1
|
Chatterjee O, Kaur GA, Shukla N, Balayan S, Singh PK, Chatterjee S, Tiwari A. Multifaceted arsenal in SELEX nanomedicine. Adv Colloid Interface Sci 2025; 342:103540. [PMID: 40344950 DOI: 10.1016/j.cis.2025.103540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Aptamers, short oligonucleotide sequences that bind specifically to cellular proteins and receptors, are emerging as versatile tools in molecular nanomedicine. Unlike passive tumor targeting via the enhanced permeability and retention (EPR) effect, aptamers enable precise drug delivery, enhancing therapeutic efficacy while minimizing side effects. Developed through the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, aptamers offer compact size, robust structure, chemical versatility, and cost-effective synthesis. They serve as effective delivery vehicles for therapeutic molecules, including miRNA, siRNA, and small-molecule drugs, and function as antibody-like ligands for applications in cancer, diabetes, and autoimmune disorders. Since the approval of Macugen, the first aptamer targeting VEGF, aptamers have also shown promise as diagnostic sensors and theranostic agents. This review explores SELEX-derived aptamers in nanomedicine, focusing on their therapeutic and diagnostic roles, particularly in precision cancer therapies. It also addresses challenges such as degradation and clinical translation alongside prospects in vaccines, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Oishika Chatterjee
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden; Department of Biological Sciences, Bose Institute Unified Academic Campus EN 80, Sector 5, Bidhan Nagar (Salt Lake City) Kolkata 700 091, WB, India
| | - Gun Anit Kaur
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Nutan Shukla
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Sapna Balayan
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Pravin Kumar Singh
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Subhrangsu Chatterjee
- Department of Biological Sciences, Bose Institute Unified Academic Campus EN 80, Sector 5, Bidhan Nagar (Salt Lake City) Kolkata 700 091, WB, India.
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden.
| |
Collapse
|
2
|
Yin L, Xu P, Huang Y, Gu X, Sun L, Zhou H, Zhou W, Xie C, Fan Q. Glutathione-Responsive Near-Infrared-II Fluorescence Probe for Early and Accurate Detection of In Situ and Metastatic Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503257. [PMID: 40434227 DOI: 10.1002/smll.202503257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/14/2025] [Indexed: 05/29/2025]
Abstract
In situ and metastatic malignant tumors are primary diseases that threaten human life. Among all the metastases, liver metastasis is the most difficult to detect. As most imaging probes have high liver accumulation, it is difficult to distinguish tiny metastases from normal liver tissue with strong background signal. In this study, the design of a novel second near-infrared window (NIR-II) fluorescence probe for precise detection of carcinoma in situ and liver metastases is presented. The probe called Tg-RGD utilizes a commercially available cyanine dye IR-806 as the signaling moiety, a disulfide bond linker as the responsive moiety, an RGD-capped poly(ethylene glycol) (PEG) as the water soluble enhancer, and the tumor targeting moiety. Tg-RGD shows good glutathione (GSH) responsiveness and selectivity, where its NIR-II fluorescence intensity can enhance 50-fold after activation. In vivo study indicates that Tg-RGD shows much better imaging and targeting effects than Tg-PEG with a similar structure but without RGD moiety for both orthotopic breast cancer and osteosarcoma. Most importantly, Tg-RGD can detect tiny liver metastases with high signal-to-background ratio (3.2). Thus, this study reports a high-performance tumor-specific NIR-II fluorescence probe for in situ and tiny metastatic tumor detection, and may further broaden the applications into related tumor lesions.
Collapse
Affiliation(s)
- Likun Yin
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Pu Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Yuxin Huang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Xuxuan Gu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Liwen Sun
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Hui Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Wen Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Chen Xie
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Quli Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| |
Collapse
|
3
|
Russo A, Tellone E, Farsaci F. Exploring Tumor Cell-Platelet Biochemical Interactions by Dielectric Measurements of Blood: A Potential Target for Tumor Detection and Staging. BIOLOGY 2025; 14:542. [PMID: 40427731 PMCID: PMC12108633 DOI: 10.3390/biology14050542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025]
Abstract
This paper aims to investigate the dielectric properties of blood for tumor detection and staging. The application of complex thermodynamic models and the study of the trend over time of some thermodynamic functions have allowed us to highlight the generation of displacement currents caused by changes in charge, i.e., by the activation and consequent accumulation of platelets on migrating tumor cells. Although few studies exist to date in this regard, the technique used has provided promising results, especially in terms of building a database. In this context, the evaluation of the dielectric parameters of healthy and cancerous blood can be exploited for the staging of cancer. The main advantages of this method include easy application, non-invasiveness, low cost, and online monitoring.
Collapse
Affiliation(s)
- Annamaria Russo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (E.T.); (F.F.)
| | | | | |
Collapse
|
4
|
Zhang J, Cheng L, Jiang S, Zhu D. Machine learning based identification of anoikis related gene classification patterns and immunoinfiltration characteristics in diabetic nephropathy. Sci Rep 2025; 15:15271. [PMID: 40312440 PMCID: PMC12046048 DOI: 10.1038/s41598-025-99395-w] [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: 07/06/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Anoikis and immune cell infiltration are pivotal factors in the pathophysiological mechanism of diabetic nephropathy (DN), yet a comprehensive understanding of the mechanism is lacking. This work aimed to pinpoint distinctive anoikis-related genes (ARGs) in DN and delve into their impact on the immune landscape. Three datasets (GSE30528, GSE47184, and GSE96804) were downloaded from the gene expression omnibus (GEO) dataset. Differentially expressed genes (DEGs) were identified using the "limma" package, while ARGs were obtained from GSEA, GeneCard, and Harmonizome datasets. The intersection of DEGs and ARGs was analyzed for Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. The CIBERSORT algorithm was employed to estimate the infiltration percentage of 22 immune cell types in DN renal tissue. Subsequently, the least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF) algorithms were adopted to screen key ARGs related to DN. After that, receiver operating characteristic (ROC) analysis was employed to assess the diagnostic accuracy of each gene and the real-time quantitative polymerase chain reaction (RT-qPCR) was adopted to quantitatively detect the expression of biomarkers in DN cell models. Finally, correlations between key genes and immune cell infiltration were analyzed, and a competitive endogenous ribonucleic acid (RNA) (ceRNA) network based on key genes was constructed. A total of 59 DEARGs were identified. GO functional annotation enrichment analysis revealed their involvement in kidney development, extracellular matrix (ECM), cytoplasmic vesicle cavity, immunoinflammatory response, and cytokine effect. KEGG pathway analysis indicated that MAPK, PI3K -Akt, IL -17, TNF, and HIF- 1 signaling pathways are critical for DN. In addition, seven key genes, including PDK4, S100A8, HTRA1, CHI3L1, WT1, CDKN1B, and EGF, were screened by machine learning algorithm. Most of these genes exhibited low expression in renal tissue of DN patients and positive correlation with neutrophils, and their expressions were verified in an external dataset cell model. The ceRNA analysis suggested potential regulatory pathways (H19/miR-15b-5p/PDK4 and KCNQ1T1/miR-1207-3p/WT1) influencing early DN progression. This work provided a comprehensive analysis of the role of DEARGs in DN for the first time, offering valuable insights for further understanding the disease mechanism and guiding clinical diagnosis, treatment, and research of DN.
Collapse
Affiliation(s)
- Jing Zhang
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China
| | - Lulu Cheng
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Shan Jiang
- Traditional Chinese Medicine Department of Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Duosheng Zhu
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China.
| |
Collapse
|
5
|
Bayrak O, Alper M, Basbinar Y, Bayrak S. The role of thrombin in the paradoxical interplay of cancer metastasis and the vascular system: A driving dynamic. Biomed Pharmacother 2025; 186:118031. [PMID: 40215647 DOI: 10.1016/j.biopha.2025.118031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
The coagulation system plays a complex role in cancer therapy. Endothelial damage and tissue factor increased by chemotherapy initiate the coagulation cascade, producing active FXa and releasing thrombin. Thrombin triggers tumor growth and metastasis, leading to severe thromboembolic events in cancer patients. Direct thrombin inhibitors do not have the expected anti-metastatic effect as PAR-2 remains active and increases the risk of bleeding. Therefore, dual inhibition of thrombin by FXa inhibition and plasmin inhibition, which converts fibrin to fibrinogen, is targeted. Clinical studies show that the use of tranexamic acid in patients on NOAC therapy may be beneficial without increasing the risk of bleeding. This approach offers a promising strategy to provide an anti-metastatic effect in cancer treatment.
Collapse
Affiliation(s)
- Ozge Bayrak
- Dokuz Eylul University, Institute of Health Sciences, Department of Oncology, Izmir, Turkey
| | - Meltem Alper
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Yasemin Basbinar
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Serdar Bayrak
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey; Dokuz Eylul University, Faculty of Medicine, Department of Cardiovascular Surgery, Izmir, Turkey.
| |
Collapse
|
6
|
Liu J, Yu Z, Liu Q, Dou C, Cao P, Xie X. A novel 5-differentially expressed gene (DEG) signature predicting the prognosis in patients with metastatic liver malignancies and the prognostic and therapeutic potential of SPP1. Int J Clin Oncol 2025; 30:956-973. [PMID: 40014188 DOI: 10.1007/s10147-025-02723-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND This study aimed to identify differentially expressed genes (DEGs) that are associated with hepatocarcinogenesis and metastasis in hepatocellular carcinoma (HCC) and to explore their value in predicting overall survival (OS). The methods used included bioinformatics analysis of gene expression datasets and in vitro experiments using HCC cell lines. METHODS Gene expression profiles from metastatic and non-metastatic liver cancer specimens were analyzed using the limma R package. Functional enrichment was performed using Metascape. A prognostic 5-gene signature was constructed using the LASSO algorithm based on TCGA-LIHC data. Kaplan-Meier survival analysis assessed the association of these genes with clinical outcomes (DFI, DSS, OS, and PFS). In vitro, Huh7 and Hep3B cells were transfected with shRNA for SPP1 knockdown. Cell viability was measured with CCK-8 assays, and migration was assessed with Transwell and wound-healing assays. Protein expression was evaluated via western blotting. RESULTS The analysis of gene expression profiles led to the identification of 11 DEGs associated with immune response, phagocytosis, and cell migration. From these DEGs, the LASSO algorithm identified a 5-DEG signature (MASP1, MASP2, MUC1, TREM1, and SPP1) that was predictive of OS in liver cancer patients. Among the five genes, SPP1 was the most upregulated in cancer samples and was significantly associated with poorer outcomes, including DFI, DSS, OS, and PFS. In vitro experiments confirmed that SPP1 knockdown in Huh7 and Hep3B cells significantly inhibited cancer cell viability and migration. Western blot analysis showed alterations in key proteins, with a reduction in vimentin and Ki-67 and an increase in E-cadherin following SPP1 knockdown. CONCLUSION This study highlights the pivotal effect of SPP1 on HCC development and underscores its potential as a biomarker for the OS of liver cancer patients. The identified DEGs may serve as predictive markers for OS and potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Jing Liu
- Department of Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zijian Yu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qiong Liu
- Department of Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Chengyun Dou
- Department of Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Peng Cao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Xia Xie
- Department of Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
7
|
Liu L, He Y, Du H, Tang M, Wang T, Tan J, Zha L, Yang L, Ashrafizadeh M, Tian Y, Zhou H. Biological profile of breast cancer brain metastasis. Acta Neuropathol Commun 2025; 13:78. [PMID: 40253355 PMCID: PMC12008903 DOI: 10.1186/s40478-025-01983-4] [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: 07/29/2024] [Accepted: 03/08/2025] [Indexed: 04/21/2025] Open
Abstract
Breast cancer is one of the leading causes of death worldwide. The aggressive behaviour of breast tumor results from their metastasis. Notably, the brain tissue is one of the common regions of metastasis, thereby reducing the overall survival of patients. Moreover, the metastatic tumors demonstrate poor response or resistance to therapies. In addition, breast cancer brain metastasis provides the poor prognosis of patients. Therefore, it is of importance to understand the mechanisms in breast cancer brain metastasis. Both cell lines and animal models have been developed for the evaluation of breast cancer brain metastasis. Moreover, different tumor microenvironment components and other factors such as lymphocytes and astrocytes can affect brain metastasis. The breast cancer cells can disrupt the blood-brain barrier (BBB) during their metastasis into brain, developing blood-tumor barrier to enhance carcinogenesis. The breast cancer brain metastasis can be increased by the dysregulation of chemokines, STAT3, Wnt, Notch and PI3K/Akt. On the other hand, the effective therapeutics have been developed for the brain metastasis such as introduction of nanoparticles. Moreover, the disruption of BBB by ultrasound can increase the entrance of bioactive compounds to the brain tissue. In order to improve specificity and selectivity, the nanoparticles for the delivery of therapeutics and crossing over BBB have been developed to suppress breast cancer brain metastasis.
