1
|
Valla SA, Chimento A, Demarchi G, Prodan EN, Werner E, Vitale DL, Romano ML, Alaniz LD, Becú-Villalobos D, Cristina C. Pituitary folliculo-stellate cells modulate tumor vasculature and extracellular matrix composition in experimental lactosomatotropinomas. Biochem Biophys Res Commun 2025; 767:151876. [PMID: 40315570 DOI: 10.1016/j.bbrc.2025.151876] [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: 01/03/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
Folliculo-stellate cells (FSCs) constitute 5-10 % of the adenohypophysis and have been proposed as paracrine regulators of pituitary cells. However, their participation in pituitary tumor development remains unclear. We generated a lacto-somatotropic tumor model by subcutaneous injection of GH3 (lacto-somatotrophs) and the FSCs TtT/GF, isolated or combined, in immunodeficient mice, to study the role of the FSCs on tumor formation, hormone secretion, vascularization and extra-cellular matrix involvement. The co-culture of both cell lines let us gain insight into the proliferative and secretory action of FSC in pituitary tumor modulation. Our results showed that initially GH3:TtT/GF tumors had an earlier onset, but lately, TtT/GF cells restrained GH3:TtT/GF tumor growth and their Prl synthesis, although no differences were observed in the proliferative potential of tumor cells in vivo. Instead, TtT/GF cells exerted a direct mitogenic action on GH3 cells in vitro. Moreover, GH3 tumors had fewer irrigating vessels, lower vascular area and a higher VEGF/bFGF ratio that correlated with Hif1a expression, consistent with the tissue hypoxia and hemorrhage. These features were downregulated in their co-injected counterparts, which interestingly showed an increased deposition of collagens, glycoproteins and mucopolysaccharides extra-cellular matrix (EMC) components. Isolated TtT/GF injected cells did not generate tumors, but they developed fibrous masses characterized by collagen and high bFGF production. In conclusion, our results demonstrate that FSCs are dual regulators of pituitary tumor growth, with a mitogenic action on tumor cells but also a restrictive tumor effect on the cancer processes angiogenesis, hypoxia, and ECM remodeling.
Collapse
Affiliation(s)
- S A Valla
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - A Chimento
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina; Comisión de Investigaciones Científicas de La Provincia de Buenos Aires, CIC, La Plata, Buenos Aires, Argentina
| | - G Demarchi
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - E N Prodan
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - E Werner
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - D L Vitale
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - M L Romano
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - L D Alaniz
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina
| | - D Becú-Villalobos
- Instituto de Biología y Medicina Experimental (IByME-CONICET), CABA, Argentina
| | - C Cristina
- Centro de Investigaciones Básicas y Aplicadas, Universidad Nacional Del Noroeste de la Provincia de Buenos Aires, Junín, Buenos Aires, Argentina; Centro de Investigaciones y Transferencia Del Noroeste de La Provincia de Buenos Aires (CITNOBA) - UNNOBA-UNSAdA-CONICET, Pergamino, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Lundholm L, Montelius M, Jalnefjord O, Schoultz E, Forssell‐Aronsson E, Ljungberg M. Cluster Analysis of VERDICT MRI for Cancer Tissue Characterization in Neuroendocrine Tumors. NMR IN BIOMEDICINE 2025; 38:e70050. [PMID: 40296332 PMCID: PMC12038086 DOI: 10.1002/nbm.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
Diffusion MRI models accounting for varying diffusion times and high b-values, such as VERDICT, hold potential for non-invasively characterizing tumor tissue types, potentially enabling improved tumor grading, and treatment evaluation. Furthermore, cluster analysis can aid in identifying multidimensional patterns in the diffusion MRI (dMRI) data that are not apparent when analyzing individual parameters in isolation. The aim of this study was to evaluate how well cluster analysis of VERDICT parameters can be used for intratumor tissue characterization compared to ADC in a mouse model of human small intestine neuroendocrine tumor (GOT1), and to validate the method by histological analysis. Mice implanted with GOT1 were irradiated and subsequently imaged using a dMRI protocol designed for estimation of VERDICT parameters and ADC values. Histological analysis using hematoxylin and eosin (H&E), Masson's trichrome, and Ki67 staining identified three distinct tumor tissue types: necrotic, fibrotic, and viable tumor tissue. ROIs were drawn on regions of high and low ADC, which spatially matched with necrosis or fibrosis, and viable tumor tissue, respectively. Among the VERDICT parameters, the cell radius index (R) was most effective in distinguishing between necrotic and fibrotic tissue, whereas the intracellular fraction (fIC) was the most effective in differentiating viable from non-viable tissue. A Gaussian mixture model (GMM) of three clusters, representing each tumor tissue type, was fitted to R and fIC of all tumor voxel data. VERDICT cluster maps corresponded well with the histology classification maps overall. Fibrotic tissue corresponded best with the cluster of low fIC and low R, necrotic tissue with the cluster of low fIC and high R, and viable tumor tissue with the cluster of high fIC and intermediate R. In conclusion, GMM cluster analysis of VERDICT MRI data shows potential in differentiating necrotic, fibrotic, and viable tumor tissue in irradiated GOT1 tumors.
Collapse
Affiliation(s)
- Lukas Lundholm
- Department of Medical Radiation SciencesInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Mikael Montelius
- Department of Medical Radiation SciencesInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Oscar Jalnefjord
- Department of Medical Radiation SciencesInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Medical Physics and Biomedical EngineeringSahlgrenska University HospitalGothenburgSweden
| | - Elin Schoultz
- Department of Medical Biochemistry and Cell BiologySahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Eva Forssell‐Aronsson
- Department of Medical Radiation SciencesInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Medical Physics and Biomedical EngineeringSahlgrenska University HospitalGothenburgSweden
| | - Maria Ljungberg
- Department of Medical Radiation SciencesInstitute of Clinical Sciences, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of Medical Physics and Biomedical EngineeringSahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
3
|
Harun A, Bendele N, Khalil MI, Vasquez I, Djuanda J, Posey R, Rashid MH, Christopher GF, Bickel U, Gruev V, Tropp J, Egan PF, Srivastava I. 3D Tumor-Mimicking Phantom Models for Assessing NIR I/II Nanoparticles in Fluorescence-Guided Surgical Interventions. ACS NANO 2025. [PMID: 40378397 DOI: 10.1021/acsnano.5c01919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Fluorescence image-guided surgery (FIGS) offers high spatial resolution and real-time feedback but is limited by shallow tissue penetration and autofluorescence from current clinically approved fluorophores. The near-infrared (NIR) spectrum, specifically the NIR-I (700-900 nm) and NIR-II (950-1700 nm), addresses these limitations with deeper tissue penetration and improved signal-to-noise ratios. However, biological barriers and suboptimal optical performance under surgical conditions have hindered the clinical translation of NIR-I/II nanoprobes. In vivo mouse models have shown promise, but these models do not replicate the complex optical scenarios encountered during real-world surgeries. Existing tissue-mimicking phantoms used to evaluate NIR-I/II imaging systems are useful but fall short when assessing nanoprobes in surgical environments. These phantoms often fail to replicate the tumor microenvironment, limiting their predictive assessment. To overcome these challenges, we propose developing tumor-mimicking phantom models (TMPs) that integrate key tumor features, such as tunable tumor cell densities, in vivo-like nanoparticle concentrations, biologically relevant factors (pH, enzymes), replicate light absorption components (hemoglobin), and light scattering components (intralipid). These TMPs enable more clinically relevant assessments of NIR-I/II nanoprobes, including optical tissue penetration profiling, tumor margin delineation, and ex vivo thoracic surgery on porcine lungs. The components of TMPs can be further modulated to closely match the optical profiles of in vivo and ex vivo tumors. Additionally, 3D bioprinting technology facilitates a high-throughput platform for screening nanoprobes under realistic conditions. This approach will identify high-performing NIR-I/II probes with superior surgical utility, bridging the gap between preclinical findings and clinical applications, and ensuring results extend beyond traditional in vivo mouse studies.
