced killing, while simultaneously reducing the potentially damaging effect on the cellular structure and function of the surrounding healthy tissues. Therefore, radiosensitizers are therapeutic agents used to boost the effectiveness of radiation treatment. The complexity and heterogeneity of cancer, and the multifactorial nature of its pathophysiology has led to many approaches to treatment. The effectiveness of each approach has been proven to some extent, but no definitive treatment to eradicate cancer has been discovered. The current review discusses a broad range of nano-radiosensitizers, summarizing possible combinations of radiosensitizing NPs with several other types of cancer therapy options, focusing on the benefits and drawbacks, challenges, and future prospects.
Collapse
Affiliation(s)
- Mohammad Varzandeh
- Department of Materials EngineeringIsfahan University of TechnologyIsfahanIran
| | - Leila Sabouri
- AmitisGen TECH Dev GroupTehranIran
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Vahid Mansouri
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical SciencesTehranIran
| | - Maliheh Gharibshahian
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeSchool of Medicine, Shahroud University of Medical SciencesShahroudIran
| | - Nima Beheshtizadeh
- Regenerative Medicine Group (REMED)Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Tissue EngineeringSchool of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
| | - Michael R. Hamblin
- Laser Research Center, Faculty of Health ScienceUniversity of JohannesburgDoornfonteinSouth Africa
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)Universal Scientific Education and Research Network (USERN)TehranIran
- Research Center for ImmunodeficienciesChildren's Medical Center, Tehran University of Medical SciencesTehranIran
- Department of ImmunologySchool of Medicine, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
36
|
Torabian P, Yousefi H, Fallah A, Moradi Z, Naderi T, Delavar MR, Ertas YN, Zarrabi A, Aref AR. Cancer stem cell-mediated drug resistance: A comprehensive gene expression profile analysis in breast cancer. Pathol Res Pract 2023; 246:154482. [PMID: 37196466 DOI: 10.1016/j.prp.2023.154482] [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: 01/23/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/19/2023]
Abstract
Breast cancer is the most frequently diagnosed malignancy in women and a major public health concern. In the current report, differential expression of the breast cancer resistance promoting genes with a focus on breast cancer stem cell related elements as well as the correlation of their mRNAs with various clinicopathologic characteristics, including molecular subtypes, tumor grade/stage, and methylation status, have been investigated using METABRIC and TCGA datasets. To achieve this goal, we downloaded gene expression data of breast cancer patients from TCGA and METABRIC. Then, statistical analyses were used to assess the correlation between the expression levels of stem cell related drug resistant genes and methylation status, tumor grades, various molecular subtypes, and some cancer hallmark gene sets such as immune evasion, metastasis, and angiogenesis. According to the results of this study, a number of stem cell related drug resistant genes are deregulated in breast cancer patients. Furthermore, we observe negative correlations between methylation of resistance genes and mRNA expression. There is a significant difference in the expression of resistance-promoting genes between different molecular subtypes. As mRNA expression and DNA methylation are clearly related, DNA methylation might be a mechanism that regulates these genes in breast cancer cells. As indicated by the differential expression of resistance-promoting genes among various breast cancer molecular subtypes, these genes may function differently in different subtypes of breast cancer. In conclusion, significant deregulation of resistance-promoting factors indicates that these genes may play a significant role in the development of breast cancer.