Collapse
Affiliation(s)
- Li Liu
- Department of Oncology, Suining Central Hospital, Suning, 629000, China
| | - Yuan He
- Department of Oncology, Yunyang County People's Hospital, Chongqing, 404500, China
| | - Hongyu Du
- Department of General Medicine, The Seventh People's Hospital of Chongqing, The Central Hospital Affiliated to Chongging University of Technology, Chongqing, 400054, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Tingting Wang
- Department of Gynecology and Obstetrics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jieren Tan
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, PR China
| | - Lisha Zha
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, PR China
| | - Li Yang
- Department of Nephrology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China.
| | - Yu Tian
- School of Public Health, Benedictine University, No.5700 College Road, Lisle, IL, 60532, USA.
- Research Center, the Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Guangzhou, 510080, China.
| |
Collapse
|
8
|
Labidi S, Mulla N, Elkholi IE, Capella MP, Rose AAN, Panasci L, Ferrario C, Basik M, Fallah P. High Ki-67 expression is associated with increased risk of distant recurrence in Oncotype Dx low risk breast cancer. Clin Breast Cancer 2025:S1526-8209(25)00092-8. [PMID: 40319004 DOI: 10.1016/j.clbc.2025.04.001] [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: 10/08/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
PURPOSE To assess whether high Ki-67 protein expression level could independently predict the distant recurrence in early-stage breast cancer with low Oncotype Dx scores (≤ 25). METHODS This single-center retrospective cohort study included 278 patients with hormone receptor positive (HR+) human epidermal growth factor receptor 2 negative (HER2-), T1-2N0M0, low Oncotype Dx recurrence score (RS) (≤ 25) breast cancer. We identified 2 groups: "high Ki-67″ ≥ 15% (n = 130, 47%) and "low Ki-67″ < 15% (n = 148, 53%). Clinical characteristics, treatment and survival were abstracted from chart review. Fisher's exact test was used to assess differences between Ki-67 groups. Cox-regression models were used to assess differences in survival. RESULTS After a median follow up of 7 years, 13 (4.7%) patients experienced distant metastasis. Recurrence rate was significantly higher in the "high Ki-67″ group 9.2% (12/130) versus the "low Ki-67″ group 0.7% (1/148) (P = .001). Distant metastasis-free survival (dMFS) was significantly shorter in the "high Ki-67″ group (HR 12.90, 95% CI, 12.53-13.27, P = .008). Tumor size ≥ 2 cm was associated with shorter dMFS (HR, 12.90; 95% CI, 12.53-13.27; P < .001). In a multivariable analysis, tumor size ≥ 2 cm and "High Ki-67″ were independent prognosis factors for dMFS. CONCLUSION Ki-67 expression level may help to identify a subset of low risk Oncotype Dx patients who could benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Soumaya Labidi
- Segal Cancer Center, Jewish General Hospital, Montréal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada
| | - Nasser Mulla
- College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Islam E Elkholi
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Quebec, Canada
| | | | - April A N Rose
- Segal Cancer Center, Jewish General Hospital, Montréal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Quebec, Canada
| | - Lawrence Panasci
- Segal Cancer Center, Jewish General Hospital, Montréal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada
| | - Cristiano Ferrario
- Segal Cancer Center, Jewish General Hospital, Montréal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada
| | - Mark Basik
- Segal Cancer Center, Jewish General Hospital, Montréal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Quebec, Canada
| | - Parvaneh Fallah
- Segal Cancer Center, Jewish General Hospital, Montréal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
9
|
Köhler B, Brieger E, Brandstätter T, Hörterer E, Wilk U, Pöhmerer J, Jötten A, Paulitschke P, Broedersz CP, Zahler S, Rädler JO, Wagner E, Roidl A. Unraveling the metastasis-preventing effect of miR-200c in vitro and in vivo. Mol Oncol 2025; 19:1029-1053. [PMID: 39404181 PMCID: PMC11977663 DOI: 10.1002/1878-0261.13712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 04/09/2025] Open
Abstract
Advanced breast cancer, as well as ineffective treatments leading to surviving cancer cells, can result in the dissemination of these malignant cells from the primary tumor to distant organs. Recent research has shown that microRNA 200c (miR-200c) can hamper certain steps of the invasion-metastasis cascade. However, it is still unclear whether miR-200c expression alone is sufficient to prevent breast cancer cells from metastasis formation. Hence, we performed a xenograft mouse experiment with inducible miR-200c expression in MDA-MB 231 cells. The ex vivo analysis of metastatic sites in a multitude of organs, including lung, liver, brain, and spleen, revealed a dramatically reduced metastatic burden in mice with miR-200c-expressing tumors. A fundamental prerequisite for metastasis formation is the motility of cancer cells and, therefore, their migration. Consequently, we analyzed the effect of miR-200c on collective- and single-cell migration in vitro, utilizing MDA-MB 231 and MCF7 cell systems with genetically modified miR-200c expression. Analysis of collective-cell migration revealed confluence-dependent motility of cells with altered miR-200c expression. Additionally, scratch assays showed an enhanced predisposition of miR-200c-negative cells to leave cell clusters. The in-between stage of collective- and single-cell migration was validated using transwell assays, which showed reduced migration of miR-200c-positive cells. Finally, to measure migration at the single-cell level, a novel assay on dumbbell-shaped micropatterns was performed, which revealed that miR-200c critically determines confined cell motility. All of these results demonstrate that sole expression of miR-200c impedes metastasis formation in vivo and migration in vitro and highlights miR-200c as a metastasis suppressor in breast cancer.
Collapse
Affiliation(s)
- Bianca Köhler
- Pharmaceutical Biotechnology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| | - Emily Brieger
- Faculty of Physics and Center for NanoScienceLudwig‐Maximilians‐Universität MünchenGermany
| | - Tom Brandstätter
- Department of Physics and AstronomyVrije Universiteit AmsterdamThe Netherlands
- Arnold‐Sommerfeld‐Center for Theoretical PhysicsLudwig‐Maximilians‐Universität MünchenGermany
| | - Elisa Hörterer
- Pharmaceutical Biotechnology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| | - Ulrich Wilk
- Pharmaceutical Biotechnology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| | - Jana Pöhmerer
- Pharmaceutical Biotechnology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| | - Anna Jötten
- Faculty of Physics and Center for NanoScienceLudwig‐Maximilians‐Universität MünchenGermany
| | - Philipp Paulitschke
- Faculty of Physics and Center for NanoScienceLudwig‐Maximilians‐Universität MünchenGermany
- PHIO Scientific GmbHMunichGermany
| | - Chase P. Broedersz
- Department of Physics and AstronomyVrije Universiteit AmsterdamThe Netherlands
- Arnold‐Sommerfeld‐Center for Theoretical PhysicsLudwig‐Maximilians‐Universität MünchenGermany
| | - Stefan Zahler
- Pharmaceutical Biology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| | - Joachim O. Rädler
- Faculty of Physics and Center for NanoScienceLudwig‐Maximilians‐Universität MünchenGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| | - Andreas Roidl
- Pharmaceutical Biotechnology, Department of PharmacyLudwig‐Maximilians‐Universität MünchenGermany
| |
Collapse
|
10
|
Ocak M, Ateş Ş, Kahveci S, Okan A, Doğanyiğit Z, Uçar S, Yılmaz S. Evaluation of the anticarcinogenic effects of Rutin on brain tissue in mice with Ehrlich ascites carcinoma by micro-computed tomography and histological methods. Asia Pac J Clin Oncol 2025; 21:174-179. [PMID: 38526529 DOI: 10.1111/ajco.14058] [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] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Studies for new treatment strategies on cancer continue, and new searches continue in the diagnosis and evaluation of cancer. This study examined the possible anticarcinogenic effect of Rutin on the brain tissues of male mice with Ehrlich ascites carcinoma (EAC). MATERIAL AND METHODS We used micro-computed tomography (micro-CT) and histologically Hematoxylin&Eosin (H&E) staining methods for evaluation. RESULTS In the evaluation results, we saw a significant decrease in the brain volume of the tumor group to the control group. The difference in volume between the Rutin treatment group and the control group was not significant. In the brain tissues of the tumor group, numerous degenerated neurons characterized by pericellular/perivascular space expansion, cell swelling, or expansion were detected in the cortex and hippocampus regions. We showed a reduction in the damage rate in the Rutin treated group. CONCLUSION As a result, Rutin was found to have an anticarcinogenic effect. In addition to the classical histological evaluation, we used a newer method, micro-CT, in our study. We believe that this study has important results both in terms of its originality and adding new information to the literature.
Collapse
Affiliation(s)
- Mert Ocak
- Department of Anatomy, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Şükrü Ateş
- Department of Anatomy, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Selda Kahveci
- Department of Histology and Embriology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Aslı Okan
- Department of Histology and Embriology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Züleyha Doğanyiğit
- Department of Histology and Embriology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Sümeyye Uçar
- Department of Anatomy, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Seher Yılmaz
- Department of Anatomy, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
11
|
Smaldone G, Di Matteo F, Castelluccio R, Napolitano V, Miranda MR, Manfra M, Campiglia P, Vestuto V. Targeting the CXCR4/CXCL12 Axis in Cancer Therapy: Analysis of Recent Advances in the Development of Potential Anticancer Agents. Molecules 2025; 30:1380. [PMID: 40142155 PMCID: PMC11945090 DOI: 10.3390/molecules30061380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer, a leading cause of premature death, arises from genetic and epigenetic mutations that transform normal cells into tumor cells, enabling them to proliferate, evade cell death, and stimulate angiogenesis. Recent evidence indicates that chemokines are essential in tumor development, activating receptors that promote proliferation, invasion, and metastasis. The CXCR4/CXCL12 signaling pathway is gaining attention as a promising target for cancer therapy. CXCR4, a chemokine receptor, is often overexpressed in various types of cancer, including kidney, lung, brain, prostate, breast, pancreas, ovarian, and melanomas. When it binds to its endogenous ligand, CXCL12, it promotes cell survival, proliferation, and migration, crucial mechanisms for the retention of hematopoietic stem cells in the bone marrow and the movement of lymphocytes. The extensive expression of CXCR4 in cancer, coupled with the constant presence of CXCL12 in various organs, drives the activation of this axis, which in turn facilitates angiogenesis, tumor progression, and metastasis. Given the detrimental role of the CXCR4/CXCL12 axis, the search for drugs acting selectively against this protein represents an open challenge. This review aims to summarize the recent advancements in the design and development of CXCR4 antagonists as potential anticancer agents.
Collapse
Affiliation(s)
- Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Francesca Di Matteo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Roberta Castelluccio
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Valeria Napolitano
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Maria Rosaria Miranda
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy;
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy;
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| |
Collapse
|
12
|
Lee CE, Noh KM, Kim S, Hong J, Kim K. Recent Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Engineering Strategies for Precise Strike Therapy against Tumor. Biomater Res 2025; 29:0170. [PMID: 40110051 PMCID: PMC11922527 DOI: 10.34133/bmr.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025] Open
Abstract
Effective drug delivery relies on the selection of suitable carriers, which is crucial for protein-based therapeutics such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). One of the key advantages of TRAIL is its ability to selectively induce apoptosis in cancer cells excluding healthy tissues by binding to death receptors DR4 and DR5, which are highly expressed in various cancer cells. Despite this promise, the clinical application of TRAIL has been limited by its short half-life, limited stability, and inefficient delivery to tumor sites. To overcome currently available clinical and engineering approaches, a series of sophisticated strategies is required: (a) the design of biomaterial-mediated carriers for enhanced targeting efficacy, particularly via optimizing selected materials, composition, formulation, and surface modulation. Moreover, (b) development of genetically modified cellular products for augmented TRAIL secretion toward tumor microenvironments and (c) cell surface engineering techniques for TRAIL immobilization onto infusible cell populations are also discussed in the present review. Among these approaches, living cell-based carriers offer the distinct advantage of systemically administered TRAIL-functionalized cells capturing circulating tumor cells in the bloodstream, thereby preventing secondary tumor formation. This review provides insight into the development of novel TRAIL delivery platforms, discusses considerations for clinical translation, and suggests future directions and complementary strategies to advance the field of TRAIL-based cancer therapeutics.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyung Mu Noh
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Jiyeon Hong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
13
|
Huilcaman R, Campos A, Contreras P, Simón L, Varas-Godoy M, Grünenwald F, Shao B, Heinecke J, Lobos-Gonzalez L, Leyton L, Quest AFG. Inclusion of ΑVβ3 integrin into extracellular vesicles in a caveolin-1 tyrosine-14- phosphorylation dependent manner and subsequent transfer to recipient melanoma cells promotes migration, invasion and metastasis. Cell Commun Signal 2025; 23:139. [PMID: 40098186 PMCID: PMC11912626 DOI: 10.1186/s12964-025-02131-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Caveolin-1 (CAV1) is a membrane protein that promotes migration, invasion and metastasis of cancer cells when phosphorylated on tyrosine-14 (Y14) by a cell intrinsic mechanism involving the activation of a novel Rab5-Rac1 signaling axis. Moreover, CAV1 expressed in aggressive cancer cells is included into extracellular vesicles (EVs) and such EVs increase the metastatic potential of recipient lower grade cancer cells. However, the relevance of CAV1 Y14 phosphorylation in these extrinsic EV-stimulated events remained to be determined. Here we used B16F10 mouse melanoma cells over-expressing wild-type CAV1, phospho-mimetic CAV1(Y14E) or phospho-null CAV1(Y14F) as models to determine how the EV protein content was affected by Y14 phosphorylation and how these EVs modulated the metastatic potential of recipient B16F10 cells lacking CAV1. EVs from B16F10 cells over-expressing wild-type and CAV1(Y14/E) contain CAV1, and other proteins linked to signaling pathways associated with cell adhesion and migration. CAV1 inclusion in EVs was reduced by the Y14F mutation and global protein composition was also significantly different. Moreover, CAV1 wild-type and CAV1(Y14E) EVs promoted migration, as well as invasion of cells lacking CAV1 [B16F10(Mock) cells]. In addition, β3 integrin was transferred via CAV1(Y14E) EVs to B16F10 (Mock) cells, and treatment with such EVs promoted metastasis of recipient B16F10(Mock) cells. Finally, CAV1(Y14E) EV-enhanced migration, invasion and metastasis of recipient cells was blocked by anti-αVβ3 antibodies. In conclusion, CAV1 phosphorylated on Y14 not only intrinsically promotes migration, invasion and metastasis of cells expressing the protein (in cis), but also favors the inclusion of CAV1 into EVs, as well as the extrinsic acquisition of malignant traits in recipient cells, through integrin transfer (in trans).