Collapse
Affiliation(s)
- Asma Harun
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas 79106, United States
| | - Nathaniel Bendele
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Mohammad Ibrahim Khalil
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Isabella Vasquez
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas 79106, United States
| | - Jonathan Djuanda
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Robert Posey
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Md Hasnat Rashid
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Gordon F Christopher
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Ulrich Bickel
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas 79106, United States
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Science Center, Amarillo, Texas 79106, United States
| | - Viktor Gruev
- Department of Electrical and Computer Engineering and Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Joshua Tropp
- Department of Chemistry & Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas 79106, United States
| | - Paul F Egan
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Indrajit Srivastava
- Department of Mechanical Engineering, Edward E. Whitacre Jr. College of Engineering, Texas Tech University, Lubbock, Texas 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas 79106, United States
| |
Collapse
|
4
|
Wang K, Shen X, Wu J, Bi Q, Gao Z, Sun Z, Wang W. Fibrogenesis-driven tumor progression in clear cell renal cell carcinoma: prognostic, therapeutic implications and the dual role of neuropilin-1. Cancer Cell Int 2025; 25:179. [PMID: 40380175 PMCID: PMC12082889 DOI: 10.1186/s12935-025-03801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the predominant subtype of renal cancer, with a poor prognosis driven by therapy resistance and a propensity for recurrence. Tumor microenvironment (TME)-associated fibrosis accelerates disease progression by fostering immune evasion. Neuropilin-1 (NRP1), a key mediator in fibrotic signaling and cancer biology, has been implicated in these processes. However, the genetic correlation between fibrogenesis and ccRCC remains largely unexplored, necessitating a focused analysis of fibrogenesis-related genes (FRGs) to identify novel prognostic markers and therapeutic strategies. METHODS This study utilized an integrative bioinformatics framework to identify prognosis-associated fibrogenesis-related genes (pFRGs) and applied non-negative matrix factorization (NMF) to stratify ccRCC patients based on fibrotic signatures. A machine learning-derived prognostic model was developed to categorize patients into high-risk and low-risk groups, with tumor microenvironment (TME) features analyzed across these subgroups. The pro-tumorigenic role of NRP1 via the TGF-β/SMAD signaling pathway was validated in vitro and in vivo. RESULTS Twelve pFRGs were identified, with elevated expression correlating with reduced survival. NMF revealed two ccRCC subtypes with different fibrotic and immune profiles. The high-fibrosis subtype showed worse survival and a pro-tumorigenic TME. The risk model demonstrated robust predictive performance (AUCs: 0.738, 0.731, 0.711 for 1-, 2-, and 3-year survival). High-risk patients, marked by immune dysfunction, exhibited worse survival but greater immunotherapy sensitivity. Among the pFRGs, NRP1 was upregulated in ccRCC, and paradoxically associated with favorable prognosis in TCGA, primarily due to stromal enrichment. In vitro and in vivo experiments confirmed that NRP1 promotes ccRCC proliferation, migration, and invasion by enhancing TGF-β/SMAD-driven epithelial-mesenchymal transition (EMT). CONCLUSION Fibrosis is a critical driver of ccRCC progression, linking fibrogenesis-related genes to poor prognosis, immune suppression, and tumor aggressiveness. NRP1 was identified as a central regulator of fibrosis-induced tumor progression through the TGF-β/SMAD signaling pathway. Combining NRP1 inhibition with anti-fibrotic therapies presents a potential strategy for enhancing therapeutic outcomes in ccRCC.
Collapse
Affiliation(s)
- Kai Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Urology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xihao Shen
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qing Bi
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zihao Gao
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Vadibeler S, Clarke S, Phyu SM, Parkes EE. Interactions between cancer-associated fibroblasts and the extracellular matrix in oesophageal cancer. Matrix Biol 2025:S0945-053X(25)00049-6. [PMID: 40379112 DOI: 10.1016/j.matbio.2025.05.003] [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: 04/07/2025] [Revised: 05/13/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Stromal components of the tumour microenvironment, such as cancer-associated fibroblasts (CAFs) and the extracellular matrix (ECM), are actively involved in tumorigenesis. CAFs and the ECM co-evolve with resultant molecular and mechanical pressure on tumour cells mediated by CAFs via the ECM. Meanwhile, ECM fibers determine CAF differentiation and activity, establishing a protumorigenic feed-forward loop. Oesophageal cancer carries a high morbidity and mortality, and curative surgical resection is only an option for a limited number of patients while early lymphatic spread and poor therapeutic responses are common. Although studies report marked heterogeneity in investigation of the stromal density of gastrointestinal cancers, it is generally accepted that oesophageal cancer is highly fibrotic, and stromal components like CAFs may outnumber cancer cells. Therefore, a comprehensive understanding of the reciprocal interaction between CAFs and the ECM in oesophageal cancer is essential to improving diagnostics and prognostication, as well as designing innovative anti-cancer strategies. Here, we summarise current understanding of oesophageal cancer from a stromal perspective. Then, we discuss that CAFs and the ECM in oesophageal cancer can independently and synergistically contribute to tumour progression and therapeutic resistance. We also summarise potential stromal targets that have been described in transcriptomic analyses, highlighting those validated in downstream experimental studies. Importantly, clinical translation of stromal-targeting strategies in oesophageal cancer is still in its infancy but holds significant promise for future therapeutic combinations.
Collapse
Affiliation(s)
- Subashan Vadibeler
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford; Department of Oncology, University of Oxford
| | - Shannique Clarke
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford; Department of Oncology, University of Oxford
| | - Su M Phyu
- Department of Oncology, University of Oxford
| | - Eileen E Parkes
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford; Department of Oncology, University of Oxford
| |
Collapse
|
6
|
Geronimo G, Rodrigues da Silva GH, de Moura LD, de Carvalho FV, Mendonça TC, Olivo LB, Verlindo de Araújo B, Dalla Costa TC, Lavareze L, Mariano FV, de Paula E. Enantiomeric Excess Bupivacaine in a Lavender Oil NLC Tested in a Melanoma Model: Prolonged Release and Anticancer Effect. Mol Pharm 2025. [PMID: 40310503 DOI: 10.1021/acs.molpharmaceut.5c00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Recent studies have highlighted the potential of local anesthetics (LA) as adjuvants in cancer treatment, specifically by increasing survival rates when used in surgical excisions. However, the clinical use of LA is restricted due to their systemic toxicity. The development of drug delivery systems could address this issue and advance the utilization of these molecules. In this research, we explored the pharmacokinetics (using microdialysis probes) and antitumor properties of a nanostructured lipid carrier (NLC) formulation containing the commercially available enantiomeric excess form of bupivacaine (BVCS75). This NLC was prepared with lavender oil (NLC-L-BVC), an excipient with inherent antitumor properties. We compared this formulation to a control (NLC-BVC) using synthetic lipids. Pharmacokinetic assessments of the NLCs confirmed the sustained release of BVCS75 within the tumor, characterized by a reduced elimination rate constant and longer half-life (∼6×). The encapsulation of BVCS75 within nanoparticles (whether natural or synthetic) enhanced its effectiveness in treating the primary tumor, resulting in the inhibition of tumor growth (70% with NLC-L-BVC and 72% with NLC-BVC), outperforming free BVC (17% inhibition). However, the association of lavender oil with BVCS75 in an NLC did not yield synergistic properties. Furthermore, all BVCS75 treatments (whether free or encapsulated) improved animal survival rates. These findings confirm that encapsulation of bupivacaine in NLC can prolong drug action at the local site, contributing to improved local antitumor therapy while mitigating systemic effects.
Collapse
Affiliation(s)
- Gabriela Geronimo
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas─UNICAMP, Campinas, SP 13083-862, Brazil
| | - Gustavo H Rodrigues da Silva
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, SP 13083-970, Brazil
| | - Ludmilla D de Moura
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas─UNICAMP, Campinas, SP 13083-862, Brazil
| | - Fabíola V de Carvalho
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas─UNICAMP, Campinas, SP 13083-862, Brazil
| | - Talita C Mendonça
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas─UNICAMP, Campinas, SP 13083-862, Brazil
| | - Laura B Olivo
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul─UFRGS, Porto Alegre, RS 90610-000, Brazil
| | - Bibiana Verlindo de Araújo
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul─UFRGS, Porto Alegre, RS 90610-000, Brazil
| | - Teresa C Dalla Costa
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul─UFRGS, Porto Alegre, RS 90610-000, Brazil
| | - Luccas Lavareze
- Department of Pathology, Faculty of Medical Sciences, UNICAMP, Campinas, SP 13083-888, Brazil
| | - Fernanda V Mariano
- Department of Pathology, Faculty of Medical Sciences, UNICAMP, Campinas, SP 13083-888, Brazil
| | - Eneida de Paula
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas─UNICAMP, Campinas, SP 13083-862, Brazil
| |
Collapse
|
7
|
Chang Z, Wang Z, Chen Y, Liu Y, Gao Y, Cui Y, Wang L, Liu Y, Cheng R, Liu R, Zhang L. Metabolism profiles of tannins in Phyllanthus emblica L. and its immunotherapeutic potential against hepatocellular carcinoma by re-educating tumor microenvironment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156576. [PMID: 40085988 DOI: 10.1016/j.phymed.2025.156576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Tumor-associated macrophages (TAMs) are key components of the immunosuppressive tumor microenvironment and represent significant obstacles to effective immunotherapy. Phyllanthus emblica L. (PE), a medicinal plant traditionally used in Tibet, has shown therapeutic promise. This study investigates the effects of the tannin fraction of PE (PE-TF) on HCC and its ability to modulate the tumor immunosuppressive microenvironment. METHODS We evaluated the antitumor efficacy of PE-TF using H22 xenografts and Hepa1-6 orthotopic mouse models. Transcriptomic analysis was performed to identify molecular targets underlying PE-TF suppression of HCC growth. Additionally, UPLC-MS/MS analysis identified the prototypic and metabolic components of PE-TF present in serum, tumor tissues, and adjacent normal liver tissues in the orthotopic HCC model. RESULTS PE-TF significantly suppressed tumor growth in both subcutaneous and orthotopic HCC models and promoted reprogramming of TAMs toward an antitumor M1 phenotype in vivo. Furthermore, PE-TF counteracted the protumoral effects mediated by bone marrow-derived macrophages (BMDMs) exposed to Hepa1-6-derived conditioned medium (HCM). Although TBH promoted macrophage M2 polarization, the reactive oxygen species (ROS)-scavenging activity of PE-TF effectively inhibited this process. Modulation of the tumor microenvironment by PE-TF-enhanced CD8+T cell infiltration and bolstered their antitumor response, as evidenced by increased transcription of perforin, IFN-γ, and IL-2. Transcriptomic analysis further revealed that T-cell receptor and cytotoxic T-cell signaling pathways are critical mediators of PE-TF' therapeutic effects. Moreover, we preliminarily characterized 79 components across serum, liver, and tumor tissues, and identified metabolic pathways for PE-TF ingredients-including methylation and glycosylation modifications of tumor-enriched constituents. Notably, seven components, such as corilagin and urolithin D, are hypothesized to possess immunomodulatory properties. CONCLUSION Our findings underscore the potential of PE-TF as an adjuvant immunotherapy for HCC. By scavenging ROS, PE-TF reverses the immunosuppressive M2-TAM phenotype and remodels the tumor microenvironment, thereby enhancing antitumor immunity. Additionally, integrating chemical and metabolic profiling offers a promising strategy for refining candidate selection in future drug discovery endeavors.