Collapse
Affiliation(s)
- Pedram Torabian
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Aysan Fallah
- Department of hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Moradi
- Department of hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tohid Naderi
- Department of Laboratory Hematology and Blood Bank, School of Allied Medicine, Shahid Beheshti University of medical sciences, Tehran, Iran
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey; Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396 Istanbul, Turkey
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Li J, Dong T, Wu Z, Zhu D, Gu H. The effects of MYC on tumor immunity and immunotherapy. Cell Death Discov 2023; 9:103. [PMID: 36966168 PMCID: PMC10039951 DOI: 10.1038/s41420-023-01403-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
The oncogene MYC is dysregulated in a host of human cancers, and as an important point of convergence in multitudinous oncogenic signaling pathways, it plays a crucial role in tumor immune regulation in the tumor immune microenvironment (TIME). Specifically, MYC promotes the expression of immunosuppressive factors and inhibits the expression of immune activation regulators. Undoubtedly, a therapeutic strategy that targets MYC can initiate a new era of cancer treatment. In this review, we summarize the essential role of the MYC signaling pathway in tumor immunity and the development status of MYC-related therapies, including therapeutic strategies targeting MYC and combined MYC-based immunotherapy. These studies have reported extraordinary insights into the translational application of MYC in cancer treatment and are conducive to the emergence of more effective immunotherapies for cancer.
Collapse
Affiliation(s)
- Jiajin Li
- Department of Pediatrics, Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Tingyu Dong
- Department of Pediatrics, Second Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Zhen Wu
- Department of Clinical Medicine, First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Dacheng Zhu
- Department of Clinical Medicine, First Clinical School of Medicine, Anhui Medical University, Hefei, China
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
38
|
Jasuja H, Solaymani Mohammadi F, Kim J, Gaba A, Katti DR, Katti KS. Patient-Derived Breast Cancer Bone Metastasis In Vitro Model Using Bone-Mimetic Nanoclay Scaffolds. J Tissue Eng Regen Med 2023; 2023:5753666. [PMID: 40226410 PMCID: PMC11919058 DOI: 10.1155/2023/5753666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/21/2023] [Indexed: 04/15/2025]
Abstract
The unavailability of reliable models for studying breast cancer bone metastasis is the major challenge associated with poor prognosis in advanced-stage breast cancer patients. Breast cancer cells tend to preferentially disseminate to bone and colonize within the remodeling bone to cause bone metastasis. To improve the outcome of patients with breast cancer bone metastasis, we have previously developed a 3D in vitro breast cancer bone metastasis model using human mesenchymal stem cells (hMSCs) and primary breast cancer cell lines (MCF-7 and MDAMB231), recapitulating late-stage of breast cancer metastasis to bone. In the present study, we have tested our model using hMSCs and patient-derived breast cancer cell lines (NT013 and NT023) exhibiting different characteristics. We investigated the effect of breast cancer metastasis on bone growth using this 3D in vitro model and compared our results with previous studies. The results showed that NT013 and NT023 cells exhibiting hormone-positive and triple-negative characteristics underwent mesenchymal to epithelial transition (MET) and formed tumors in the presence of bone microenvironment, in line with our previous results with MCF-7 and MDAMB231 cell lines. In addition, the results showed upregulation of Wnt-related genes in hMSCs, cultured in the presence of excessive ET-1 cytokine released by NT013 cells, while downregulation of Wnt-related genes in the presence of excessive DKK-1, released by NT023 cells, leading to stimulation and abrogation of the osteogenic pathway, respectively, ultimately mimicking different types of bone lesions in breast cancer patients.