Collapse
Grants
- FONDECYT grants 1210644 (A.F.G.Q.), 1200836, 1240888 (L.L.), 1211223 (L.L.-G.), 1190928 (M.V.G.), FONDAP grants 15130011 and 1523A0008 (A.F.G.Q., L.L., L.L.-G., M.V.G.), ANID/BASAL/FB210008 (M.V.G.), ANID postdoctoral fellowship award Becas Chile (A.C.), 3170169 (J.D.), ANID PhD fellowship awards 21130102 (AC), 21161246 (R.H.). FONDECYT, FONDAP, ANID/BASAL/, ANID postdoctoral fellowship award Becas Chile, ANID PhD fellowship awards
Collapse
Affiliation(s)
- R Huilcaman
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Facultad de Ciencias de la Salud, Escuela de Tecnología Médica, Universidad Bernardo OHiggins, General Gana 1702, Santiago, 8370854, Chile
| | - A Campos
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Cancer Research UK Scotland Institute, Garscube Estate. Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - P Contreras
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - L Simón
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Nutrition and Dietetic School, Universidad Finis Terrae, Santiago, Chile
| | - M Varas-Godoy
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, 7510156, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago, 8580702, Chile
| | - F Grünenwald
- Laboratory of Reproductive Biology, Center for Biomedical Research, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Baohai Shao
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98195- 8055, USA
| | - Jay Heinecke
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98195- 8055, USA
| | - L Lobos-Gonzalez
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile
| | - L Leyton
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile.
| | - A F G Quest
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
14
|
Liu Z, Song H, Wang Z, Hu Y, Zhong X, Liu H, Zeng J, Ye Z, Ning W, Liang Y, Yuan S, Deng Z, Jin L, Mo J, Ren J, Yao M. A novel optimized orthotopic mouse model for brain metastasis with sustained cerebral blood circulation and capability of multiple delivery. Clin Exp Metastasis 2025; 42:19. [PMID: 40095206 PMCID: PMC11913983 DOI: 10.1007/s10585-025-10336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025]
Abstract
Brain metastasis is thought to be related to the high mortality and poor prognosis of lung cancer. Despite significant advances in the treatment of primary lung cancer, the unique microenvironment of the brain renders current therapeutic strategies largely ineffective against brain metastasis. The lack of effective drugs for brain metastasis treatment is primarily due to the incomplete understanding of the mechanisms underlying its initiation and progression. Currently, our understanding of brain metastasis remains limited, primarily due to the absence of appropriate models that can realistically simulate the entire process of tumor cell detachment from the primary site, circulation through the bloodstream, and eventual colonization of the brain. Therefore, there is a pressing need to develop more suitable lung cancer brain metastasis models that can effectively replicate these critical stages of metastasis. Here, based on the traditional carotid artery injection model, we established a novel orthotopic mouse model by using a light-controlled hydrogel to repair the puncture site on the carotid artery, with sustained cerebral blood circulation and the capability of multiple delivery cancer cell to mimic lung cancer brain metastasis. The optimized orthotopic mouse model significantly reduced cerebral ischemia and improved cerebral oxygenation by 60% compared to the traditional orthotopic mouse model, enhancing post-operative survival rates. It also showed a reduction in pro-inflammatory cytokines and featured less inflammatory and more resting states of microglial and astrocyte cells. Furthermore, the optimized orthotopic mouse model markedly increased the success rate and absolute number of the metastatic clones in the brain. Additionally, the multiple delivery model based on the optimized orthotopic mouse model substantially augmented the tumor clone number and formation rates compared to single injection in the optimized orthotopic mouse model. This model overcomes previous limitations by maintaining cerebral circulation, providing a more accurate simulation of the continuous entry of tumor cells into cerebral circulation. It offers a robust platform for studying the interactions of cancer cells with the brain microenvironment and testing new therapeutic approaches.
Collapse
Affiliation(s)
- Zihao Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Huisheng Song
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Zhenning Wang
- Department of Neurosurgery, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523059, China
| | - Yang Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Xiaoxuan Zhong
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing andCommunication, Institute of Photonics Technology, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Huiling Liu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Zhiming Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Wenfeng Ning
- Ningyuan County People's Hospital, Yongzhou, 425699, Hunan, China
| | - Yizhi Liang
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing andCommunication, Institute of Photonics Technology, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Shengfang Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Zijun Deng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Long Jin
- Guangdong Provincial Key Laboratory of Optical Fiber Sensing andCommunication, Institute of Photonics Technology, College of Physics & Optoelectronic Engineering, Jinan University, Guangzhou, 510632, China
| | - Jieying Mo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China
| | - Jiaoyan Ren
- School of Food Sciences and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Maojin Yao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, People's Republic of China.
| |
Collapse
|
15
|
Doodmani SM, Safari MH, Akbari M, Farahani N, Alimohammadi M, Aref AR, Tajik F, Maghsoodlou A, Daneshi S, Tabari T, Taheriazam A, Entezari M, Nabavi N, Hashemi M. Metastasis and chemoresistance in breast cancer: Crucial function of ZEB1/2 proteins. Pathol Res Pract 2025; 267:155838. [PMID: 39954369 DOI: 10.1016/j.prp.2025.155838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Breast cancer remains one of the leading causes of mortality worldwide. While advancements in chemotherapy, immunotherapy, radiotherapy, and targeted therapies have significantly improved breast cancer treatment, many patients are diagnosed at advanced stages, where tumor cells exhibit aggressive behavior and therapy resistance. Understanding the mechanisms driving breast cancer progression is therefore critical. Metastasis is a major factor that drastically reduces patient prognosis and survival, accounting for most breast cancer-related deaths. ZEB proteins have emerged as key regulators of cancer metastasis. Beyond their role in metastasis, ZEB proteins also influence drug resistance. This review focuses on the role of ZEB1 and ZEB2 in regulating breast cancer metastasis. These proteins interact with components of the tumor microenvironment (TME) to drive cancer progression and metastasis. Additionally, ZEB proteins regulate angiogenesis through interactions with VEGF. Targeting ZEB proteins offers potential therapeutic benefits, particularly for aggressive breast cancer subtypes such as triple-negative breast cancer (TNBC), which often show poor therapeutic response. ZEB proteins also influence the sensitivity of breast cancer cells to chemotherapy, making them promising targets for enhancing treatment efficacy. Given their upregulation in breast cancer, ZEB proteins can serve as valuable diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Seyed Mohammad Doodmani
- Department of Pathobiology, Faculty of Specialized Veterinary Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences,Tehran, Iran
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Fatemeh Tajik
- Department of Surgery, University of California, Irvine Medical Center, Orange, CA, USA
| | - Amin Maghsoodlou
- Young Researchers and Elite Club, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Teimour Tabari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
16
|
Codrich M, Biasotto A, D’Aurizio F. Circulating Biomarkers of Thyroid Cancer: An Appraisal. J Clin Med 2025; 14:1582. [PMID: 40095491 PMCID: PMC11900207 DOI: 10.3390/jcm14051582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 03/19/2025] Open
Abstract
Thyroid cancer is the most prevalent endocrine cancer. The prognosis depends on the type and stage at diagnosis. Thyroid cancer treatments involve surgery, possibly followed by additional therapeutic options such as hormone therapy, radiation therapy, targeted therapy and chemotherapy. Besides the well-known thyroid tumor biomarkers, new circulating biomarkers are now emerging. Advances in genomic, transcriptomic and proteomic technologies have allowed the development of novel tumor biomarkers. This review explores the current literature data to critically analyze the benefits and limitations of routinely measured circulating biomarkers for the diagnosis and monitoring of thyroid cancer. The review also sheds light on new circulating biomarkers, focusing on the challenges of their use in the clinical management of thyroid cancer, underlining the need for the identification of a new generation of circulating biomarkers.
Collapse
Affiliation(s)
- Marta Codrich
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy; (M.C.); (A.B.)
| | - Alessia Biasotto
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy; (M.C.); (A.B.)
- Institute of Clinical Pathology, Academic Hospital “Santa Maria della Misericordia”, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Federica D’Aurizio
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy; (M.C.); (A.B.)
- Institute of Clinical Pathology, Academic Hospital “Santa Maria della Misericordia”, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| |
Collapse
|
17
|
Peralta M, Dupas A, Larnicol A, Lefebvre O, Goswami R, Stemmelen T, Molitor A, Carapito R, Girardo S, Osmani N, Goetz JG. Endothelial calcium firing mediates the extravasation of metastatic tumor cells. iScience 2025; 28:111690. [PMID: 39898056 PMCID: PMC11787530 DOI: 10.1016/j.isci.2024.111690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Metastatic dissemination is driven by genetic, biochemical, and biophysical cues that favor the distant colonization of organs and the formation of life-threatening secondary tumors. We have previously demonstrated that endothelial cells (ECs) actively remodel during extravasation by enwrapping arrested tumor cells (TCs) and extruding them from the vascular lumen while maintaining perfusion. In this work, we dissect the cellular and molecular mechanisms driving endothelial remodeling. Using high-resolution intravital imaging in zebrafish embryos, we demonstrate that the actomyosin network of ECs controls tissue remodeling and subsequent TC extravasation. Furthermore, we uncovered that this cytoskeletal remodeling is driven by altered endothelial-calcium (Ca2+) signaling caused by arrested TCs. Accordingly, we demonstrated that the inhibition of voltage-dependent calcium L-type channels impairs extravasation. Lastly, we identified P2X4, TRP, and Piezo1 mechano-gated Ca2+ channels as key mediators of the process. These results further highlight the central role of endothelial remodeling during the extravasation of TCs and open avenues for successful therapeutic targeting.
Collapse
Affiliation(s)
- Marina Peralta
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Amandine Dupas
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Annabel Larnicol
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Olivier Lefebvre
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ruchi Goswami
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Tristan Stemmelen
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091 Strasbourg, France
| | - Anne Molitor
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Raphael Carapito
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091 Strasbourg, France
| | - Salvatore Girardo
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Naël Osmani
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jacky G. Goetz
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
18
|
Ferdousmakan S, Mansourian D, Seyedi Asl FS, Fathi Z, Maleki-Sheikhabadi F, Afjadi MN, Zalpoor H. Autophagy induced by metabolic processes leads to solid tumor cell metastatic dormancy and recurrence. Med Oncol 2025; 42:62. [PMID: 39899220 DOI: 10.1007/s12032-025-02607-6] [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: 10/22/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
A crucial cellular mechanism that has a complex impact on the biology of cancer, particularly in solid tumors, is autophagy. This review explores how metabolic processes trigger autophagy, which helps metastatic tumor cells go dormant and recur. During metastasis, tumor cells frequently encounter severe stressors, such as low oxygen levels and nutritional deprivation, which causes them to activate autophagy as a survival tactic. This process allows cancer stem cells (CSCs) to withstand severe conditions while also preserving their features. After years of dormancy, dormant disseminated tumor cells (DTCs) may reappear as aggressive metastatic cancers. The capacity of autophagy to promote resistance to treatments and avoid immune detection is intimately related to this phenomenon. According to recent research, autophagy promotes processes, such as the epithelial-to-mesenchymal transition (EMT) and helps build a pre-metastatic niche, which makes treatment strategies more challenging. Autophagy may be a promising therapeutic target because of its dual function as a tumor suppressor in early-stage cancer and a survival promoter in advanced stages. To effectively treat metastatic diseases, it is crucial to comprehend how metabolic processes interact with autophagy and affect tumor behavior. In order to find novel therapeutic approaches that can interfere with these processes and improve patient outcomes, this study highlights the critical need for additional investigation into the mechanisms by which autophagy controls tumor dormancy and recurrence.