Collapse
Affiliation(s)
- Zihao Chang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Zhaohui Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yinxin Chen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yuqi Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Ye Gao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yitong Cui
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Le Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Yue Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China
| | - Ruiyang Cheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| | - Runping Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China.
| | - Lanzhen Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, PR China.
| |
Collapse
|
8
|
Sergi CM, Guerra L, Hager J. Autosomal Dominant Polycystic Kidney Disease-Related Multifocal Renal Cell Carcinoma: A Narrative Iconographic Review. Int J Mol Sci 2025; 26:3965. [PMID: 40362206 PMCID: PMC12072103 DOI: 10.3390/ijms26093965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/19/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common inheritable disease of cystic degeneration in the kidney. ADPKD is a significant cause of end-stage renal disease (ESRD). Autosomal Dominant Polycystic Liver Disease (ADPLD) results in substantial PLD with minimal PKD. Currently, there are eight genes which have been associated with ADPKD (PKD1 and PKD2), ADPLD (PRKCSH, SEC63, LRP5, ALG8, and SEC61B), or both (GANAB). The severity of ADPKD can show an extremely broad range, but the evolution to ESRD is doubtless unavoidable. In some patients, carcinogenesis develops with inflammation as a potential promoting factor. In this chapter, we illustrate the severity of ADPKD and the fate to develop renal cell carcinoma (RCC).
Collapse
Affiliation(s)
- Consolato M. Sergi
- Anatomic Pathology, Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Luis Guerra
- Pediatric Urology, Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON K1H 8L1, Canada;
| | - Josef Hager
- Pediatric Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
9
|
Huang C, Shao Y, Bai J, Zhao Y, Ogawa R. Fibroproliferative conditions: the 3R approach bridging fibrosis and tumors. Trends Mol Med 2025:S1471-4914(25)00060-7. [PMID: 40268589 DOI: 10.1016/j.molmed.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 02/10/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025]
Abstract
Soft-tissue fibroproliferative conditions (FPCs) affect many organs. All demonstrate the accumulation of (myo)fibroblasts and extracellular matrix. Currently, FPCs are classified according to the affected body site/organ. To promote research into the etiological mechanisms that drive pathological FPCs, we propose a new, more clinically grounded, FPC classification that is based on the intent and severity of the fibroproliferation. There are three categories: responsive, replacement, and reconstructive FPCs. Reconstructive FPCs (e.g., keloids) have quasi-neoplastic behaviors, including local invasiveness, and serve as a bridge between fibrosis and cancers. Comparisons of reconstructive FPCs to both cancers and the other FPC categories may help elucidate their pathogenic cellular properties, microenvironmental components, and intracellular-signaling mechanisms. Thus, the new FPC classification may promote research in the fibrosis field.
Collapse
Affiliation(s)
- Chenyu Huang
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China.
| | - Yue Shao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China.
| | - Jianbo Bai
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Yi Zhao
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
10
|
Akabane M, Kawashima J, Woldesenbetm S, Macedo AB, Altaf A, Aucejo F, Popescu I, Kitago M, Poultsides GA, Sasaki K, Imaoka Y, Ruzzenente A, Endo I, Pawlik TM. Enhancing outcome prediction in patients with colorectal liver metastases undergoing hepatectomy: the synergistic impact of FIB-4 index and tumor burden score across KRAS profiles. HPB (Oxford) 2025:S1365-182X(25)00546-5. [PMID: 40287297 DOI: 10.1016/j.hpb.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The prognostic value of Fibrosis-4 (FIB-4) index, concerning KRAS status (wild-type [wtKRAS] vs. mutated [mutKRAS]) remains unclear in post-hepatectomy colorectal liver metastases (CRLM). We evaluated the combined impact of FIB-4 and Tumor Burden Score (TBS) on overall survival (OS)/recurrence-free survival (RFS), stratified by KRAS status. METHODS CRLM patients undergoing hepatectomy (2000-2020) were analyzed, grouped by TBS/FIB-4. RESULTS Among 828 patients, 196 had high FIB-4. High TBS had worse 5-year OS (P < 0.001). In wtKRAS, high TBS correlated with worse OS (P < 0.001), but not in mutKRAS. High FIB-4 correlated with worse OS (P = 0.01). Sub-stratification showed no OS difference by FIB-4 in wtKRAS, but a difference in mutKRAS (P = 0.03). Multivariable analysis identified mutKRAS (HR: 1.90), high TBS (HR: 1.62), and FIB-4 (HR: 1.15) as mortality risk factors. The TBS-FIB-4-KRAS index had highest predictive accuracy. For RFS, TBS and FIB-4 independently stratified outcomes. High TBS was associated with worse RFS in wtKRAS (P < 0.001) but not in mutKRAS. High FIB-4 decreased RFS in mutKRAS (P = 0.001) but not in wtKRAS. FIB-4 was associated with a 10% increased recurrence risk. CONCLUSION TBS and FIB-4, alongside KRAS status, should be considered to improve outcome predictions.
Collapse
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Jun Kawashima
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Selamawit Woldesenbetm
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Amanda B Macedo
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Abdullah Altaf
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Federico Aucejo
- Department of General Surgery, Cleveland Clinic Foundation, OH, USA
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | | | - Kazunari Sasaki
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Yuki Imaoka
- Department of Surgery, Stanford University, Stanford, CA, USA
| | | | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
11
|
Selvam P, Tseng CH, Wang CT, Sun YY, Chen YL, Kao YT, Dahms HU, Cheng CM. 4-Anilinoquinolinylchalcone derivatives mediate antifibrotic effects through ERK/MRTF-a signaling pathway crosstalk. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2025; 32:11685-11696. [PMID: 40234319 DOI: 10.1007/s11356-025-36382-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Quinolones and their analogues are a remarkable group of drugs that have multiple impacts on the human immune system. They are suspected to mediate anti-cancer and anti-inflammatory responses. However, due to their effectiveness in treating a number of significant diseases, such as genitourinary cancer and breast cancer, as well as their antiangiogenic and immunomodulatory qualities, interest in this group of traditional medicines has recently increased. Unfortunately, numerous side effects were observed, such as diarrhea, skin rashes, nausea, vomiting, bleeding, and abnormal liver functions. To overcome these restrictions and to enhance the pharmacological profile, research efforts are focusing on the synthesis and optimization of novel quinolone analogues that lack severe side effects. The present study focuses on the mechanism of action and the signaling pathway involving the 4-anilinoquinolinylchalcone derivative. The objective of the present work was to better understand the mechanism by which anti-fibrosis is mediated by screening 6 synthesized 4-anilinoquinolinylchalcone derivatives for their potential as novel anti-fibrosis therapeutics.