Collapse
Affiliation(s)
- Haneesh Jasuja
- Department of Civil Construction and Environmental Engineering, North Dakota State University, Fargo, ND 58108, USA
| | | | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Anu Gaba
- Sanford Roger Maris Cancer Center, Fargo, ND 58102, USA
| | - Dinesh R. Katti
- Department of Civil Construction and Environmental Engineering, North Dakota State University, Fargo, ND 58108, USA
| | - Kalpana S. Katti
- Department of Civil Construction and Environmental Engineering, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
39
|
Li W, Jiang Y, Lu J. Nanotechnology-enabled immunogenic cell death for improved cancer immunotherapy. Int J Pharm 2023; 634:122655. [PMID: 36720448 PMCID: PMC9975075 DOI: 10.1016/j.ijpharm.2023.122655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Tumor immunotherapy has revolutionized the field of oncology treatments in recent years. As one of the promising strategies of cancer immunotherapy, tumor immunogenic cell death (ICD) has shown significant potential for tumor therapy. Nanoparticles are widely used for drug delivery due to their versatile characteristics, such as stability, slow blood elimination, and tumor-targeting ability. To increase the specificity of ICD inducers and improve the efficiency of ICD induction, functionally specific nanoparticles, such as liposomes, nanostructured lipid carriers, micelles, nanodiscs, biomembrane-coated nanoparticles and inorganic nanoparticles have been widely reported as the vehicles to deliver ICD inducers in vivo. In this review, we summarized the strategies of different nanoparticles for ICD-induced cancer immunotherapy, and systematically discussed their advantages and disadvantages as well as provided feasible strategies for solving these problems. We believe that this review will offer some insights into the design of effective nanoparticulate systems for the therapeutic delivery of ICD inducers, thus, promoting the development of ICD-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson 85721, United States.
| |
Collapse
|
40
|
Gao T, Soldatov R, Sarkar H, Kurkiewicz A, Biederstedt E, Loh PR, Kharchenko PV. Haplotype-aware analysis of somatic copy number variations from single-cell transcriptomes. Nat Biotechnol 2023; 41:417-426. [PMID: 36163550 PMCID: PMC10289836 DOI: 10.1038/s41587-022-01468-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022]
Abstract
Genome instability and aberrant alterations of transcriptional programs both play important roles in cancer. Single-cell RNA sequencing (scRNA-seq) has the potential to investigate both genetic and nongenetic sources of tumor heterogeneity in a single assay. Here we present a computational method, Numbat, that integrates haplotype information obtained from population-based phasing with allele and expression signals to enhance detection of copy number variations from scRNA-seq. Numbat exploits the evolutionary relationships between subclones to iteratively infer single-cell copy number profiles and tumor clonal phylogeny. Analysis of 22 tumor samples, including multiple myeloma, gastric, breast and thyroid cancers, shows that Numbat can reconstruct the tumor copy number profile and precisely identify malignant cells in the tumor microenvironment. We identify genetic subpopulations with transcriptional signatures relevant to tumor progression and therapy resistance. Numbat requires neither sample-matched DNA data nor a priori genotyping, and is applicable to a wide range of experimental settings and cancer types.
Collapse
Affiliation(s)
- Teng Gao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Ruslan Soldatov
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Hirak Sarkar
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Adam Kurkiewicz
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Evan Biederstedt
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Po-Ru Loh
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Altos Labs, San Diego, CA, USA.
| |
Collapse
|
41
|
Liu Z, Zhang C, Cui B, Wang Y, Lim K, Li K, Thiery JP, Chen J, Ho CL. Targeted EpCAM-binding for the development of potent and effective anticancer proteins. Biomed Pharmacother 2023; 161:114443. [PMID: 36863098 DOI: 10.1016/j.biopha.2023.114443] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Protein-based cancer therapies are considered an alternative to conventional anticancer regimens, providing multifunctional properties while showing low toxicity. However, its widespread use is limited by absorption and instability issues, resulting in higher dosage requirements and a prolonged onset of bioactivity to elicit the desired response. Here, we developed a non-invasive antitumor treatment using designed ankyrin repeat protein (DARPin)-anticancer protein-conjugate that specifically targets the cancer biomarker, epithelial cell adhesion molecule (EpCAM). The DARPin-anticancer proteins bind to EpCAM-positive cancer cells and improve the in vitro anticancer efficacy by over 100-folds within 24 h, where the DARPin-tagged human lactoferrin fragment (drtHLF4) IC50 value is within the nanomolar range. Orally administered drtHLF4 was readily absorbed into the systemic flow of the HT-29 cancer murine model, exerting its anticancer effect on other tumors in the host body. Orally administered drtHFL4 cleared HT29-colorectal tumors using a single dose, whereas intratumoral injection cleared HT29-subcutaneous tumors within three doses. This approach addresses the limitations of other protein-based anticancer treatments by providing a non-invasive anticancer therapy with improved potency and tumor-specificity.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Chen Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Beiming Cui
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Yijie Wang
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Kaisheng Lim
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Kai Li
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Jean Paul Thiery
- Guangzhou Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China.