Collapse
Affiliation(s)
- Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | - Dorrin Mansourian
- Faculty of Pharmacy, Eastern Mediterranean University, Gazimagusa TRNC via Mersin 10, Mersin, Turkey
| | | | - Zeinab Fathi
- Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
Lim BJW, Liu M, Wang L, Kong SLY, Yin T, Yan C, Xiang K, Cao C, Wu H, Mihai A, Tay FPL, Wang E, Jiang Q, Ma Z, Tan L, Chia RN, Qin D, Pan CC, Wang XF, Li QJ. Neoadjuvant anti-4-1BB confers protection against spontaneous metastasis through low-affinity intratumor CD8 + T cells in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635356. [PMID: 39975187 PMCID: PMC11838326 DOI: 10.1101/2025.01.29.635356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neoadjuvant immunotherapy seeks to harness the primary tumor as a source of relevant tumor antigens to enhance systemic anti-tumor immunity through improved immunological surveillance. Despite having revolutionized the treatment of patients with high-risk early-stage triple-negative breast cancer (TNBC), a significant portion of patients remain unresponsive and succumb to metastatic recurrence post-treatment. Here, we found that optimally scheduled neoadjuvant administration of anti-4-1BB monotherapy was able to counteract metastases and prolong survival following surgical resection. Phenotypic and transcriptional profiling revealed enhanced 4-1BB expression on tumor-infiltrating intermediate (T int ), relative to progenitor (T prog ) and terminally exhausted (T term ) T cells. Furthermore, T int was enriched in low-affinity T cells. Treatment with anti-4-1BB drove clonal expansion of T int , with reduced expression of tissue-retention marker CD103 in T prog . This was accompanied by increased TCR clonotype sharing between paired tumors and pre-metastatic lungs. Further interrogation of sorted intratumor T cells confirmed enhanced T cell egress into circulation following anti-4-1BB treatment. In addition, gene signature extracted from anti-4-1BB treated T int was consistently associated with improved clinical outcomes in BRCA patients. Combinatorial neoadjuvant anti-4-1BB and ablation of tumor-derived CXCL16 resulted in enhanced therapeutic effect. These findings illustrate the intratumor changes underpinning the efficacy of neoadjuvant anti-4-1BB, highlighting the reciprocity between local tissue-retention and distant immunologic fortification, suggesting treatment can reverse the siphoning of intratumor T cells to primary tumor, enabling redistribution to distant tissues and subsequent protection against metastases.
Collapse
|
20
|
Chen Q, Yang M, Duan X, Zhang J, Shi F, Chen R, Li Y. Linker Histone H1.4 Inhibits the Growth, Migration and EMT Process of Non-Small Cell Lung Cancer by Regulating ERK1/2 Expression. Biochem Genet 2025; 63:576-591. [PMID: 38472566 DOI: 10.1007/s10528-024-10760-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
H1.4 is one of the 11 variants of linker histone H1, and is associated with tumorigenesis and development of various cancers. However, it is unclear for the role of histone H1.4 in non-small cell lung cancer (NSCLC). In this study, we found that overexpression of H1.4 significantly inhibited the cell viability, migration, invasion and epithelial-mesenchymal transition (EMT) processes, whereas silencing H1.4 by shRNA knockdown promoted these processes in NSCLC cell lines A549 and H1299. We further showed that H1.4 overexpression reduced ERK1/2 expression or its phosphorylation levels, while H1.4 knockdown increased ERK1/2 expression or phosphorylation levels in NSCLC. Furthermore, we demonstrated that H1.4 bound to the promoter of ERK1/2, and acted as a transcriptional suppressor to inhibit ERK1/2 expression in A549 or H1299 cells. Importantly, we found that ERK ecto-expression can largely recovered the inhibitory effects of H1.4 on cell viability, migration, invasion and EMT processes. In summary, our study reveals that the H1.4-ERK pathway is crucial for cell viability, migration, invasion and EMT of NSCLC and could be a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Qian Chen
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Mengqi Yang
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Xinyue Duan
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Jie Zhang
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Fan Shi
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Rong Chen
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China
| | - Yong Li
- School of Life Sciences, Anhui University, Hefei, Anhui Province, 230601, PR China.
- Center for Stem Cell and Translational Medicine, Anhui University, Hefei, Anhui Province, China.
| |
Collapse
|
21
|
Krishnamurthy M, Dhall A, Sahoo S, Schultz CW, Baird MA, Desai P, Odell J, Takahashi N, Nirula M, Zhuang S, Huang Y, Schroeder B, Zhang Y, Thomas MS, Redon C, Robinson C, Thang L, Ileva L, Patel NL, Kalen JD, Varlet AA, Zuela-Sopilniak N, Jha A, Wangsa D, Butcher D, Morgan T, Afzal AN, Chari R, Baktiar K, Kumar S, Pongor L, Difilippantonio S, Aladjem MI, Pommier Y, Jolly MK, Lammerding J, Sharma AK, Thomas A. Metastatic organotropism in small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.07.617066. [PMID: 39416100 PMCID: PMC11483079 DOI: 10.1101/2024.10.07.617066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Metastasis is the leading cause of cancer-related deaths, yet its regulatory mechanisms are not fully understood. Small-cell lung cancer (SCLC) is the most metastatic form of lung cancer, with most patients presenting with widespread disease, making it an ideal model for studying metastasis. However, the lack of suitable preclinical models has limited such studies. We utilized rapid autopsy-derived tumors to develop xenograft models that mimic key features of SCLC, including histopathology, rapid and widespread development of metastasis to the liver, brain, adrenal, bone marrow, and kidneys within weeks, and response to chemotherapy. By integrating in vivo lineage selection with comprehensive bulk and single cell multiomic profiling of transcriptomes and chromatin accessibility, we identified critical cellular programs driving metastatic organotropism to the liver and brain, the most common sites of SCLC metastasis. Our findings reveal the key role of nuclear-cytoskeletal interactions in SCLC liver metastasis. Specifically, the loss of the nuclear envelope protein lamin A/C, encoded by the LMNA gene, increased nuclear deformability and significantly increased the incidence of liver metastasis. Human liver metastases exhibited reduced LMNA expression compared to other metastatic sites, correlating with poorer patient outcomes and increased mortality. This study introduces novel preclinical models for SCLC metastasis and highlights pathways critical for organ-specific metastasis, offering new avenues for the development of targeted therapies to prevent or treat metastatic disease.
Collapse
Affiliation(s)
- Manan Krishnamurthy
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anjali Dhall
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science, Bangalore, India
| | - Christopher W. Schultz
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michelle A. Baird
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health; Bethesda, USA
| | - Parth Desai
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Hematology & Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Jacob Odell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Nobuyuki Takahashi
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Medical Oncology, National Cancer Center East Hospital, Kashiwa, Japan
| | - Michael Nirula
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sophie Zhuang
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yue Huang
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Brett Schroeder
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yang Zhang
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maria Sebastian Thomas
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christophe Redon
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christina Robinson
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701
| | - Lai Thang
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701
| | - Lilia Ileva
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Nimit L. Patel
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Joseph D. Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Alice-Anaïs Varlet
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Noam Zuela-Sopilniak
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ankita Jha
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health; Bethesda, USA
| | - Darawalee Wangsa
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Donna Butcher
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tamara Morgan
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Alyah N. Afzal
- Laboratory Animal Sciences Program, Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Raj Chari
- Laboratory Animal Sciences Program, Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Karim Baktiar
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Suresh Kumar
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lorinc Pongor
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Cancer Genomics and Epigenetics Core Group, Szeged, Hungary
| | - Simone Difilippantonio
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701
| | - Mirit I. Aladjem
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore, India
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ajit Kumar Sharma
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Anish Thomas
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
22
|
Yu Y, Zhu C, Wang X, Shi Y, Gao Y, Yu Z. hERG activators exhibit antitumor effects in breast cancer through calcineurin and β-catenin-mediated signaling pathways. Front Pharmacol 2025; 16:1545300. [PMID: 39917621 PMCID: PMC11799564 DOI: 10.3389/fphar.2025.1545300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Background Breast cancer remains a leading cause of mortality among women worldwide, with existing therapeutic options often accompanied by significant side effects and a persistent risk of disease recurrence. This highlights the need for novel drug candidates with new mechanisms of action by targeting alternative signaling pathways. While hERG channel is notoriously regarded as an off-target due to drug-induced cardiotoxicity, its therapeutic potential as a drug target remains largely unexplored. Methods This study investigated the role of hERG in breast cancer progression and its impact on patient survival. The anti-proliferative, anti-migratory, anti-invasive and pro-apoptotic effects of hERG activators were evaluated using the Cell Counting Kit-8, wound healing assay, transwell assay and cell apoptosis assay, respectively. Western blotting, Ca2+ imaging and immunofluorescence assays were employed to study their antitumor mechanisms of actions. Results We identified two novel hERG activators, SDUY429 and SDUY436, which effectively inhibited the proliferation and migration of MDA-MB-231 and MCF-7 cells. In addition, SDUY436 demonstrated significant anti-invasive and pro-apoptotic effects in MDA-MB-231 cells. Mechanistically, the anti-proliferative activity of hERG activators were mediated through calcineurin activation via enhanced calcium ion influx, which facilitated the nuclear translocation of nuclear factor of activated T cells (NFAT) and upregulated p21Waf/Cip expression. Furthermore, both SDUY429 and SDUY436 remarkably suppressed the migration and invasion of MDA-MB-231 cells by downregulating the protein kinase B (AKT)/glycogen synthase kinase-3 beta (GSK3β)/β-catenin signaling pathway. The observed reduction in phospho-AKT-Ser473 (pAKTS473) expression resulted in the decreased levels of phospho-GSK3β-Ser9 (pGSK3βS9), thereby limiting the nuclear localization of β-catenin, which led to the inhibition of cell migration and invasion. Notably, combining SDUY429 or SDUY436 with the AKT inhibitor MK-2206 produced synergistic anti-proliferative effects. Conclusion These findings suggest that hERG activators hold promise as new potential therapeutic agents for the treatment of breast cancer, paving the way for future investigations into their clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhiyi Yu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
23
|
Wang W, Lokman NA, Barry SC, Oehler MK, Ricciardelli C. LGR5: An emerging therapeutic target for cancer metastasis and chemotherapy resistance. Cancer Metastasis Rev 2025; 44:23. [PMID: 39821694 PMCID: PMC11742290 DOI: 10.1007/s10555-024-10239-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
Cancer stem cells play an important role in tumor progression and chemotherapy resistance. Leucine-rich G repeat-containing protein-coupled receptor 5 (LGR5) has been identified as a cancer stem cell marker in several cancer types. LGR5 is involved in cancer development and progression via several pathways including WNT/β-catenin signaling pathway. LGR5 plays a role in tumor progression by promoting cancer cell migration, invasion, metastasis, and angiogenesis in many cancers including colorectal, brain, gastric, and ovarian cancer. This review summarises the current knowledge on the expression and functional role of LGR5 in cancers, the molecular mechanisms regulated by LGR5, and the relationship between LGR5 and chemotherapy resistance. The review also includes highlights potential strategies to inhibit LGR5 expression and function. The majority of functional studies have shown that LGR5 plays an important role in promoting cancer progression, metastasis and chemotherapy resistance however, in some contexts LGR5 can also activate tumor-suppressive pathways and LGR5 negative cells can also promote cancer progression. The review highlights that targeting LGR5 is a promising anti-cancer treatment but the functional effect of LGR5 on tumor cells is complex may be dependent on cancer type, tumor microenvironment and cross-talk with other molecules in the LGR5 signaling pathway.