Collapse
Affiliation(s)
- Padhmavathi Selvam
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan
| | - Chih Hua Tseng
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Fragrance & Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, 801, Taiwan
| | - Ching Tung Wang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan
| | - Yu-Yo Sun
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung City, 804, Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
| | - Yu-Tse Kao
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
| | - Hans-Uwe Dahms
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung City, 804, Taiwan
| | - Chih Mei Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
12
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00798-8. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Rajesh A, Gurusamy DS, Manikkam R. Evaluating the Tumor Burden, Histological Changes, and Immune Landscape of Breast Cancer Post-neoadjuvant Chemotherapy: Insights From 50 Cases. Cureus 2025; 17:e80258. [PMID: 40061852 PMCID: PMC11890627 DOI: 10.7759/cureus.80258] [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] [Accepted: 03/08/2025] [Indexed: 03/21/2025] Open
Abstract
Breast cancer is a heterogeneous disease with variable responses to neoadjuvant chemotherapy (NACT). Evaluating the histopathological and immune changes in post-NACT breast cancer specimens is crucial for understanding treatment response and guiding further management. This study aims to assess tumor burden using the Residual Cancer Burden (RCB) index, examine histological alterations, evaluate immune activity through tumor-infiltrating lymphocytes (TILs), and analyze proliferative capacity via Ki-67 expression in post-NACT breast cancer specimens. A cross-sectional study of 50 modified radical mastectomy (MRM) specimens post-NACT was conducted. The histopathological analysis included tumor regression changes, stromal and cellular alterations, and nodal involvement. Immune response was assessed by quantifying TILs, and proliferation was measured using the Ki-67 index. Statistical correlations were made between clinicopathological parameters, TILs, and Ki-67 expression. Residual disease was detected in 39 cases (78%), and 11 cases (22%) had no residual disease. Among the 39 cases with residual disease, the majority were classified as RCB II (22 cases, 56%), 16 cases (41%) were classified as RCB III, and one case (3%) was classified as RCB I. Common histological changes post-NACT included fibrosis in 31 cases (62%), necrosis in 19 cases (38%), and infiltration by foamy histiocytes in 16 cases (32%). Malignant epithelial cells more frequently exhibited foamy cytoplasm (16 cases (41%) vs. two cases (5%); p=0.0003), hyperchromatic nucleus (26 cases (67%) vs. six cases (15%); p=0.0001), and prominent nucleoli (26 cases (67%) vs. four cases (10%); p=0.0001) compared to benign cells. Among the 39 cases with residual disease, low TIL and high Ki-67 expression were observed in 20 cases (51%), while 12 cases (32%) showed high TIL and low Ki-67. Residual tumors with high TIL and high Ki-67 (four cases, 10%) and low TIL and low Ki-67 (three cases, 8%) were less common. A significant inverse relationship was found between TIL levels and Ki-67 expression (p=0.0002), as tumors with low TIL were more likely to have high Ki-67 expression (20 cases, 51%), whereas those with high TIL more frequently exhibited low Ki-67 expression (12 cases, 32%). Post-NACT evaluation of tumor burden, immune landscape, and proliferation provides valuable prognostic insights. Integrating RCB, TILs, and Ki-67 into routine pathological assessment may aid patient stratification and guide personalized treatment strategies. Further large-scale studies are needed to validate these findings and improve therapeutic decision-making in breast cancer management.
Collapse
Affiliation(s)
- Arasi Rajesh
- Pathology, Government Medical College, Tuticorin, IND
| | | | | |
Collapse
|
14
|
Zhou S, Zhao Z, Wang Z, Xu H, Li Y, Xu K, Li W, Yang J. Cancer-associated fibroblasts in carcinogenesis. J Transl Med 2025; 23:50. [PMID: 39806363 PMCID: PMC11727299 DOI: 10.1186/s12967-025-06071-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
In contemporary times, cancer poses the most significant threat to human life and safety. Scientists have relentlessly pursued the intricacies of carcinogenesis and explored ways to prevent and treat cancer. Carcinogenesis is a complex, multi-faceted, and multi-stage process, with numerous underlying causes, including inflammation and fibrosis. Cancer-associated fibroblasts (CAFs), however, occupy a pivotal and substantial role within the tumor microenvironment, facilitating carcinogenesis through diverse mechanisms such as creating inflammation, fostering a fibrotic tumor microenvironment, and immunosuppression. In this paper, we introduce the concept of carcinogenesis, explain its causes, describe the characteristics of CAFs and their sources, and highlight the roles and mechanisms of CAFs in promoting carcinogenesis. Ultimately, our aim is to contribute to the development of novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Shufen Zhou
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zekun Zhao
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojun Wang
- Department of Thyroid and Breast Surgery, The DingLi Clinical, The Wenzhou Central Hospital, College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hanzheng Xu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yijie Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China.
| | - Wei Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Jiahua Yang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
15
|
Dorjkhorloo G, Shiraishi T, Erkhem-Ochir B, Sohda M, Okami H, Yamaguchi A, Shioi I, Komine C, Nakazawa N, Shibasaki Y, Okada T, Osone K, Sano A, Sakai M, Ogawa H, Katayama A, Oyama T, Yokobori T, Shirabe K, Saeki H. High levels of fibrotic tumor components are associated with recurrence and intratumoral immune status in advanced colorectal cancer patients. Sci Rep 2024; 14:30735. [PMID: 39730445 DOI: 10.1038/s41598-024-80489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/19/2024] [Indexed: 12/29/2024] Open
Abstract
The importance of collagen and elastin remains incompletely understood concerning tumor immunity in cancer tissues. This study explored the clinical significance of collagen and elastin deposition on tumor immunity in advanced colorectal cancer patients. The collagen and elastin contents were assessed simultaneously using elastic van Gieson (EVG) histochemical staining. Immunohistochemical staining was performed to measure the immune cell markers CD3, CD8, CD86, and CD163 in surgically resected primary tumors from 78 pT4 colorectal cancer patients. High collagen, elastin, and EVG scores are associated with aggressive characteristics and short disease-free survival. A high EVG score was identified as an independent predictor of poor disease-free survival. Furthermore, tumors with high collagen and EVG scores exhibited significantly fewer intratumoral CD3 + and CD8 + cells. Evaluating tumor fibrosis using the classical and straightforward EVG staining method could be a reliable predictor of recurrence in high-risk colorectal cancer patients with tumor immune tolerance.
Collapse
Affiliation(s)
- Gendensuren Dorjkhorloo
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuya Shiraishi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Bilguun Erkhem-Ochir
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Haruka Okami
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Arisa Yamaguchi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ikuma Shioi
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Chika Komine
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Nobuhiro Nakazawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yuta Shibasaki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuhisa Okada
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Katsuya Osone
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Akihiko Sano
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sakai
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroomi Ogawa
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ayaka Katayama
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Takehiko Yokobori
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroshi Saeki
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
16
|
Trautmann T, Yakobian N, Nguyen R. CAR T-cells for pediatric solid tumors: where to go from here? Cancer Metastasis Rev 2024; 43:1445-1461. [PMID: 39317919 PMCID: PMC11554711 DOI: 10.1007/s10555-024-10214-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Despite the great success that chimeric antigen receptor (CAR) T-cells have had in patients with B-cell malignancies and multiple myeloma, they continue to have limited efficacy against most solid tumors. Especially in the pediatric population, pre- and post-treatment biopsies are rarely performed due to ethical reasons, and thus, our understanding is still very limited regarding the mechanisms in the tumor microenvironment by which tumor cells exclude effectors and attract immune-suppressive cells. Nevertheless, based on the principles that are known, current T-cell engineering has leveraged some of these processes and created more potent CAR T-cells. The recent discovery of new oncofetal antigens and progress made in CAR design have expanded the potential pool of candidate antigens for therapeutic development. The most promising approaches to enhance CAR T-cells are novel CAR gating strategies, creative ways of cytokine delivery to the TME without enhancing systemic toxicity, and hijacking the chemokine axis of tumors for migratory purposes. With these new modifications, the next step in the era of CAR T-cell development will be the clinical validation of these promising preclinical findings.
Collapse
Affiliation(s)
- Tina Trautmann
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Natalia Yakobian
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, NCI, NIH, NCI, 10 Center Drive, 1W-5832, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Jang Y, Kang S, Han H, Kang CM, Cho NH, Kim BG. Fibrosis-Encapsulated Tumoroid, A Solid Cancer Assembloid Model for Cancer Research and Drug Screening. Adv Healthc Mater 2024; 13:e2402391. [PMID: 39233539 PMCID: PMC11650424 DOI: 10.1002/adhm.202402391] [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: 06/30/2024] [Revised: 08/12/2024] [Indexed: 09/06/2024]
Abstract
Peritumoral fibrosis is known to promote cancer progression and confer treatment resistance in various solid tumors. Consequently, developing accurate cancer research and drug screening models that replicate the structure and function of a fibrosis-surrounded tumor mass is imperative. Previous studies have shown that self-assembly three-dimensional (3D) co-cultures primarily produce cancer-encapsulated fibrosis or maintain a fibrosis-encapsulated tumor mass for a short period, which is inadequate to replicate the function of fibrosis, particularly as a physical barrier. To address this limitation, a multi-layer spheroid formation method is developed to create a fibrosis-encapsulated tumoroid (FET) structure that maintains structural stability for up to 14 days. FETs exhibited faster tumor growth, higher expression of immunosuppressive cytokines, and equal or greater resistance to anticancer drugs compared to their parental tumoroids. Additionally, FETs serve as a versatile model for traditional cancer research, enabling the study of exosomal miRNA and gene functions, as well as for mechanobiology research when combined with alginate hydrogel. Our findings suggest that the FET represents an advanced model that more accurately mimics solid cancer tissue with peritumoral fibrosis. It may show potential superiority over self-assembly-based 3D co-cultures for cancer research and drug screening, and holds promise for personalized drug selection in cancer treatment.