| | - Jun Chen
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Chun Loong Ho
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
42
|
Ren XH, Han D, He XY, Guo T, Chen XS, Pang X, Cheng SX. Multi-Targeting Nano-Systems Targeting Heterogeneous Cancer Cells for Therapeutics and Biomarker Detection. Adv Healthc Mater 2023; 12:e2202155. [PMID: 36333906 DOI: 10.1002/adhm.202202155] [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: 08/25/2022] [Revised: 10/15/2022] [Indexed: 11/06/2022]
Abstract
Cancer heterogeneity plays a vital part in cancer resistance and metastasis. To provide a reliable approach to exert a therapy action and evaluate its efficiency in heterogeneous cancer cells, a multiple targeting delivery vector composed of histone encapsulating the therapeutic or diagnostic agent, hyaluronic acid targeting CD44 overexpressed in stem tumor cells, SYL3C aptamer targeting epithelial cell adhesion molecule (EpCAM) overexpressed in epithelial cancer cells, and CL4 aptamer targeting epidermal growth factor receptor (EGFR) overexpressed in mesenchymal cancer cells, is developed. The vector can efficiently target different cancer cells and circulating tumor cells (CTCs) in the peripheral blood of patients for mucin 1 (MUC1) knockout. Furthermore, the multiple targeting vector can be used to co-encapsulate three types of molecular beacons for probing various mRNA biomarkers at single-cell resolution after genome editing. This study provides an efficient approach for exerting therapeutic actions in heterogeneous cancer cells and assessing the therapeutic efficacy by detection of cancer biomarkers via liquid biopsy.
Collapse
Affiliation(s)
- Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, 430072, China
| | - Di Han
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiao-Yan He
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xue-Si Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Xuan Pang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
43
|
Attalla S, Taifour T, Muller W. Tailoring therapies to counter the divergent immune landscapes of breast cancer. Front Cell Dev Biol 2023; 11:1111796. [PMID: 36910138 PMCID: PMC9992199 DOI: 10.3389/fcell.2023.1111796] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/24/2023] Open
Abstract
Breast cancer remains a significant clinical concern affecting millions of women worldwide. Immunotherapy is a rapidly growing drug class that has revolutionized cancer treatment but remains marginally successful in breast cancer. The success of immunotherapy is dependent on the baseline immune responses as well as removing the brakes off pre-existing anti-tumor immunity. In this review, we summarize the different types of immune microenvironment observed in breast cancer as well as provide approaches to target these different immune subtypes. Such approaches have demonstrated pre-clinical success and are currently under clinical evaluation. The impact of combination of these approaches with already approved chemotherapies and immunotherapies may improve patient outcome and survival.