Collapse
Affiliation(s)
- Wanqi Wang
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
| | - Simon C Barry
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, 5005, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, 5000, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia.
| |
Collapse
|
24
|
Ding Y, Chen Y, Xie S, Qiu Q, Guo X, Feng Y, Li H, Zhu F, Liu Y. Chemokine family significance and prognostic value in colorectal cancer. Front Immunol 2025; 15:1404768. [PMID: 39835121 PMCID: PMC11743568 DOI: 10.3389/fimmu.2024.1404768] [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: 03/21/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Background Colorectal cancer (CRC) poses a substantial global health concern, exhibits inconspicuous early symptoms, and is typically diagnosed at advanced stages leading to unfavorable outcomes. The intricate tumor microenvironment plays a crucial role in CRC development and progression, where chemokines contribute significantly. These chemokines exhibit widespread expression within tumor cells, facilitating immune cell infiltration, angiogenesis, and the establishment of distant metastases. The dysregulation of various chemokines in the context of CRC has emerged as a pivotal factor in the disease's pathogenesis. Methods To explore the relationship between chemokine gene expression and CRC patient survival, as well as to clarify their biological roles,We conducted RNA-sequencing (RNA-seq) analysis on a cohort of 88 CRC patients with tumor samples, thereby enabling a detailed exploration of chemokine involvement in CRC. This study was rigorously augmented using comprehensive datasets from The Cancer Genome Atlas (TCGA), ensuring a robust analysis of gene expression patterns associated with clinical outcomes. Results Through data analysis, we identified key genes from the chemokine family thought pertinent to CRC outcomes. Consequently, we constructed a novel prognostic model based on the risk score derived from these chemokine expressions. Validation against clinical metadata, executed through immunohistochemistry analysis, affirmed the relevance and accuracy of our model in predicting patient survival. Conclusion Our findings illuminate the critical role of chemokines in shaping the immune microenvironment of CRC, thereby highlighting potential therapeutic targets for future treatment strategies. Our new prognostic model could provide important information for the development of targeted therapies for CRC, enhancing personalized treatment approaches andultimately improving survival for CRC patients.
Collapse
Affiliation(s)
- Yi Ding
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yinnan Chen
- Department of High Talent, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Hubei Province Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Siyun Xie
- Information Science and Technology, Northwest University, Xi’an, Shaanxi, China
| | - Quanpeng Qiu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaolong Guo
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yun Feng
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Clinical Medical Research Center for Digestive Diseases of Shaanxi Province (Oncology), The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hongxia Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Clinical Medical Research Center for Digestive Diseases of Shaanxi Province (Oncology), The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Fang Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaping Liu
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of High Talent, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Clinical Medical Research Center for Digestive Diseases of Shaanxi Province (Oncology), The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
25
|
Mao C, Zhang J, Yang C, Mei L, Feng Y, Dai F, Huang Y, Xiao H, Deng B. BCAR1 facilitates the survival of lung adenocarcinoma cells by augmenting the unfolded protein response, autophagy, and the formation of vasculogenic mimicry. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167558. [PMID: 39488300 DOI: 10.1016/j.bbadis.2024.167558] [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: 03/29/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Our objective was to elucidate the pivotal roles of BCAR1 in unfolded protein response (UPR), autophagy and vasculogenic mimicry (VM) formation, processes that essential for the metastasis of lung adenocarcinoma (LUAD) cells. METHODS The morphological assessment of endoplasmic reticulum (ER) status and autolysosomes in H1975 and H1299 LUAD cells following BCAR1 knockout (KO) was conducted using transmission electron microscope. The expression of markers and cellular functions related to the UPR, autophagy, and VM formation were examined in LUAD cells tissues. Additionally, proteomic analysis of LUAD cells was performed via mass spectrometry, and the pertinent signaling pathways were analyzed using bioinformatics tools. RESULTS BCAR1-KO inhibited autophagy and UPR induced triggered starvation in LUAD cells. Cleaved-ATF6a-mediated UPR and subsequent autophagy, enhanced by BCAR1, were confirmed using the UPR stimulator and blocker. High BCAR1 expression, along with elevated UPR and autophagy, predicts poor prognosis in LUAD patients. BCAR1-KO reduced tube formation and VM markers expressions in LUAD cells. Additionally, BCAR1 expression positively correlated with VM formation in BALB/c-nu mice xenografts and LUAD patient tissues. CONCLUSION BCAR1 promotes LUAD metastasis by enhancing cancer cell survival in nutrient-poor environments through ATF6-mediated UPR activation and autophagy. As BCAR1 induces VM formation, metastatic lesions eventually colonize. Thus, BCAR1 is a promising anti-metastasis target.
Collapse
Affiliation(s)
- Chengyi Mao
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jingge Zhang
- Thoracic Surgery Department, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Chuan Yang
- Thoracic Surgery Department, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Longyong Mei
- Thoracic Surgery Department, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yonggeng Feng
- Thoracic Surgery Department, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Fuqiang Dai
- Thoracic Surgery Department, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yi Huang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China.
| | - Hualiang Xiao
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing 400042, China.
| | - Bo Deng
- Thoracic Surgery Department, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
26
|
Kabak EC, Foo SL, Rafaeva M, Martin I, Bentires-Alj M. Microenvironmental Regulation of Dormancy in Breast Cancer Metastasis: "An Ally that Changes Allegiances". ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:373-395. [PMID: 39821034 DOI: 10.1007/978-3-031-70875-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Breast cancer remission after treatment is sometimes long-lasting, but in about 30% of cases, there is a relapse after a so-called dormant state. Cellular cancer dormancy, the propensity of disseminated tumor cells (DTCs) to remain in a nonproliferative state for an extended period, presents an opportunity for therapeutic intervention that may prevent reawakening and the lethal consequences of metastatic outgrowth. Therefore, identification of dormant DTCs and detailed characterization of cancer cell-intrinsic and niche-specific [i.e., tumor microenvironment (TME) mediated] mechanisms influencing dormancy in different metastatic organs are of great importance in breast cancer. Several microenvironmental drivers of DTC dormancy in metastatic organs, such as the lung, bone, liver, and brain, have been identified using in vivo models and/or in vitro three-dimensional culture systems. TME induction and persistence of dormancy in these organs are mainly mediated by signals from immune cells, stromal cells, and extracellular matrix components of the TME. Alterations of the TME have been shown to reawaken dormant DTCs. Efforts to capitalize on these findings often face translational challenges due to limited availability of representative patient samples and difficulty in designing dormancy-targeting clinical trials. In this chapter, we discuss current approaches to identify dormant DTCs and provide insights into cell-extrinsic (i.e., TME) mechanisms driving breast cancer cell dormancy in distant organs.
Collapse
Affiliation(s)
- Evrim Ceren Kabak
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sok Lin Foo
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Maria Rafaeva
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
27
|
Shen JJ, Yang X, Yu M, Li QC, Wang RY, Yu WY, Zhang JL, Chen YL, Zhu WT, Li J, Zhan ZJ, Wu R. Discovery of aurovertin B as a potent metastasis inhibitor against triple-negative breast cancer: Elucidating the complex role of the ATF3-DUSP1 axis. J Pharmacol Exp Ther 2025; 392:100005. [PMID: 39893015 DOI: 10.1124/jpet.124.002264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by high mortality rates, primarily due to its propensity for metastasis. Addressing this challenge necessitates the development of effective antimetastatic therapies. This study aimed to identify natural compounds with potential antimetastatic properties mainly based on the high-throughput phenotypic screening system. This system, utilizing luciferase reporter gene assays combined with scratch wound assays, evaluates compounds based on their influence on the epithelial-mesenchymal transition (EMT) marker E-cadherin. Through this approach, aurovertin B (AVB) was revealed to have significant antimetastatic capability. Notably, AVB exhibited substantial metastasis suppression in many TNBC cell lines, including MDA-MB-231, HCC1937, and 4T1. Also, its remarkable antimetastatic activity was demonstrated in vivo via the orthotopic breast cancer mouse model. Further exploration revealed a pronounced association between AVB-induced upregulation of dual-specificity phosphatase 1 (DUSP1) and its inhibitory effect on TNBC metastasis. Additionally, microarray analysis conducted to elucidate the underlying mechanism of the AVB-DUSP1 interaction identified activating transcription factor 3 (ATF3) as a critical transcription factor instrumental in DUSP1 transcriptional activation. This discovery, coupled with observations of enhanced ATF3-DUSP1 expression and consequent reduction in TNBC metastatic foci in response to AVB, provides novel insights into the metastatic mechanisms of TNBC. SIGNIFICANCE STATEMENT: This study constructs a high-throughput phenotypic screening system utilizing epithelial-mesenchymal transition marker E-cadherin promoter luciferase reporter gene combined with scratch wound assays. Aurovertin B was revealed to possess significant antimetastatic activity through this approach, which was further demonstrated via in vivo and in vitro experiments. The discovery of the regulatory role of the ATF3-DUSP1 pathway enriches our understanding of TNBC metastasis mechanism and suggests the potential of ATF3 and DUSP1 as biomarkers for diagnosing TNBC metastasis.
Collapse
Affiliation(s)
- Jian-Jun Shen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Xi Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Meng Yu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Qing-Cui Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Ru-Yu Wang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Wen-Yan Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jia-Li Zhang
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Li Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Wen-Ting Zhu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Jia Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Zha-Jun Zhan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| | - Rui Wu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
28
|
Basmaeil Y, Subayyil AA, Kulayb HB, Kondkar AA, Alrodayyan M, Khatlani T. Partial Inhibition of Epithelial-to-Mesenchymal Transition (EMT) Phenotypes by Placenta-Derived DBMSCs in Human Breast Cancer Cell Lines, In Vitro. Cells 2024; 13:2131. [PMID: 39768220 PMCID: PMC11674051 DOI: 10.3390/cells13242131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Stem cell-based therapies hold significant potential for cancer treatment due to their unique properties, including migration toward tumor niche, secretion of bioactive molecules, and immunosuppression. Mesenchymal stem cells (MSCs) from adult tissues can inhibit tumor progression, angiogenesis, and apoptosis of cancer cells. We have previously reported the isolation and characterization of placenta-derived decidua basalis mesenchymal stem cells (DBMSCs), which demonstrated higher levels of pro-migratory and anti-apoptotic genes, indicating potential anti-cancer effects. In this study, we analyzed the anti-cancer effects of DBMSCs on human breast cancer cell lines MDA231 and MCF7, with MCF 10A used as control. We also investigated how these cancer cells lines affect the functional competence of DBMSCs. By co-culturing DBMSCs with cancer cells, we analyzed changes in functions of both cell types, as well as alterations in their genomic and proteomic profile. Our results showed that treatment with DBMSCs significantly reduced the functionality of MDA231 and MCF7 cells, while MCF 10A cells remained unaffected. DBMSC treatment decreased epithelial-to-mesenchymal transition (EMT)-related protein levels in MDA231 cells and modulated expression of other cancer-related genes in MDA231 and MCF7 cells. Although cancer cells reduced DBMSC proliferation, they increased their expression of anti-apoptotic genes. These findings suggest that DBMSCs can inhibit EMT-related proteins and reduce the invasive characteristics of MDA231 and MCF7 breast cancer cells, highlighting their potential as candidates for cell-based cancer therapies.
Collapse
Affiliation(s)
- Yasser Basmaeil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Abdullah Al Subayyil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Haya Bin Kulayb
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia;
| | - Maha Alrodayyan
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| |
Collapse
|
29
|
Wu Y, Shang J, Zhang X, Li N. Advances in molecular imaging and targeted therapeutics for lymph node metastasis in cancer: a comprehensive review. J Nanobiotechnology 2024; 22:783. [PMID: 39702277 PMCID: PMC11657939 DOI: 10.1186/s12951-024-02940-4] [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: 01/30/2024] [Accepted: 10/19/2024] [Indexed: 12/21/2024] Open
Abstract
Lymph node metastasis is a critical indicator of cancer progression, profoundly affecting diagnosis, staging, and treatment decisions. This review article delves into the recent advancements in molecular imaging techniques for lymph nodes, which are pivotal for the early detection and staging of cancer. It provides detailed insights into how these techniques are used to visualize and quantify metastatic cancer cells, resident immune cells, and other molecular markers within lymph nodes. Furthermore, the review highlights the development of innovative, lymph node-targeted therapeutic strategies, which represent a significant shift towards more precise and effective cancer treatments. By examining cutting-edge research and emerging technologies, this review offers a comprehensive overview of the current and potential impact of lymph node-centric approaches on cancer diagnosis, staging, and therapy. Through its exploration of these topics, the review aims to illuminate the increasingly sophisticated landscape of cancer management strategies focused on lymph node assessment and intervention.