Collapse
Affiliation(s)
- Yeonsue Jang
- Department of Urological Science InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Suki Kang
- Department of PathologyYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hyunho Han
- Department of Urological Science InstituteYonsei University College of MedicineSeoul03722Republic of Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of SurgeryYonsei University College of MedicineSeoul03722Republic of Korea
| | - Chang Moo Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of SurgeryYonsei University College of MedicineSeoul03722Republic of Korea
| | - Nam Hoon Cho
- Department of PathologyYonsei University College of MedicineSeoul03722Republic of Korea
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoul03722South Korea
| | - Baek Gil Kim
- Department of PathologyYonsei University College of MedicineSeoul03722Republic of Korea
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoul03722South Korea
| |
Collapse
|
18
|
Akabane M, Pawlik TM. ASO Author Reflections: Synergistic Role of FIB-4 Index and Tumor Burden Score in Predicting Recurrence After Hepatectomy for Intrahepatic Cholangiocarcinoma. Ann Surg Oncol 2024:10.1245/s10434-024-16536-7. [PMID: 39542965 DOI: 10.1245/s10434-024-16536-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
19
|
Akabane M, Kawashima J, Woldesenbet S, Macedo AB, Cauchy F, Shen F, Maithel SK, Groot Koerkamp B, Alexandrescu S, Kitago M, Weiss M, Martel G, Pulitano C, Aldrighetti L, Poultsides GA, Imaoka Y, Guglielmi A, Bauer TW, Endo I, Gleisner A, Marques HP, Pawlik TM. Improving Recurrence Prediction in Intrahepatic Cholangiocarcinoma: The Synergistic Impact of the FIB-4 Index and Tumor Burden Score on Post-hepatectomy Outcomes. Ann Surg Oncol 2024:10.1245/s10434-024-16455-7. [PMID: 39511008 DOI: 10.1245/s10434-024-16455-7] [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: 09/06/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND The prognostic role of the fibrosis-4 (FIB-4) index relative to intrahepatic cholangiocarcinoma (ICC) after hepatectomy remains unclear. This study sought to characterize the impact of the FIB-4 index and tumor burden score (TBS) on recurrence and overall survival (OS). METHODS ICC patients undergoing hepatectomy (2000-2020) were identified using a multi-institutional database. Patients were categorized as low (low TBS/low FIB-4 index), intermediate (low TBS/high FIB-4 index or high TBS/low FIB-4 index), and high (high TBS/high FIB-4 index). RESULTS Among 1168 patients in different TBS and FIB-4 index cohorts, 3-year recurrence varied considerably. For instance, among the patients with low TBS, individuals with a high FIB-4 index had a greater risk of recurrence than patients with a low FIB-4 index (59.9 vs. 47.7%; P = 0.01). Among patients with a high TBS, individuals with a high versus a low FIB-4 index had a higher incidence of recurrence (76.8 vs. 69.0%; P = 0.04). A similar pattern was observed among patients with both a low FIB-4 index (low [47.7%] vs. high [69.0%] TBS) and a high FIB-4 index (low [59.9%] vs. high [76.8%] TBS; both P < 0.001). Patients with a high [27.5%] versus a low [48.8%] TBS; P < 0.001) and patients with a high [34.2%] versus a low [43.5%] FIB-4 index; P = 0.01) had a worse OS. The multivariable analysis demonstrated an increasing risk of recurrence in the intermediate-index (hazard ratio [HR], 1.61; 95% confidence interval [CI], 1.20-2.16; P = 0.001) and high-index (HR, 2.13; 95% CI 1.45-3.13; P < 0.001) groups versus the low-index group. CONCLUSIONS Both tumor-related and non-tumorous characteristics should be used to predict risk of recurrence and survival more accurately among patients with ICC following hepatic resection.
Collapse
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Jun Kawashima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Selamawit Woldesenbet
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Amanda B Macedo
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - François Cauchy
- Department of Hepatobiliopancreatic Surgery, APHP, Beaujon Hospital, Clichy, France
| | - Feng Shen
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | | | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | - Matthew Weiss
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Guillaume Martel
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Carlo Pulitano
- Department of Surgery, Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia
| | | | | | - Yuki Imaoka
- Department of Surgery, Stanford University, Stanford, CA, USA
| | | | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Ana Gleisner
- Department of Surgery, University of Colorado, Denver, CO, USA
| | - Hugo P Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA.
- Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, Oncology, Health Services Management and Policy, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
20
|
Zhang J, Zang X, Jiao P, Wu J, Meng W, Zhao L, Lv Z. Alterations of Ceramides, Acylcarnitines, GlyceroLPLs, and Amines in NSCLC Tissues. J Proteome Res 2024; 23:4343-4358. [PMID: 39317643 DOI: 10.1021/acs.jproteome.4c00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Abnormal lipid metabolism plays an important role in cancer development. In this study, nontargeted lipidomic study on 230 tissue specimens from 79 nonsmall cell lung cancer (NSCLC) patients was conducted using ultraperformance liquid chromatography-high-resolution mass spectrometry (UPLC-HRMS). Downregulation of sphingosine and medium-long-chain ceramides and short-medium-chain acylcarnitine, upregulation of long-chain acylcarnitine C20:0, and enhanced histamine methylation were revealed in NSCLC tissues. Compared with paired noncancerous tissues, adenocarcinoma (AC) tissues had significantly decreased levels of sphingosine, medium-long-chain ceramides (Cer d18:1/12:0 and Cer d16:1/14:0, Cer d18:0/16:0, Cer d18:1/16:0, Cer d18:2/16:0, Cer d18:2/18:0), short-medium-chain (C2-C16) acylcarnitines, LPC 20:0 and LPC 22:1, and significantly increased levels of the long-chain acylcarnitine C20:0, LPC 16:0, LPC P-16:0, LPC 20:1, LPC 20:2, glyceroPC, LPE 16:0, and LPE 18:2. In squamous cell carcinoma (SCC) tissues, sphingosine, Cer d18:2/16:0 and Cer d18:2/18:0, and short-medium-chain acylcarnitines had significantly lower levels, while long-chain acylcarnitines (C20:0, and C22:0 or C22:0 M), LPC 20:1, LPC 20:2, and N1,N12-diacetylspermine had significantly higher levels compared to controls. In AC and SCC tissues, the levels of LPG 18:0, LPG 18:1, and LPS 18:1 were significantly decreased, while the levels of ceramide-1-phosphate (C1P) d18:0/3:0 or LPE P-16:0, N1-acetylspermidine, and 1-methylhistamine were significantly increased than controls. Furthermore, an orthogonal partial least-squares-discriminant analysis (OPLS-DA) model based on a 4-lipid panel was established, showing good discrimination ability between cancerous and noncancerous tissues.
Collapse
Affiliation(s)
- Jie Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
| | - Xiaoling Zang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
- Qingdao Marine Science and Technology Center, Qingdao, Shandong 266235, P. R. China
| | - Peng Jiao
- Department of Thoracic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Jiangyu Wu
- Department of Thoracic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Wei Meng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
| | - Lizhen Zhao
- College of Physics, Qingdao University, Qingdao, Shandong 266071, P. R. China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
- Qingdao Marine Science and Technology Center, Qingdao, Shandong 266235, P. R. China
| |
Collapse
|
21
|
Li Y, Huang X, Li Y, Qiao Q, Chen C, Chen Y, Zhong W, Liu H, Sun T. WRN Nuclease-Mediated EcDNA Clearance Enhances Antitumor Therapy in Conjunction with Trehalose Dimycolate/Mesoporous Silica Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407026. [PMID: 39206698 PMCID: PMC11516056 DOI: 10.1002/advs.202407026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Current research on tumor fibrosis has focused on cancer-associated fibroblasts, which may exert dual functions of tumor promotion and inhibition. Little attention has been paid to whether tumor cells themselves can undergo fibrotic transformation and whether they can inhibit parenchymal cells similar to pulmonary fibrosis, thus achieving the goal of inhibiting the malignant progression of tumors. To explore the significance of inducing tumor fibrosis for cancer treatment. This study utilizes mesoporous silica nanoparticles (MSN) loaded with Trehalose dimycolate (TDM) to induce tumor cell fibrosis through the dual effects of TDM-induced inflammatory granuloma and MSN-induced foreign body granuloma. The results show that TDM/MSN (TM) can effectively induce tumor fibrosis, manifested specifically by collagen internalization, and suppression of proliferation and invasion capabilities, suggesting the potential role of tumor fibrosis therapy. However, further investigation reveals that extrachromosomal DNA (ecDNA) mediates resistance to fibrosis induction. To comprehensively enhance the efficacy, WRN exonuclease is conjugated to TM to form new nanoparticles (TMW) capable of effectively eliminating ecDNA, globally promoting tumor cell fibroblast-like transformation, and validated in a PDX model to inhibit cancer progression. Therefore, TMW, through inducing tumor cell fibrosis to inhibit its malignant progression, holds great potential as a clinical treatment strategy.