Collapse
Affiliation(s)
- Sherif Attalla
- Department Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tarek Taifour
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Department Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - William Muller
- Department Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Department Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
44
|
D’Accardo C, Porcelli G, Mangiapane LR, Modica C, Pantina VD, Roozafzay N, Di Franco S, Gaggianesi M, Veschi V, Lo Iacono M, Todaro M, Turdo A, Stassi G. Cancer cell targeting by CAR-T cells: A matter of stemness. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:1055028. [PMID: 39086964 PMCID: PMC11285689 DOI: 10.3389/fmmed.2022.1055028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/01/2022] [Indexed: 08/02/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy represents one of the most innovative immunotherapy approaches. The encouraging results achieved by CAR-T cell therapy in hematological disorders paved the way for the employment of CAR engineered T cells in different types of solid tumors. This adoptive cell therapy represents a selective and efficacious approach to eradicate tumors through the recognition of tumor-associated antigens (TAAs). Binding of engineered CAR-T cells to TAAs provokes the release of several cytokines, granzyme, and perforin that ultimately lead to cancer cells elimination and patient's immune system boosting. Within the tumor mass a subpopulation of cancer cells, known as cancer stem cells (CSCs), plays a crucial role in drug resistance, tumor progression, and metastasis. CAR-T cell therapy has indeed been exploited to target CSCs specific antigens as an effective strategy for tumor heterogeneity disruption. Nevertheless, a barrier to the efficacy of CAR-T cell-based therapy is represented by the poor persistence of CAR-T cells into the hostile milieu of the CSCs niche, the development of resistance to single targeting antigen, changes in tumor and T cell metabolism, and the onset of severe adverse effects. CSCs resistance is corroborated by the presence of an immunosuppressive tumor microenvironment (TME), which includes stromal cells, cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and immune cells. The relationship between TME components and CSCs dampens the efficacy of CAR-T cell therapy. To overcome this challenge, the double strategy based on the use of CAR-T cell therapy in combination with chemotherapy could be crucial to evade immunosuppressive TME. Here, we summarize challenges and limitations of CAR-T cell therapy targeting CSCs, with particular emphasis on the role of TME and T cell metabolic demands.
Collapse
Affiliation(s)
- Caterina D’Accardo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Laura Rosa Mangiapane
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Chiara Modica
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Narges Roozafzay
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Melania Lo Iacono
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| |
Collapse
|
45
|
Lee-Theilen M, Fadini DD, Hadhoud JR, van Dongen F, Kroll G, Rolle U, Fiegel HC. Hepatoblastoma Cancer Stem Cells Express PD-L1, Reveal Plasticity and Can Emerge upon Chemotherapy. Cancers (Basel) 2022; 14:5825. [PMID: 36497307 PMCID: PMC9736435 DOI: 10.3390/cancers14235825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
The biology of cancer stem cells (CSCs) of pediatric cancers, such as hepatoblastoma, is sparsely explored. This is mainly due to the very immature nature of these tumors, which complicates the distinction of CSCs from the other tumor cells. Previously, we identified a CSC population in hepatoblastoma cell lines expressing the CSC markers CD34 and CD90, cell surface Vimentin (csVimentin) and binding of OV-6. In this study, we detected the co-expression of the immune escape factor PD-L1 in the CSC population, whereas the other tumor cells remained negative. FACS data revealed that non-CSCs give rise to CSCs, reflecting plasticity of CSCs and non-CSCs in hepatoblastoma as seen in other tumors. When we treated cells with cisplatin and decitabine, a new CD34+/lowOV-6lowCD90+ population emerged that lacked csVimentin and PD-L1 expression. Expression analyses showed that this new CSC subset shared similar pluripotency and EMT features with the already-known CSCs. FACS results further revealed that this subset is also generated from non-CSCs. In conclusion, we showed that hepatoblastoma CSCs express PD-L1 and that the biology of hepatoblastoma CSCs is of a plastic nature. Chemotherapeutic treatment leads to another CSC subset, which is highly chemoresistant and could be responsible for a poor prognosis after postoperative chemotherapy.