Collapse
Affiliation(s)
- Yunhao Wu
- Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jin Shang
- Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinyue Zhang
- The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Nu Li
- The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
30
|
Wang W, Kang Y, Qu X, Li Y, Zhou H. A rare case report of renal clear cell carcinoma with multiple skin metastases and a review of the literature. Front Oncol 2024; 14:1461791. [PMID: 39697235 PMCID: PMC11652365 DOI: 10.3389/fonc.2024.1461791] [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: 08/09/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Renal cell carcinoma is the most common type of primary renal cancer, and clear cell carcinoma is the most common subtype, accounting for approximately 70% of all adult renal cell carcinoma cases. At the time of diagnosis, many patients already have metastatic manifestations. Cutaneous metastasis of renal clear cell carcinoma is very rare and usually represents a poor prognosis, mostly affecting the head and neck. In this paper, we report a case of renal clear cell carcinoma with multiple cutaneous metastases, including a chest wall mass for more than 10 years and an abdominal wall mass for 1 year. A 69-year-old man with a history of diabetes mellitus was admitted to the hospital for examination of a right chest and abdominal wall mass and peripheral pain, and ultrasonography suggested a solid space-occupying lesion in the left kidney, which was considered malignant, and a solid mass in the right chest and abdominal wall, which was considered metastatic. A subsequent abdominal CT scan showed malignant tumors in the left kidney and adrenal region, and multiple metastatic tumors in the liver, pancreas, right thoracoabdominal wall, and the abdomen. To clarify the nature of the pathology, an ultrasound-guided puncture of the right abdominal wall mass was performed, and the pathological diagnosis was clear cell carcinoma, with immunohistochemistry suggesting a renal clear cell carcinoma origin. The patient died within 6 months.
Collapse
Affiliation(s)
- Wei Wang
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yongcun Kang
- Department of Radiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaona Qu
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Li
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hongyan Zhou
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
31
|
L Rocha H, Aguilar B, Getz M, Shmulevich I, Macklin P. A multiscale model of immune surveillance in micrometastases gives insights on cancer patient digital twins. NPJ Syst Biol Appl 2024; 10:144. [PMID: 39627216 PMCID: PMC11614875 DOI: 10.1038/s41540-024-00472-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024] Open
Abstract
Metastasis is the leading cause of death in patients with cancer, driving considerable scientific and clinical interest in immunosurveillance of micrometastases. We investigated this process by creating a multiscale mathematical model to study the interactions between the immune system and the progression of micrometastases in general epithelial tissue. We analyzed the parameter space of the model using high-throughput computing resources to generate over 100,000 virtual patient trajectories. We demonstrated that the model could recapitulate a wide variety of virtual patient trajectories, including uncontrolled growth, partial response, and complete immune response to tumor growth. We classified the virtual patients and identified key patient parameters with the greatest effect on the simulated immunosurveillance. We highlight the lessons derived from this analysis and their impact on the nascent field of cancer patient digital twins (CPDTs). While CPDTs could enable clinicians to systematically dissect the complexity of cancer in each individual patient and inform treatment choices, our work shows that key challenges remain before we can reach this vision. In particular, we show that there remain considerable uncertainties in immune responses, unreliable patient stratification, and unpredictable personalized treatment. Nonetheless, we also show that in spite of these challenges, patient-specific models suggest strategies to increase control of clinically undetectable micrometastases even without complete parameter certainty.
Collapse
Affiliation(s)
- Heber L Rocha
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | | | - Michael Getz
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | | | - Paul Macklin
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
32
|
Yamaguchi N, Wu YG, Ravetch E, Takahashi M, Khan AG, Hayashi A, Mei W, Hsu D, Umeda S, de Stanchina E, Lorenz IC, Iacobuzio-Donahue CA, Tavazoie SF. A Targetable Secreted Neural Protein Drives Pancreatic Cancer Metastatic Colonization and HIF1α Nuclear Retention. Cancer Discov 2024; 14:2489-2508. [PMID: 39028915 PMCID: PMC11611693 DOI: 10.1158/2159-8290.cd-23-1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 07/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an increasingly diagnosed cancer that kills 90% of afflicted patients, with most patients receiving palliative chemotherapy. We identified neuronal pentraxin 1 (NPTX1) as a cancer-secreted protein that becomes overexpressed in human and murine PDAC cells during metastatic progression and identified adhesion molecule with Ig-like domain 2 (AMIGO2) as its receptor. Molecular, genetic, biochemical, and pharmacologic experiments revealed that secreted NPTX1 acts cell-autonomously on the AMIGO2 receptor to drive PDAC metastatic colonization of the liver-the primary site of PDAC metastasis. NPTX1-AMIGO2 signaling enhanced hypoxic growth and was critically required for hypoxia-inducible factor-1α (HIF1α) nuclear retention and function. NPTX1 is overexpressed in human PDAC tumors and upregulated in liver metastases. Therapeutic targeting of NPTX1 with a high-affinity monoclonal antibody substantially reduced PDAC liver metastatic colonization. We thus identify NPTX1-AMIGO2 as druggable critical upstream regulators of the HIF1α hypoxic response in PDAC. Significance: We identified the NPTX1-AMIGO2 axis as a regulatory mechanism upstream of HIF1α-driven hypoxia response that promotes PDAC liver metastasis. Therapeutic NPTX1 targeting outperformed a common chemotherapy regimen in inhibiting liver metastasis and suppressed primary tumor growth in preclinical models, revealing a novel therapeutic strategy targeting hypoxic response in PDAC.
Collapse
Affiliation(s)
- Norihiro Yamaguchi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Y Gloria Wu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Ethan Ravetch
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Mai Takahashi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Abdul G. Khan
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - Akimasa Hayashi
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wenbin Mei
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Dennis Hsu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Shigeaki Umeda
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ivo C. Lorenz
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | | | - Sohail F. Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
33
|
Liu C, Chen S, Zhang Y, Zhou X, Wang H, Wang Q, Lan X. Mechanisms of Rho GTPases in regulating tumor proliferation, migration and invasion. Cytokine Growth Factor Rev 2024; 80:168-174. [PMID: 39317522 DOI: 10.1016/j.cytogfr.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
The occurrence of most cancers is due to the clonal proliferation of tumor cells, immune evasion, and the ability to spread to other body parts. Rho GTPases, a family of small GTPases, are key regulators of cytoskeleton reorganization and cell polarity. Additionally, Rho GTPases are key proteins that induce the proliferation and metastasis of tumor cells. This review focuses on the complex regulatory mechanisms of Rho GTPases, exploring their critical role in promoting tumor cell proliferation and dissemination. Regarding tumor cell proliferation, attention is given to the role of Rho GTPases in regulating the cell cycle and mitosis. In terms of tumor cell dissemination, the focus is on the role of Rho GTPases in regulating cell migration and invasion. Overall, this review elucidates the mechanisms of Rho GTPases members in the development of tumor cells, aiming to provide theoretical references for the treatment of mammalian tumor diseases and related applications.
Collapse
Affiliation(s)
- Cheng Liu
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Shutao Chen
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Yu Zhang
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Xinyi Zhou
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Haiwei Wang
- Chongqing Academy Of Animal Sciences, Chongqing 402460, China.
| | - Qigui Wang
- Chongqing Academy Of Animal Sciences, Chongqing 402460, China.
| | - Xi Lan
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
34
|
Lu YT, Lin CW, Su SC, Ho YT, Yeh FL, Hsin CH, Yang SF. L48H37, a curcumin analog, suppresses matrix metalloproteinase-9 expression and activity to hamper nasopharyngeal cancer cell migration. Oral Oncol 2024; 159:107038. [PMID: 39284263 DOI: 10.1016/j.oraloncology.2024.107038] [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: 07/18/2024] [Revised: 08/21/2024] [Accepted: 09/10/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE Metastatic disease is a major issue of treatment failure in nasopharyngeal carcinoma (NPC) patients and often linked to high mortality. L48H37, a synthetic analog of curcumin with augmented bioavailability over its parent compound, has demonstrated several oncostatic characteristics. This study was aimed to explore the anti-metastatic effect of L48H37 on NPC cancer cells and its underlying mechanism. METHODS Cell viability was evaluated using MTT assay. Regulation of signaling pathways was elucidated by immunoblotting, and specific kinase inhibitors. RESULTS In this study, we showed that L48H37 suppressed TPA-stimulated invasive and migratory capacities of NPC cell lines and gave rise to very little cytotoxic responses. Such anti-cancer effect of L48H37 was accompanied with attenuated expression levels and enzymatic activities of matrix metalloproteinase-9 (MMP-9), a pivotal mediator of metastatic processes. In addition, L48H37 interfered with TPA-induced JNK activation, and the treatment of L48H37 combined with a JNK antagonist demonstrated a synergistic effect on restraining TPA-stimulated MMP-9 activity and migration events in NPC cells. CONCLUSIONS Our results revealed that L48H37 impeded the invasive potential of NPC cells via impairment of MMP-9 function and abundance, highlighting possible complementary therapies using curcumin or its effective analogs to manage NPC dissemination.
Collapse
Affiliation(s)
- Yen-Ting Lu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan; Department of Otolaryngology, St. Martin De Porres Hospital, Chiayi, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Ting Ho
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Fang-Ling Yeh
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA
| | - Chung-Han Hsin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
35
|
Guruvayurappan GK, Frankenbach-Désor T, Laubach M, Klein A, von Bergwelt-Baildon M, Cusan M, Aszodi A, Holzapfel BM, Böcker W, Mayer-Wagner S. Clinical challenges in prostate cancer management: Metastatic bone-tropism and the role of circulating tumor cells. Cancer Lett 2024; 606:217310. [PMID: 39486571 DOI: 10.1016/j.canlet.2024.217310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Prostate cancer (PCa) metastasis is one of the leading causes of cancer-related mortality in men worldwide, primarily due to its tendency to metastasize, with bones of axial skeleton being the favored target-site. PCa bone-metastasis (PCa-BM) presents significant clinical challenges, especially by the weakening of bone architecture, majorly due to the formation of osteoblastic lesions, leading to severe bone fractures. Another complication is that the disease predominantly affects elderly men. Further exploration is required to understand how the circulating tumor cells (CTCs) adapt to varying microenvironments and other biomechanical stresses encountered during the sequential steps in metastasis, finally resulting in colonization specifically in the bone niche, in PCa-BM. Deciphering how CTCs encounter and adapt to different biochemical, biomechanical and microenvironmental factors may improve the prospects of PCa diagnosis, development of novel therapeutics and prognosis. Moreover, the knowledge developed is expected to have broader implications for cancer research, paving the way for better therapeutic strategies and targeted therapies in the realm of metastatic cancer progression across different types of cancers. Our review begins with analyzing the challenges in PCa diagnosis, treatment and management, and delves into the formation and dynamics of CTCs, highlighting their role in PCa metastasis and bone-tropism. We further explore the pivotal role of individual factors in dictating the predisposition of tumors to metastasize to specific secondary sites, such as the noteworthy tendency of PCa bone-metastasis. Finally, we highlight the unresolved questions and potential avenues for further exploration.
Collapse
Affiliation(s)
- Gayathri K Guruvayurappan
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Tina Frankenbach-Désor
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Markus Laubach
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexander Klein
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Monica Cusan
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Attila Aszodi
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris M Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Böcker
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Susanne Mayer-Wagner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
36
|
Li Z, Tan W, Li X, Wang Y, Dang Z, Zhang Z, Guan S, Zhu S, Li F, Zhang M. Unlocking lysosomal acidity to activate membranolytic module for accurately cancer theranostics. Bioorg Chem 2024; 153:107764. [PMID: 39232344 DOI: 10.1016/j.bioorg.2024.107764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Chemotherapy drug efflux, toxic side effects, and low efficacy against drug-resistant cells have plagued safe and efficient cancer theranostics. However, the materials or methods that resolve these defects all-in-one are scarce. Here, a new cancer theranostics strategy is proposed by utilizing changes in lysosomal acidity in cancer cells to activate the membranolytic model to overcome these obstacles together. Therefore, a simple fluorescent anthracene derivative Lyso-Mito is developed, which has a perfect pKa (4.62) value that falls between the pH of lysosomes in cancer and normal cells. Lyso-Mito itself can precisely target and convert the pH perturbation of lysosomes in cancer cells to fluorescent response and membranolytic module activity to accomplish the low drug efflux, weak toxic side effects, and low drug-resistant cancer diagnosis and treatment without linking other functional units or any additional assistance. Hereby, a new cancer theranostics strategy of integrating organelle microenvironment and the membranolytic model is realized.