Collapse
Affiliation(s)
- Yinan Li
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
| | - Xiu Huang
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative DrugsTianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjin300450China
| | - Yingying Li
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative DrugsTianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjin300450China
| | - Qingqing Qiao
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
| | - Caihong Chen
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
| | - Yang Chen
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
| | - Weilong Zhong
- Tianjin Key Laboratory of Digestive DiseasesDepartment of Gastroenterology and HepatologyTianjin Institute of Digestive DiseasesTianjin Medical University General HospitalTianjin300052China
| | - Huijuan Liu
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative DrugsTianjin Key Laboratory of Molecular Drug ResearchTianjin International Joint Academy of BiomedicineTianjin300450China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjin300350China
| |
Collapse
|
22
|
Hernandez-Padilla C, Joosten B, Franco A, Cambi A, van den Dries K, Nain AS. Dendritic cell force-migration coupling on aligned fiber networks. Biophys J 2024; 123:3120-3132. [PMID: 38993114 PMCID: PMC11427780 DOI: 10.1016/j.bpj.2024.07.011] [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: 08/21/2023] [Revised: 01/12/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that reside in peripheral tissues and are responsible for initiating adaptive immune responses. As gatekeepers of the immune system, DCs need to continuously explore their surroundings, for which they can rapidly move through various types of connective tissue and basement membranes. DC motility has been extensively studied on flat 2D surfaces, yet the influences of a contextual 3D fibrous environment still need to be described. Using ECM-mimicking suspended fiber networks, we show how immature DCs (iDCs) engage in migratory cycles that allow them to transition from persistent migration to slow migratory states. For a subset of iDCs with high migratory potential, we report the organization of protrusions at the front of the cell body, which reverses upon treatment with inflammation agent PGE2. We identify an unusual migratory response to aligned fiber networks, whereby iDCs use filamentous protrusions to attach laterally and exert forces on fibers to migrate independent of fiber alignment. Increasing the fiber diameter from 200 to 500 nm does not significantly affect the migratory response; however, iDCs respond by forming denser actin bundles around larger diameters. Overall, the correlation between force-coupling and random migration of iDCs in aligned fibrous topography offers new insights into how iDCs might move in fibrous environments in vivo.
Collapse
Affiliation(s)
| | - Ben Joosten
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aime Franco
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Koen van den Dries
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Amrinder S Nain
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia.
| |
Collapse
|
23
|
Migita T. Is End-Stage Renal Disease Tumor Suppressive? Dispelling the Myths. Cancers (Basel) 2024; 16:3135. [PMID: 39335107 PMCID: PMC11430482 DOI: 10.3390/cancers16183135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
The prevalence of end-stage renal disease is increasing worldwide. Malignancies accompanying end-stage renal disease are detected in approximately 120 individuals per 10,000 person-years. Most studies have suggested that end-stage renal disease causes carcinogenesis and promotes tumor development; however, this theory remains questionable. Contrary to the theory that end-stage renal disease is predominantly carcinogenic, recent findings have suggested that after controlling for biases and sampling errors, the overall cancer risk in patients with end-stage renal disease might be lower than that in the general population, except for renal and urothelial cancer risks. Additionally, mortality rates associated with most cancers are lower in patients with end-stage renal disease than in the general population. Several biological mechanisms have been proposed to explain the anticancer effects of end-stage renal disease, including premature aging and senescence, enhanced cancer immunity, uremic tumoricidal effects, hormonal and metabolic changes, and dialysis therapy-related factors. Despite common beliefs that end-stage renal disease exacerbates cancer risk, emerging evidence suggests potential tumor-suppressive effects. This review highlights the potential anticancer effects of end-stage renal disease, proposing reconsideration of the hypothesis that end-stage renal disease promotes cancer development and progression.
Collapse
Affiliation(s)
- Toshiro Migita
- Tokyo Nephrology Clinic, Tokyo 170-0003, Japan; ; Tel.: +81-3-3949-5801
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Medical Laboratory Sciences, Kitasato University, Kanagawa 252-0373, Japan
| |
Collapse
|
24
|
Watson SS, Zomer A, Fournier N, Lourenco J, Quadroni M, Chryplewicz A, Nassiri S, Aubel P, Avanthay S, Croci D, Abels E, Broekman MLD, Hanahan D, Huse JT, Daniel RT, Hegi ME, Homicsko K, Cossu G, Hottinger AF, Joyce JA. Fibrotic response to anti-CSF-1R therapy potentiates glioblastoma recurrence. Cancer Cell 2024; 42:1507-1527.e11. [PMID: 39255775 DOI: 10.1016/j.ccell.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 06/06/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Abstract
Glioblastoma recurrence is currently inevitable despite extensive standard-of-care treatment. In preclinical studies, an alternative strategy of targeting tumor-associated macrophages and microglia through CSF-1R inhibition was previously found to regress established tumors and significantly increase overall survival. However, recurrences developed in ∼50% of mice in long-term studies, which were consistently associated with fibrotic scars. This fibrotic response is observed following multiple anti-glioma therapies in different preclinical models herein and in patient recurrence samples. Multi-omics analyses of the post-treatment tumor microenvironment identified fibrotic areas as pro-tumor survival niches that encapsulated surviving glioma cells, promoted dormancy, and inhibited immune surveillance. The fibrotic treatment response was mediated by perivascular-derived fibroblast-like cells via activation by transforming growth factor β (TGF-β) signaling and neuroinflammation. Concordantly, combinatorial inhibition of these pathways inhibited treatment-associated fibrosis, and significantly improved survival in preclinical trials of anti-colony-stimulating factor-1 receptor (CSF-1R) therapy.
Collapse
Affiliation(s)
- Spencer S Watson
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Anoek Zomer
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Translational Data Science Facility, SIB Swiss Institute of Bioinformatics, Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland
| | - Joao Lourenco
- Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Translational Data Science Facility, SIB Swiss Institute of Bioinformatics, Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland
| | - Manfredo Quadroni
- Proteomics Core Facility, University of Lausanne, 1011 Lausanne, Switzerland
| | - Agnieszka Chryplewicz
- Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Sina Nassiri
- Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Translational Data Science Facility, SIB Swiss Institute of Bioinformatics, Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland
| | - Pauline Aubel
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Simona Avanthay
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Davide Croci
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Erik Abels
- Department of Neurosurgery, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Department of Neurosurgery, Haaglanden Medical Center, 2597 The Hague, the Netherlands
| | - Marike L D Broekman
- Department of Neurosurgery, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Department of Neurosurgery, Haaglanden Medical Center, 2597 The Hague, the Netherlands
| | - Douglas Hanahan
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne, Geneva, Switzerland
| | - Jason T Huse
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roy T Daniel
- Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Department of Neurosurgery, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| | - Monika E Hegi
- Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Department of Clinical Neurosciences, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Krisztian Homicsko
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| | - Giulia Cossu
- Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Department of Oncology, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Lundin Brain Tumour Centre, University Hospital Lausanne, 1011 Lausanne, Switzerland; Swiss Cancer Center Leman (SCCL), Lausanne, Geneva, Switzerland.
| |
Collapse
|
25
|
Di Carlo E, Sorrentino C. The multifaceted role of the stroma in the healthy prostate and prostate cancer. J Transl Med 2024; 22:825. [PMID: 39238004 PMCID: PMC11378418 DOI: 10.1186/s12967-024-05564-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
Prostate cancer (PC) is an age-related disease and represents, after lung cancer, the second cause of cancer death in males worldwide. Mortality is due to the metastatic disease, which mainly involves the bones, lungs, and liver. In the last 20 years, the incidence of metastatic PC has increased in Western Countries, and a further increase is expected in the near future, due to the population ageing. Current treatment options, including state of the art cancer immunotherapy, need to be more effective to achieve long-term disease control. The most significant anatomical barrier to overcome to improve the effectiveness of current and newly designed drug strategies consists of the prostatic stroma, in particular the fibroblasts and the extracellular matrix, which are the most abundant components of both the normal and tumor prostatic microenvironment. By weaving a complex communication network with the glandular epithelium, the immune cells, the microbiota, the endothelium, and the nerves, in the healthy prostatic microenvironment, the fibroblasts and the extracellular matrix support organ development and homeostasis. However, during inflammation, ageing and prostate tumorigenesis, they undergo dramatic phenotypic and genotypic changes, which impact on tumor growth and progression and on the development of therapy resistance. Here, we focus on the characteristics and functions of the prostate associated fibroblasts and of the extracellular matrix in health and cancer. We emphasize their roles in shaping tumor behavior and the feasibility of manipulating and/or targeting these stromal components to overcome the limitations of current treatments and to improve precision medicine's chances of success.
Collapse
Affiliation(s)
- Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti- Pescara, Via dei Vestini, Chieti, 66100, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, Chieti, 66100, Italy.