Collapse
Affiliation(s)
- Mieun Lee-Theilen
- Department of Pediatric Surgery and Pediatric Urology, University Hospital, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Fründt T, von Felden J, Krause J, Heumann A, Li J, Riethdorf S, Pantel K, Huber S, Lohse AW, Wege H, Schulze K. Circulating tumor cells as a preoperative risk marker for occult metastases in patients with resectable cholangiocarcinoma. Front Oncol 2022; 12:941660. [PMID: 36439492 PMCID: PMC9685781 DOI: 10.3389/fonc.2022.941660] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/21/2022] [Indexed: 11/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive tumor associated with a high rate of recurrence after resection. An important risk factor for recurrence is the presence of occult metasta-ses, which are not radiologically detectable at the time of diagnosis. There are currently no biomarkers for the preoperative assessment of micrometastases. A previous study demonstrated the prognostic relevance of circulating tumor cells (CTC) in patients with advanced CCA but the potential of CTCs as a preoperative marker for detecting occult metastases has not been investigated so far. In this two-phase study, we first recruited a cohort of 27 patients with histologically proven, metastatic CCA or gallbladder cancer (GBCA) to assess feasibility (feasibility cohort, FC). CTCs were measured in the peripheral blood using the CellSearch System (CSS) between October 2012 and January 2017. Subsequently, in 11 patients undergoing curative-intended resection for CCA (intrahepatic CCA: n =4; extrahepatic CCA n= 6; gallbladder cancer: n=1), peripheral and central venous blood specimens were obtained to improve detection rate by simultaneous measurement and to elucidate distribution of CTCs in different venous compartments. Presence of CTCs detection was correlated with postoperative TNM-status. In the FC, CTCs (range 1-3 cells, median: 1) were detected in 40% (11/27) patients and were signifi-cantly associated with worse overall survival (hazard ratio: 3.59; 95% CI: 1.79- 7.1; p = 0.04). By combined peripheral and central measurement, CTC detection was increased to 54% (6/11) in the resection cohort (RC) and was associated with metastases that were only identified during the surgical procedure (peritoneal carcinoma: n = 1; infiltration of the duodenum: n = 1) or immediately after surgery (evidence of pulmonary metastases by CT scan two days after resection, not evident on initial tumor staging prior resection). Taken together, in this single center pilot study, we demonstrated that CTCs are detectable in CCA patients and are associated with significantly impaired survival in patients at metastatic stage. Detection rate prior to surgery was improved to >50% by combined peripheral and central measurement. Moreover, preoperative CTC detection may indicate existing metastases and could help to stratify patients more accurately.
Collapse
|
47
|
Guo H, Zhang W, Wang L, Shao Z, Huang X. Biomimetic cell membrane-coated glucose/oxygen-exhausting nanoreactor for remodeling tumor microenvironment in targeted hypoxic tumor therapy. Biomaterials 2022; 290:121821. [DOI: 10.1016/j.biomaterials.2022.121821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/10/2022] [Accepted: 09/23/2022] [Indexed: 11/02/2022]
|
48
|
Tian R, Li X, Zhang H, Ma L, Zhang H, Wang Z. Ulex Europaeus Agglutinin-I-Based Magnetic Isolation for the Efficient and Specific Capture of SW480 Circulating Colorectal Tumor Cells. ACS OMEGA 2022; 7:30405-30411. [PMID: 36061664 PMCID: PMC9435041 DOI: 10.1021/acsomega.2c03702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
The efficient and specific capture of circulating tumor cells (CTCs) from patients' peripheral blood is of significant value in precise cancer diagnosis and cancer therapy. As fine targeting molecules, lectins can recognize cancer cells specifically due to the abnormal glycosylation of molecules on the cancer cell membrane and the specific binding of lectin with glycoconjugates. Herein, a Ulex europaeus agglutinin-I (UEA-I)-based magnetic isolation strategy was developed to efficiently and specifically capture α-1,2-fucose overexpression CTCs from colorectal cancer (CRC) patients' peripheral blood. Using UEA-I-modified Fe3O4 magnetic beads (termed MB-UEA-I), up to 94 and 89% of target cells (i.e., SW480 CRC cells) were captured from the cell spiking complete cell culture medium and whole blood, respectively. More than 90% of captured cells show good viability and proliferation ability without detaching from MB-UEA-I. In combination with three-color immunocytochemistry (ICC) identification, MB-UEA-I has been successfully used to capture CTCs from CRC patients' peripheral blood. The experimental results indicate a correlation between CTC characterization and tumor metastasis. Specifically, MB-UEA-I can be applied to screen early CRC by capturing CTCs when served as a liquid biopsy. The presented work offers a new insight into developing cost-effective lectin-functionalized methods for biomedical applications.