Collapse
Affiliation(s)
- Zhuo Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wenjia Tan
- China-Japan Union Hospital of Jilin University, Changchun 130041, China
| | - Xinru Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - YaJun Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zetao Dang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhaoxia Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Shuwen Guan
- College of Life Science, Jilin University, Changchun 130012, China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Feng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Ming Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| |
Collapse
|
37
|
Cantoni F, Barbe L, Roy A, Wicher G, Simonsson S, Forsberg-Nilsson K, Tenje M. On-chip fabrication of tailored 3D hydrogel scaffolds to model cancer cell invasion and interaction with endothelial cells. APL Bioeng 2024; 8:046113. [PMID: 39634677 PMCID: PMC11617029 DOI: 10.1063/5.0227135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/09/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
The high mortality associated with certain cancers can be attributed to the invasive nature of the tumor cells. Yet, the complexity of studying invasion hinders our understanding of how the tumor spreads. This work presents a microengineered three-dimensional (3D) in vitro model for studying cancer cell invasion and interaction with endothelial cells. The model was generated by printing a biomimetic hydrogel scaffold directly on a chip using 2-photon polymerization that simulates the brain's extracellular matrix. The scaffold's geometry was specifically designed to facilitate the growth of a continuous layer of endothelial cells on one side, while also allowing for the introduction of tumor cells on the other side. This arrangement confines the cells spatially and enables in situ microscopy of the cancer cells as they invade the hydrogel scaffold and interact with the endothelial layer. We examined the impact of 3D printing parameters on the hydrogel's physical properties and used patient derived glioblastoma cells to study their effect on cell invasion. Notably, the tumor cells tended to infiltrate faster when an endothelial cell barrier was present. The potential for adjusting the hydrogel scaffold's properties, coupled with the capability for real-time observation of tumor-endothelial cell interactions, offers a platform for studying tumor invasion and tumor-endothelial cell interactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maria Tenje
- Author to whom correspondence should be addressed:
| |
Collapse
|
38
|
Fan L, Lin Y, Fu Y, Wang J. Small cell lung cancer with liver metastases: from underlying mechanisms to treatment strategies. Cancer Metastasis Rev 2024; 44:5. [PMID: 39585433 DOI: 10.1007/s10555-024-10220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Small cell lung cancer (SCLC) represents an aggressive neuroendocrine (NE) tumor within the pulmonary region, characterized by very poor prognoses. Druggable targets for SCLC remain limited, thereby constraining treatment options available to patients. Immuno-chemotherapy has emerged as a pivotal therapeutic strategy for extensive-stage SCLC (ES-SCLC), yet it fails to confer significant efficacy in cases involving liver metastases (LMs) originating from SCLC. Therefore, our attention is directed towards the challenging subset of SCLC patients with LMs. Disease progression of LM-SCLC patients is affected by various factors in the tumor microenvironment (TME), including immune cells, blood vessels, inflammatory mediators, metabolites, and NE substances. Beyond standard immuno-chemotherapy, ongoing efforts to manage LMs in SCLC encompass anti-angiogenic therapy, radiotherapy, microwave ablation (MWA) / radiofrequency ablation (RFA), trans-arterial chemoembolization (TACE), and systemic therapies in conjunction with local interventions. Prospective experimental and clinical investigations into SCLC should prioritize precise and individualized approaches to enhance the prognosis across distinct patient cohorts.
Collapse
Affiliation(s)
- Linjie Fan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, 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, 100021, China
| | - Yiwen Lin
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, 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, 100021, China
| | - Yunjie Fu
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, 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, 100021, China.
| |
Collapse
|
39
|
Zhang L, Yan L, Fu X, Tao Z, Liu S, Li R, Wang T, Mao Y, Shang W, Gong M, Jia X, Wang F. PDK1 promotes epithelial ovarian cancer progression by upregulating BGN. Acta Biochim Biophys Sin (Shanghai) 2024; 57:712-726. [PMID: 39578715 DOI: 10.3724/abbs.2024186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
Pyruvate dehydrogenase kinase 1 (PDK1) is a new therapeutic target that is dysregulated in multiple tumors. This study aims to explore the potential role and regulatory mechanism of PDK1 in epithelial ovarian cancer (EOC). We detect PDK1 expression in EOC tissues and cells using qRT-PCR and western blot analysis, and the effects of PDK1 on EOC cell malignant behaviors are explored. RNA sequencing analyses are performed to explore the differentially expressed genes in PDK1-silenced EOC cells. Furthermore, tumor-bearing mouse models are established to assess the impacts of PDK1 and BGN on EOC tumor growth and metastasis in vivo. The results show that PDK1 is upregulated in EOC tissues and cell lines. Biglycan (BGN) is downregulated in PDK1-silenced EOC cells, and its expression is positively correlated with PDK1 levels in EOC tissues. PDK1 depletion inhibits EOC cell proliferation, migration and invasion. Mechanistically, PDK1 and BGN are colocalized in the cytoplasm of EOC cells and interact with each other. PDK1 positively regulates BGN expression by enhancing BGN mRNA stability. BGN overexpression partially reverses the anti-tumor effects of PDK1 depletion on EOC cell malignant behaviors. PDK1 has also been revealed to upregulate BGN to activate the NF-κB oncogenic pathway in EOC cells. Additionally, PDK1 accelerates tumor growth and metastasis by modulating BGN expression. In conclusion, PDK1 functions as an oncogene, facilitating EOC progression by upregulating BGN and activating the NF-κB pathway. These findings may provide valuable biomarkers for the diagnosis and treatment of EOC.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
- Department of Gynecology, the Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian 223300, China
| | - Lina Yan
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Xin Fu
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
- Clinical Laboratory, Baoshan People's Hospital, Baoshan 678000, China
| | - Ziqi Tao
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Shuna Liu
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Rong Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Ting Wang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Yepeng Mao
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Wenwen Shang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Mi Gong
- Department of Gynecology, the Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian 223300, China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| |
Collapse
|
40
|
Ghaemi A, Abnous K, Taghdisi SM, Vakili-Azghandi M, Ramezani M, Alibolandi M. Robust aptamer-targeted CRISPR/Cas9 delivery using mesenchymal stem cell membrane -liposome hybrid: BIRC5 gene knockout against melanoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102778. [PMID: 39127174 DOI: 10.1016/j.nano.2024.102778] [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: 04/16/2024] [Revised: 07/10/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
In this study, a platform was fabricated by combining a cationic lipid, 1,2-Dioleoyl-3-trimethylammonium-propane (DOTAP) with mesenchymal stem cell membrane (MSCM) to produce a positively charged hybrid vesicle. The prepared hybrid vesicle was used to condense BIRC5 CRISPR/Cas9 plasmid for survivin (BIRC5) gene editing. The Sgc8-c aptamer (against protein tyrosine kinase 7) was then attached to the surface of the prepared NPs through electrostatic interactions. In this regard, melanoma cancer cells (B16F0 cell line) overexpressing PTK7 receptor could be targeted. Investigations were conducted on this system to evaluate its transfection efficiency, cellular toxicity, and therapeutic performance in preclinical stage using B16F0 tumor bearing C57BL/6 J mice. The results verified the superiority of the Hybrid/ BIRC5 compared to Liposome/ BIRC5 in terms of cellular toxicity and transfection efficiency. The cells exposure to Hybrid/BIRC5 significantly enhanced cytotoxicity. Moreover, Apt-Hybrid/BIRC5 showed higher anti-proliferation activity toward PTK7-positive B16F0 cancer cells than that of the PKT7-negative CHO cell line. The active tumor targeting nanoparticles increased the cytotoxicity through down-regulation of BIRC5 expression as confirmed by Western blot analysis. In preclinical stage, Apt-Hybrid/BIRC5 showed remarkable tumor growth suppression toward B16F0 tumorized mice. Thus, our study suggested that genome editing for BIRC5 through the CRISPR/Cas9 system could provide a potentially safe approach for melanoma cancer therapy and has great potential for clinical translation.
Collapse
Affiliation(s)
- Asma Ghaemi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Vakili-Azghandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Engelken JA, Butelmann T, Tribukait-Riemenschneider F, Shastri VP. Towards a 3D-Printed Millifluidic Device for Investigating Cellular Processes. MICROMACHINES 2024; 15:1348. [PMID: 39597157 PMCID: PMC11596629 DOI: 10.3390/mi15111348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Microfluidic devices (µFDs) have been explored extensively in drug screening and studying cellular processes such as migration and metastasis. However, the fabrication and implementation of microfluidic devices pose cost and logistical challenges that limit wider-spread adoption. Despite these challenges, light-based 3D printing offers a potential alternative to device fabrication. This study reports on the development of millifluidic devices (MiFDs) for disease modeling and elucidates the methods and implications of the design, production, and testing of 3D-printed MiFDs. It further details how such millifluidic devices can be cost-efficiently and effortlessly produced. The MiFD was developed through an iterative process with analytical tests (flow tests, leak tests, cytotoxicity assays, and microscopic analyses), driving design evolution and determination of the suitability of the devices for disease modeling and cancer research. The design evolution also considered flow within tissues and replicates interstitial flow between the main flow path and the modules designed to house and support organ-mimicking cancer cell spheroids. Although the primary stereolithographic (SLA) resin used in this study showed cytotoxic potential despite its biocompatibility certifications, the MiFDs possessed essential attributes for cell culturing. In summary, SLA 3D printing enables the production of MiFDs as a cost-effective, rapid prototyping alternative to standard µFD fabrication for investigating disease-related processes.
Collapse
Affiliation(s)
- Jared A. Engelken
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany; (J.A.E.); (F.T.-R.)
| | - Tobias Butelmann
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany; (J.A.E.); (F.T.-R.)
| | | | - V. Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany; (J.A.E.); (F.T.-R.)
- BIOSS Centre of Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
42
|
Srivastava P, Jha S, Singh SK, Vyas H, Sethupathi P, Nair RS, Ramachandran K, Rana B, Kumar S, Rana A. Protease activated receptor-1 regulates mixed lineage kinase-3 to drive triple-negative breast cancer tumorigenesis. Cancer Lett 2024; 603:217200. [PMID: 39222677 DOI: 10.1016/j.canlet.2024.217200] [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: 05/28/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Triple-negative breast cancer (TNBC) is difficult to treat breast cancer subtype due to lack or insignificant expressions of targetable estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2). Therefore, finding a targetable protein or signaling pathway in TNBC would impact patient care. Here, we report that a member of the Mixed Lineage Kinase (MLK) family, MLK3, is an effector of G-protein-coupled protease-activated receptors 1 (PAR1) and targeting MLK3 by a small-molecule inhibitor prevented PAR1-mediated TNBC tumorigenesis. In silico and immunohistochemistry analysis of human breast tumors showed overexpression of PAR1 and MLK3 in TNBC tumors. Treating α-thrombin and PAR1 agonist increased MLK3 and JNK activities and induced cell migration in TNBC cells. The PAR1 positive/high (PAR1+/hi) population of TNBC cells showed aggressive tumor phenotype with increased MLK3 signaling. Moreover, combined inhibition of the PAR1 and MLK3 mitigated the TNBC tumor burden in preclinical TNBC models. Our data suggests that activation of the PAR1-MLK3 axis promotes TNBC tumorigenesis. Therefore, combinatorial therapy targeting MLK3 and PAR1 could effectively reduce TNBC tumor burden.
Collapse
Affiliation(s)
- Piush Srivastava
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Saket Jha
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Harsh Vyas
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Periannan Sethupathi
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rakesh Sathish Nair
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kheerthivasan Ramachandran
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA; Research Unit, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL, 60612, USA; Research Unit, Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
43
|
Zhou S, Li H, Zhao C, Zhao W, Pan X, Jian W, Wang J. Single‑cell RNA sequencing reveals heterogeneity in ovarian cancer and constructs a prognostic signature for prognostic prediction and immunotherapy. Int Immunopharmacol 2024; 140:112855. [PMID: 39133955 DOI: 10.1016/j.intimp.2024.112855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Ovarian cancer (OC) is one of the cancers with a high incidence at present, which poses a severe threat to women's health. This study focused on identifying the heterogeneity among malignant epithelial cell OC and constructing an effective prognostic signature to predict prognosis and immunotherapy according to a multidisciplinary study. METHODS The InterCNV algorithm was used to identify the heterogeneity of OC based on the scRNA-seq and bulk RNA-seq data. Six algorithms selected EMTscore. An effective prognostic signature was conducted using the COX and Least Absolute Shrinkage and Selection Operator (LASSO) regression algorithms. The texting datasets were used to assess the accuracy of the prognostic signature. We evaluated different immune characteristics and immunotherapy response differences among other risk groups. RESULTS A prognostic signature including 14 genes was established. The patients in the high-risk group have poor survival outcomes. We also found that the patients in the low-risk group have higher immune cell infiltration, enrichment of immune checkpoints, and immunotherapy response, suggesting that the patients in the low-risk group may be more sensitive to immunotherapy. Finally, the laboratory test results showed that KREMEN2 was identified as a novel biomarker and therapeutic target for OC patients. CONCLUSIONS Our study established a GRG signature consisting of 16 genes based on the scRNA-seq and bulk RNA-seq data, which provides a new perspective on the prediction of prognosis and treatment strategy for OC.