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti- Pescara, Via dei Vestini, Chieti, 66100, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, Chieti, 66100, Italy
| |
Collapse
|
26
|
Adi W, Rubio Perez BE, Liu Y, Runkle S, Eliceiri KW, Yesilkoy F. Machine learning-assisted mid-infrared spectrochemical fibrillar collagen imaging in clinical tissues. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:093511. [PMID: 39364328 PMCID: PMC11448345 DOI: 10.1117/1.jbo.29.9.093511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Significance Label-free multimodal imaging methods that can provide complementary structural and chemical information from the same sample are critical for comprehensive tissue analyses. These methods are specifically needed to study the complex tumor-microenvironment where fibrillar collagen's architectural changes are associated with cancer progression. To address this need, we present a multimodal computational imaging method where mid-infrared spectral imaging (MIRSI) is employed with second harmonic generation (SHG) microscopy to identify fibrillar collagen in biological tissues. Aim To demonstrate a multimodal approach where a morphology-specific contrast mechanism guides an MIRSI method to detect fibrillar collagen based on its chemical signatures. Approach We trained a supervised machine learning (ML) model using SHG images as ground truth collagen labels to classify fibrillar collagen in biological tissues based on their mid-infrared hyperspectral images. Five human pancreatic tissue samples (sizes are in the order of millimeters) were imaged by both MIRSI and SHG microscopes. In total, 2.8 million MIRSI spectra were used to train a random forest (RF) model. The other 68 million spectra were used to validate the collagen images generated by the RF-MIRSI model in terms of collagen segmentation, orientation, and alignment. Results Compared with the SHG ground truth, the generated RF-MIRSI collagen images achieved a high average boundary F -score (0.8 at 4-pixel thresholds) in the collagen distribution, high correlation (Pearson's R 0.82) in the collagen orientation, and similarly high correlation (Pearson's R 0.66) in the collagen alignment. Conclusions We showed the potential of ML-aided label-free mid-infrared hyperspectral imaging for collagen fiber and tumor microenvironment analysis in tumor pathology samples.
Collapse
Affiliation(s)
- Wihan Adi
- University of Wisconsin-Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Bryan E. Rubio Perez
- University of Wisconsin-Madison, Department of Electrical and Computer Engineering, Madison, Wisconsin, United States
| | - Yuming Liu
- University of Wisconsin-Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
| | - Sydney Runkle
- University of Wisconsin-Madison, Department of Computer Science, Madison, Wisconsin, United States
| | - Kevin W. Eliceiri
- University of Wisconsin-Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- University of Wisconsin-Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Filiz Yesilkoy
- University of Wisconsin-Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| |
Collapse
|
27
|
Zhou X, LeBleu VS, Fletcher-Sananikone E, Kim J, Dai J, Li B, Wu CC, Sugimoto H, Miyake T, Becker LM, Volpert OV, Lawson E, Espinosa Da Silva C, Patel SI, Kizu A, Ehsanipour EA, Sha D, Karam JA, McAndrews KM, Kalluri R. Vascular heterogeneity of tight junction Claudins guides organotropic metastasis. NATURE CANCER 2024; 5:1371-1389. [PMID: 39289595 PMCID: PMC11987010 DOI: 10.1038/s43018-024-00813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 07/23/2024] [Indexed: 09/19/2024]
Abstract
Carcinomas are associated with metastasis to specific organs while sparing others. Breast cancer presents with lung metastasis but rarely kidney metastasis. Using this difference as an example, we queried the mechanism(s) behind the proclivity for organ-specific metastasis. We used spontaneous and implant models of metastatic mammary carcinoma coupled with inflammatory tissue fibrosis, single-cell sequencing analyses and functional studies to unravel the causal determinants of organ-specific metastasis. Here we show that lung metastasis is facilitated by angiopoietin 2 (Ang2)-mediated suppression of lung-specific endothelial tight junction protein Claudin 5, which is augmented by the inflammatory fibrotic microenvironment and prevented by anti-Ang2 blocking antibodies, while kidney metastasis is prevented by non-Ang2-responsive Claudins 2 and 10. Suppression of Claudins 2 and 10 was sufficient to induce the emergence of kidney metastasis. This study illustrates the influence of organ-specific vascular heterogeneity in determining organotropic metastasis, independent of cancer cell-intrinsic mechanisms.
Collapse
Affiliation(s)
- Xunian Zhou
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Eliot Fletcher-Sananikone
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiha Kim
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianli Dai
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bingrui Li
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chia-Chin Wu
- Department of Genomic Medicine, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Toru Miyake
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisa M Becker
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Olga V Volpert
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erica Lawson
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Espinosa Da Silva
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah I Patel
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Akane Kizu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ehsan A Ehsanipour
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Di Sha
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jose Antonio Karam
- Department of Urology, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kathleen M McAndrews
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
28
|
Graham MK, Wang R, Chikarmane R, Abel B, Vaghasia A, Gupta A, Zheng Q, Hicks J, Sysa-Shah P, Pan X, Castagna N, Liu J, Meyers J, Skaist A, Zhang Y, Rubenstein M, Schuebel K, Simons BW, Bieberich CJ, Nelson WG, Lupold SE, DeWeese TL, De Marzo AM, Yegnasubramanian S. Convergent alterations in the tumor microenvironment of MYC-driven human and murine prostate cancer. Nat Commun 2024; 15:7414. [PMID: 39198404 PMCID: PMC11358296 DOI: 10.1038/s41467-024-51450-2] [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: 09/29/2023] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
How prostate cancer cells and their precursors mediate changes in the tumor microenvironment (TME) to drive prostate cancer progression is unclear, in part due to the inability to longitudinally study the disease evolution in human tissues. To overcome this limitation, we perform extensive single-cell RNA-sequencing (scRNA-seq) and molecular pathology of the comparative biology between human prostate cancer and key stages in the disease evolution of a genetically engineered mouse model (GEMM) of prostate cancer. Our studies of human tissues reveal that cancer cell-intrinsic activation of MYC signaling is a common denominator across the well-known molecular and pathological heterogeneity of human prostate cancer. Cell communication network and pathway analyses in GEMMs show that MYC oncogene-expressing neoplastic cells, directly and indirectly, reprogram the TME during carcinogenesis, leading to a convergence of cell state alterations in neighboring epithelial, immune, and fibroblast cell types that parallel key findings in human prostate cancer.
Collapse
Affiliation(s)
- Mindy K Graham
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Rulin Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Roshan Chikarmane
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Bulouere Abel
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Ajay Vaghasia
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Anuj Gupta
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Qizhi Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jessica Hicks
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Polina Sysa-Shah
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Xin Pan
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Nicole Castagna
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jianyong Liu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jennifer Meyers
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Alyza Skaist
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Yan Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Michael Rubenstein
- Department of Biological Sciences, University of Maryland at Baltimore County, Baltimore, MD, USA
| | - Kornel Schuebel
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Brian W Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Charles J Bieberich
- Department of Biological Sciences, University of Maryland at Baltimore County, Baltimore, MD, USA
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Shawn E Lupold
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Theodore L DeWeese
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- inHealth Precision Medicine Program, Johns Hopkins Medicine, Baltimore, MD, USA.
| |
Collapse
|
29
|
Chong X, Madeti Y, Cai J, Li W, Cong L, Lu J, Mo L, Liu H, He S, Yu C, Zhou Z, Wang B, Cao Y, Wang Z, Shen L, Wang Y, Zhang X. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol 2024; 17:65. [PMID: 39123202 PMCID: PMC11316403 DOI: 10.1186/s13045-024-01578-x] [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: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The past few decades have witnessed the rise of immunotherapy for Gastrointestinal (GI) tract cancers. The role of immune checkpoint inhibitors (ICIs), particularly programmed death protein 1 (PD-1) and PD ligand-1 antibodies, has become increasingly pivotal in the treatment of advanced and perioperative GI tract cancers. Currently, anti-PD-1 plus chemotherapy is considered as first-line regimen for unselected advanced gastric/gastroesophageal junction adenocarcinoma (G/GEJC), mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer (CRC), and advanced esophageal cancer (EC). In addition, the encouraging performance of claudin18.2-redirected chimeric antigen receptor T-cell (CAR-T) therapy in later-line GI tract cancers brings new hope for cell therapy in solid tumour treatment. Nevertheless, immunotherapy for GI tumour remains yet precise, and researchers are dedicated to further maximising and optimising the efficacy. This review summarises the important research, latest progress, and future directions of immunotherapy for GI tract cancers including EC, G/GEJC, and CRC.
Collapse
Affiliation(s)
- Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yelizhati Madeti
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Wenfei Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jialin Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Liyang Mo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Huizhen Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Siyi He
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Chao Yu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhiruo Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Boya Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
30
|
Fiehn F, Beisel C, Binder M. Hepatitis C virus and hepatocellular carcinoma: carcinogenesis in the era of direct-acting antivirals. Curr Opin Virol 2024; 67:101423. [PMID: 38925094 DOI: 10.1016/j.coviro.2024.101423] [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: 05/07/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024]
Abstract
Chronic hepatitis C virus (HCV) infection is a major cause of hepatic fibrosis and cirrhosis, with a risk for the development of hepatocellular carcinoma (HCC). Although highly effective direct-acting antivirals (DAAs) are available, the incidence, morbidity, and mortality of HCV-associated HCC are still high. This article reviews the current knowledge of the mechanisms of HCV-induced carcinogenesis with a special focus on those processes that continue after virus clearance and outlines implications for patient surveillance after DAA treatment.