Collapse
Affiliation(s)
- Rongrong Tian
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- University
of Science and Technology of China, Jinzhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| | - Xiaodong Li
- Department
of Radiology, The First Hospital of Jilin
University, Changchun, Jilin 130021, P. R. China
| | - Hua Zhang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Lina Ma
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Huimao Zhang
- Department
of Radiology, The First Hospital of Jilin
University, Changchun, Jilin 130021, P. R. China
| | - Zhenxin Wang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- University
of Science and Technology of China, Jinzhai Road, Baohe District, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
49
|
Wang L, Jin Z, Master RP, Maharjan CK, Carelock ME, Reccoppa TBA, Kim MC, Kolb R, Zhang W. Breast Cancer Stem Cells: Signaling Pathways, Cellular Interactions, and Therapeutic Implications. Cancers (Basel) 2022; 14:3287. [PMID: 35805056 PMCID: PMC9265870 DOI: 10.3390/cancers14133287] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/02/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
Breast cancer stem cells (BCSCs) constitute a small population of cells within breast cancer and are characterized by their ability to self-renew, differentiate, and recapitulate the heterogeneity of the tumor. Clinically, BCSCs have been correlated with cancer progression, metastasis, relapse, and drug resistance. The tumorigenic roles of BCSCs have been extensively reviewed and will not be the major focus of the current review. Here, we aim to highlight how the crucial intrinsic signaling pathways regulate the fate of BCSCs, including the Wnt, Notch, Hedgehog, and NF-κB signaling pathways, as well as how different cell populations crosstalk with BCSCs within the TME, including adipocytes, endothelial cells, fibroblasts, and immune cells. Based on the molecular and cellular activities of BCSCs, we will also summarize the targeting strategies for BCSCs and related clinical trials. This review will highlight that BCSC development in breast cancer is impacted by both BCSC endogenous signaling and external factors in the TME, which provides an insight into how to establish a comprehensively therapeutic strategy to target BCSCs for breast cancer treatments.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Immunology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zeng Jin
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan P. Master
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Chandra K. Maharjan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Madison E. Carelock
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tiffany B. A. Reccoppa
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Department of Biology, College of Liberal Arts & Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
50
|
Biomimetic Nanotherapeutics: Employing Nanoghosts to fight Melanoma. Eur J Pharm Biopharm 2022; 177:157-174. [PMID: 35787429 DOI: 10.1016/j.ejpb.2022.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022]
Abstract
Melanoma is a cancer of melanocytes present at the basal layer of the skin. Nanomedicine has armed us with competent platform to manage such fatal neoplastic diseases. Nevertheless, it suffers from numerous pitfalls such as rapid clearance and opsonization of surface-functionalized carriers, biocompatibility and idiopathic reactions which could be difficult to predict in the patient. Biomimetic approach, a novel step towards personalized medicine bridges these drawbacks by employing endogenous cell membranes to traverse physiological barriers. Camouflaged carriers coated with natural cell membranes possess unique characteristics such as high circulatory periods, and the absence of allogenic and xenogenic responses. Proteins residing on the cell membranes render a diverse range of utilities to the coated nanoparticles including natural efficiency to identify cellular targets, homologous targeting, reticuloendothelial system evasion, biocompatibility and reduced adverse and idiopathic effects. In the present article, we have focused on cell membrane camouflaged nanocarriers for melanoma management. We have discussed various types of biomimetic systems, their processing and coating approaches, and their characterization. We have also enumerated novel avenues in melanoma treatment and the combination of biomimetic systems with smart nanoparticulate systems with the potential to bring breakthroughs in the near future. Additionally, immunotherapy-based biomimetic systems to combat melanoma have been highlighted. Hurdles towards clinical translation and ways to overcome them have been explained in detail.
Collapse
|