Collapse
Affiliation(s)
- Shisi Zhou
- Department of Gynaecology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Huiyan Li
- Department of Rheumatology and Immunology, The Fourth Affiliated Hospital, China Medical University, Shenyang 110000, China
| | - Chengzhi Zhao
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Wancheng Zhao
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue Pan
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weilan Jian
- Department of Anesthesiology, Shanghai Tenth People's Hospital, Shanghai, China.
| | - Jieli Wang
- Department of Gynaecology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China.
| |
Collapse
|
44
|
Ye Y, Wang J, Izban MG, Ballard BR, Barsky SH. Initiation of tumor dormancy by the lymphovascular embolus. Oncotarget 2024; 15:726-740. [PMID: 39392391 PMCID: PMC11468568 DOI: 10.18632/oncotarget.28658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Cancer dormancy followed by recurrence remains an enigma in cancer biology. Since both local and systemic recurrences are thought to emanate from dormant micrometastasis which take origin from lymphovascular tumor emboli we wondered whether the process of dormancy might initiate within lymphovascular emboli. This study combines experimental studies with a patient-derived xenograft (PDX) of inflammatory breast cancer (Mary-X) that spontaneously forms spheroids in vitro and budding lymphovascular tumor emboli in vivo with observational studies utilizing tissue microarrays (TMAs) of human breast cancers. In the experimental studies, Mary-X during both lymphovascular emboli formation in vivo and spheroidgenesis in vitro exhibited decreased proliferation, a G0/G1 cell cycle arrest and decreased mTOR signaling. This induction of dormancy required calpain-mediated E-cadherin proteolysis and was mediated by decreased P13K signaling, resulting in decreased mTOR activity. In observational human breast cancer studies, increased E-cadherin immunoreactivity due to increased E-cad/NTF-1 but both decreased Ki-67 and mTOR activity was observed selectively and differentially within the lymphovascular tumor emboli. Both our experimental as well as observational studies indicate that in vivo lymphovascular tumor emboli and their in vitro spheroid equivalent initiate dormancy through these pathways.
Collapse
Affiliation(s)
- Yin Ye
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| | - Justin Wang
- Department of Graduate Medical Education, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Michael G. Izban
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| | - Billy R. Ballard
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| | - Sanford H. Barsky
- Department of Pathology, Anatomy and Cell Biology and the Clinical and Translational Research Center of Excellence, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
45
|
Sahoo S, Ghosh S, Areekkadan AM, Chakrabarty A, Banerjee R. Cancer Cell-Selective Inhibition of Migration by Styrenic Catiomer Emulsions. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53599-53609. [PMID: 39340815 DOI: 10.1021/acsami.4c14410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Cancer metastasis remains the most formidable cause of mortality and morbidity in cancer patients. Developing an effective and economical method toward cancer antimetastatic strategy demands immediate attention in anticancer therapy. Herein, we followed a cost-effective greener method for preparing a small family of amphiphilic catiomers with varied styrene content (45, 63, and 83%), which revealed the unique potential of promoting normal cell migration while retarding cancer metastasis. The styrenic polymers formed micellar self-assembly in aqueous phase and exhibited a cationic charge. Polymers were quite nontoxic up to 200 μg/mL concentration toward human embryonic kidney cell HEK293 as well as human, triple negative breast cancer cell MDAMB-231, mouse melanoma cell B16F10, and human oral squamous carcinoma cell FaDu. Confocal imaging and fluorescence activated cell sorting (FACS) showed effective incorporation of polymers within cells. Interestingly, the polymer-treated HEK293 cells underwent prominent wound healing in scratch assay. However, the as-synthesized polymer-treated cancer cells resisted migration as analyzed from the scratch assay. A mechanistic study using immunoblotting assay established upregulation of migratory proteins vimentin and TGF-β and downregulation of E-cadherin in normal HEK293 cells. Remarkably, this trend was completely reversed in cancer cell MDAMB-231. This study describes the extraordinary potential of styrenic catiomers as wound healers for normal cells while inhibiting cancer metastasis.
Collapse
Affiliation(s)
- Subhasish Sahoo
- Department of Oils, Lipid Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Souma Ghosh
- Department of Oils, Lipid Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abdul Malik Areekkadan
- Department of Polymers & Functional Materials, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Arindam Chakrabarty
- Department of Polymers & Functional Materials, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rajkumar Banerjee
- Department of Oils, Lipid Science & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
46
|
Schmid KF, Zeinali S, Moser SK, Dubey C, Schneider S, Deng H, Haefliger S, Marti TM, Guenat OT. Assessing the metastatic potential of circulating tumor cells using an organ-on-chip model. Front Bioeng Biotechnol 2024; 12:1457884. [PMID: 39439549 PMCID: PMC11493642 DOI: 10.3389/fbioe.2024.1457884] [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: 07/01/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Metastatic lung cancer remains a leading cause of death worldwide, with its intricate metastatic cascade posing significant challenges to researchers and clinicians. Despite substantial progress in understanding this cascade, many aspects remain elusive. Microfluidic-based vasculature-on-chip models have emerged as powerful tools in cancer research, enabling the simulation of specific stages of tumor progression. In this study, we investigate the extravasation behaviors of A549 lung cancer cell subpopulations, revealing distinct differences based on their phenotypes. Our results show that holoclones, which exhibit an epithelial phenotype, do not undergo extravasation. In contrast, paraclones, characterized by a mesenchymal phenotype, demonstrate a notable capacity for extravasation. Furthermore, we observed that paraclones migrate significantly faster than holoclones within the microfluidic model. Importantly, we found that the depletion of vascular endothelial growth factor (VEGF) effectively inhibits the extravasation of paraclones. These findings highlight the utility of microfluidic-based models in replicating key aspects of the metastatic cascade. The insights gained from this study underscore the potential of these models to advance precision medicine by facilitating the assessment of patient-specific cancer cell dynamics and drug responses. This approach could lead to improved strategies for predicting metastatic risk and tailoring personalized cancer therapies, potentially involving the sampling of cancer cells from patients during tumor resection or biopsies.
Collapse
Affiliation(s)
- Karin F. Schmid
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Soheila Zeinali
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Susanne K. Moser
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Christelle Dubey
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sabine Schneider
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Simon Haefliger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas M. Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
47
|
Fabisiewicz A, Szostakowska-Rodzos M, Grzybowska EA. Improving the Prognostic and Predictive Value of Circulating Tumor Cell Enumeration: Is Longitudinal Monitoring the Answer? Int J Mol Sci 2024; 25:10612. [PMID: 39408942 PMCID: PMC11476589 DOI: 10.3390/ijms251910612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Circulating tumor cell (CTC) numbers in the blood of cancer patients can indicate the progression and invasiveness of tumors, and their prognostic and predictive value has been repeatedly demonstrated. However, the standard baseline CTC count at the beginning of treatment, while informative, is not completely reliable and may not adequately reflect the state of the disease. A growing number of studies indicate that the long-term monitoring of CTC numbers in the same patient provides more comprehensive prognostic data and should be incorporated into clinical practice, as a factor that contributes to therapeutic decisions. This review describes the current status of CTC enumeration as a prognostic and predictive factor, highlights the shortcomings of current solutions, and advocates for longitudinal CTC analysis as a more effective method of the evaluation of developing disease, treatment efficacy, and the long term-monitoring of the minimal residual disease. As evidenced by the described reports, the longitudinal monitoring of CTCs should provide a better and more sensitive prediction of the course of the disease, and its incorporation in clinical practice should be beneficial.
Collapse
Affiliation(s)
| | | | - Ewa A. Grzybowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (A.F.); (M.S.-R.)
| |
Collapse
|
48
|
He C. Activating Invasion and Metastasis in Small Cell Lung Cancer: Role of the Tumour Immune Microenvironment and Mechanisms of Vasculogenesis, Epithelial-Mesenchymal Transition, Cell Migration, and Organ Tropism. Cancer Rep (Hoboken) 2024; 7:e70018. [PMID: 39376011 PMCID: PMC11458887 DOI: 10.1002/cnr2.70018] [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: 03/12/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) harbours the most aggressive phenotype of all lung cancers to correlate with its bleak prognosis. The aggression of SCLC is partially attributable to its strong metastatic tendencies. The biological processes facilitating the metastasis in SCLC are still poorly understood and garnering a deeper understanding of these processes may enable the exploration of additional targets against this cancer hallmark in the treatment of SCLC. RECENT FINDINGS This narrative review will discuss the proposed molecular mechanisms by which the cancer hallmark of activating invasion and metastasis is featured in SCLC through important steps of the metastatic pathway, and address the various molecular targets that may be considered for therapeutic intervention. The tumour immune microenvironment plays an important role in facilitating immunotherapy resistance, whilst the poor infiltration of natural killer cells in particular fosters a pro-metastatic environment in SCLC. SCLC vasculogenesis is achieved through VEGF expression and vascular mimicry, and epithelial-mesenchymal transition is facilitated by the expression of the transcriptional repressors of E-cadherin, the suppression of the Notch signalling pathway and tumour heterogeneity. Nuclear factor I/B, selectin and B1 integrin hold important roles in SCLC migration, whilst various molecular markers are expressed by SCLC to assist organ-specific homing during metastasis. The review will also discuss a recent article observing miR-1 mRNA upregulation as a potential therapeutic option in targeting the metastatic activity of SCLC. CONCLUSION Treatment of SCLC remains a clinical challenge due to its recalcitrant and aggressive nature. Amongst the many hallmarks used by SCLC to enable its aggressive behaviour, that of its ability to invade surrounding tissue and metastasise is particularly notable and understanding the molecular mechanisms in SCLC metastasis can identify therapeutic targets to attenuate SCLC aggression and improve mortality.
Collapse
Affiliation(s)
- Carl He
- Department of Oncology, Eastern HealthUniversity of MelbourneMelbourneAustralia
| |
Collapse
|
49
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
50
|
Bekker RA, Obertopp N, Redler G, Penagaricano J, Caudell JJ, Yamoah K, Pilon-Thomas S, Moros EG, Enderling H. Spatially fractionated GRID radiation potentiates immune-mediated tumor control. Radiat Oncol 2024; 19:121. [PMID: 39272128 PMCID: PMC11401399 DOI: 10.1186/s13014-024-02514-6] [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: 02/06/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Tumor-immune interactions shape a developing tumor and its tumor immune microenvironment (TIME) resulting in either well-infiltrated, immunologically inflamed tumor beds, or immune deserts with low levels of infiltration. The pre-treatment immune make-up of the TIME is associated with treatment outcome; immunologically inflamed tumors generally exhibit better responses to radio- and immunotherapy than non-inflamed tumors. However, radiotherapy is known to induce opposing immunological consequences, resulting in both immunostimulatory and inhibitory responses. In fact, it is thought that the radiation-induced tumoricidal immune response is curtailed by subsequent applications of radiation. It is thus conceivable that spatially fractionated radiotherapy (SFRT), administered through GRID blocks (SFRT-GRID) or lattice radiotherapy to create areas of low or high dose exposure, may create protective reservoirs of the tumor immune microenvironment, thereby preserving anti-tumor immune responses that are pivotal for radiation success. METHODS We have developed an agent-based model (ABM) of tumor-immune interactions to investigate the immunological consequences and clinical outcomes after 2 Gy × 35 whole tumor radiation therapy (WTRT) and SFRT-GRID. The ABM is conceptually calibrated such that untreated tumors escape immune surveillance and grow to clinical detection. Individual ABM simulations are initialized from four distinct multiplex immunohistochemistry (mIHC) slides, and immune related parameter rates are generated using Latin Hypercube Sampling. RESULTS In silico simulations suggest that radiation-induced cancer cell death alone is insufficient to clear a tumor with WTRT. However, explicit consideration of radiation-induced anti-tumor immunity synergizes with radiation cytotoxicity to eradicate tumors. Similarly, SFRT-GRID is successful with radiation-induced anti-tumor immunity, and, for some pre-treatment TIME compositions and modeling parameters, SFRT-GRID might be superior to WTRT in providing tumor control. CONCLUSION This study demonstrates the pivotal role of the radiation-induced anti-tumor immunity. Prolonged fractionated treatment schedules may counteract early immune recruitment, which may be protected by SFRT-facilitated immune reservoirs. Different biological responses and treatment outcomes are observed based on pre-treatment TIME composition and model parameters. A rigorous analysis and model calibration for different tumor types and immune infiltration states is required before any conclusions can be drawn for clinical translation.
Collapse
Affiliation(s)
- Rebecca A Bekker
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, 33612, USA
| | - Nina Obertopp
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, 33612, USA
| | - Gage Redler
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - José Penagaricano
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Jimmy J Caudell
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Kosj Yamoah
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Eduardo G Moros
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Heiko Enderling
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|