Collapse
Affiliation(s)
- Felix Fiehn
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (D430), German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Claudia Beisel
- Department of Internal Medicine IV, Gastroenterology and Infectious Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Marco Binder
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (D430), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
31
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
32
|
Das M, Teli P, Vaidya A, Kale V. Expression of CD45 in non-hematopoietic cells: implications in regenerative medicine and disease management. Regen Med 2024; 19:407-419. [PMID: 39058408 PMCID: PMC11370962 DOI: 10.1080/17460751.2024.2378627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
CD45 plays a crucial role in the regulation of hematopoiesis. However, a comprehensive understanding of its role in non-hematopoietic cells is lacking. Several tissue precursors express CD45, indicating its crucial role in tissue regeneration. These precursors would fall prey to the recent therapies involving CD45 as a target. CD45+ double-positive tumor cells contribute to cancer progression, but whether CD45 is involved in the process needs to be investigated. Recently, we showed that aging induces CD45 expression in mesenchymal stromal cells and affects their differentiation potential. In this review, we, for the first time, unravel the important implications of the expression of CD45 in non-hematopoietic cells and provide novel insights into its potential therapeutic target in regenerative medicine and disease management.
Collapse
Affiliation(s)
- Madhurima Das
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India
| | - Prajakta Teli
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India
| |
Collapse
|
33
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis part III: genitourinary system. J Transl Med 2024; 22:616. [PMID: 38961396 PMCID: PMC11223291 DOI: 10.1186/s12967-024-05333-1] [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: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Fibrosis is a pathological process involving the abnormal deposition of connective tissue, resulting from improper tissue repair in response to sustained injury caused by hypoxia, infection, or physical damage. It can impact any organ, leading to their dysfunction and eventual failure. Additionally, tissue fibrosis plays an important role in carcinogenesis and the progression of cancer.Early and accurate diagnosis of organ fibrosis, coupled with regular surveillance, is essential for timely disease-modifying interventions, ultimately reducing mortality and enhancing quality of life. While extensive research has already been carried out on the topics of aberrant wound healing and fibrogenesis, we lack a thorough understanding of how their relationship reveals itself through modern imaging techniques.This paper focuses on fibrosis of the genito-urinary system, detailing relevant imaging technologies used for its detection and exploring future directions.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
34
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis part II: abdominal organs. J Transl Med 2024; 22:610. [PMID: 38956593 PMCID: PMC11218138 DOI: 10.1186/s12967-024-05346-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: 02/12/2024] [Accepted: 05/25/2024] [Indexed: 07/04/2024] Open
Abstract
Fibrosis is the aberrant process of connective tissue deposition from abnormal tissue repair in response to sustained tissue injury caused by hypoxia, infection, or physical damage. It can affect almost all organs in the body causing dysfunction and ultimate organ failure. Tissue fibrosis also plays a vital role in carcinogenesis and cancer progression. The early and accurate diagnosis of organ fibrosis along with adequate surveillance are helpful to implement early disease-modifying interventions, important to reduce mortality and improve quality of life. While extensive research has already been carried out on the topic, a thorough understanding of how this relationship reveals itself using modern imaging techniques has yet to be established. This work outlines the ways in which fibrosis shows up in abdominal organs and has listed the most relevant imaging technologies employed for its detection. New imaging technologies and developments are discussed along with their promising applications in the early detection of organ fibrosis.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
35
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis. Part I: Thoracic organs. J Transl Med 2024; 22:609. [PMID: 38956586 PMCID: PMC11218337 DOI: 10.1186/s12967-024-05244-1] [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: 02/12/2024] [Accepted: 04/27/2024] [Indexed: 07/04/2024] Open
Abstract
Sustained injury from factors such as hypoxia, infection, or physical damage may provoke improper tissue repair and the anomalous deposition of connective tissue that causes fibrosis. This phenomenon may take place in any organ, ultimately leading to their dysfunction and eventual failure. Tissue fibrosis has also been found to be central in both the process of carcinogenesis and cancer progression. Thus, its prompt diagnosis and regular monitoring is necessary for implementing effective disease-modifying interventions aiming to reduce mortality and improve overall quality of life. While significant research has been conducted on these subjects, a comprehensive understanding of how their relationship manifests through modern imaging techniques remains to be established. This work intends to provide a comprehensive overview of imaging technologies relevant to the detection of fibrosis affecting thoracic organs as well as to explore potential future advancements in this field.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
36
|
Mancini A, Gentile MT, Pentimalli F, Cortellino S, Grieco M, Giordano A. Multiple aspects of matrix stiffness in cancer progression. Front Oncol 2024; 14:1406644. [PMID: 39015505 PMCID: PMC11249764 DOI: 10.3389/fonc.2024.1406644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
The biophysical and biomechanical properties of the extracellular matrix (ECM) are crucial in the processes of cell differentiation and proliferation. However, it is unclear to what extent tumor cells are influenced by biomechanical and biophysical changes of the surrounding microenvironment and how this response varies between different tumor forms, and over the course of tumor progression. The entire ensemble of genes encoding the ECM associated proteins is called matrisome. In cancer, the ECM evolves to become highly dysregulated, rigid, and fibrotic, serving both pro-tumorigenic and anti-tumorigenic roles. Tumor desmoplasia is characterized by a dramatic increase of α-smooth muscle actin expressing fibroblast and the deposition of hard ECM containing collagen, fibronectin, proteoglycans, and hyaluronic acid and is common in many solid tumors. In this review, we described the role of inflammation and inflammatory cytokines, in desmoplastic matrix remodeling, tumor state transition driven by microenvironment forces and the signaling pathways in mechanotransduction as potential targeted therapies, focusing on the impact of qualitative and quantitative variations of the ECM on the regulation of tumor development, hypothesizing the presence of matrisome drivers, acting alongside the cell-intrinsic oncogenic drivers, in some stages of neoplastic progression and in some tumor contexts, such as pancreatic carcinoma, breast cancer, lung cancer and mesothelioma.
Collapse
Affiliation(s)
- Alessandro Mancini
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- BioUp Sagl, Lugano, Switzerland
| | - Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University “Giuseppe De Gennaro,” Casamassima, Bari, Italy
| | - Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, NA, Italy
- Sbarro Health Research Organization (S.H.R.O.) Italia Foundation ETS, Candiolo, TO, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
37
|
Wang X, Chen B, Zhang H, Peng L, Liu X, Zhang Q, Wang X, Peng S, Wang K, Liao L. Integrative analysis identifies molecular features of fibroblast and the significance of fibrosis on neoadjuvant chemotherapy response in breast cancer. Int J Surg 2024; 110:4083-4095. [PMID: 38546506 PMCID: PMC11254208 DOI: 10.1097/js9.0000000000001360] [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: 12/12/2023] [Accepted: 03/03/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND The molecular features of fibroblasts and the role of fibrosis in neoadjuvant chemotherapy (NAC) response and breast cancer (BRCA) prognosis remain unclear. Therefore, this study aimed to investigate the impact of interstitial fibrosis on the response and prognosis of patients with BRCA undergoing NAC treatment. MATERIALS AND METHODS The molecular characteristics of pathologic complete response (pCR) and non-pCR (npCR) in patients with BRCA were analyzed using multiomics analysis. A clinical cohort was collected to investigate the predictive value of fibrosis in patients with BRCA. RESULTS Fibrosis-related signaling pathways were significantly upregulated in patients with npCR. npCR may be associated with distinct and highly active fibroblast subtypes. Patients with high fibrosis had lower pCR rates. The fibrosis-dependent nomogram for pCR showed efficient predictive ability [training set: area under the curve [AUC]=0.871, validation set: AUC=0.792]. Patients with low fibrosis had a significantly better prognosis than those with high fibrosis, and those with a high fibrotic focus index had significantly shorter overall and recurrence-free survival. Therefore, fibrosis can be used to predict pCR. Our findings provide a basis for decision-making in the treatment of BRCA. CONCLUSIONS npCR is associated with a distinct and highly active fibroblast subtype. Furthermore, patients with high fibrosis have lower pCR rates and shorter long-term survival. Therefore, fibrosis can predict pCR. A nomogram that includes fibrosis can provide a basis for decision-making in the treatment of BRCA.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital
- Clinical Research Center For Breast Cancer In Hunan Province
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, People’s Republic of China
| | - Hanghao Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Clinical Research Center For Breast Cancer In Hunan Province
| | - Lushan Peng
- Department of Pathology, Xiangya Hospital, Central South University
| | - Xiangyan Liu
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Clinical Research Center For Breast Cancer In Hunan Province
| | - Qian Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Clinical Research Center For Breast Cancer In Hunan Province
| | - Xiaoxiao Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Clinical Research Center For Breast Cancer In Hunan Province
| | - Shuai Peng
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Clinical Research Center For Breast Cancer In Hunan Province
| | - Kuangsong Wang
- Department of Pathology, Xiangya Hospital, Central South University
| | - Liqiu Liao
- Department of Breast Surgery, Xiangya Hospital, Central South University
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University
- Clinical Research Center For Breast Cancer In Hunan Province
| |
Collapse
|
38
|
Ashworth JC, Cox TR. The importance of 3D fibre architecture in cancer and implications for biomaterial model design. Nat Rev Cancer 2024; 24:461-479. [PMID: 38886573 DOI: 10.1038/s41568-024-00704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 06/20/2024]
Abstract
The need for improved prediction of clinical response is driving the development of cancer models with enhanced physiological relevance. A new concept of 'precision biomaterials' is emerging, encompassing patient-mimetic biomaterial models that seek to accurately detect, treat and model cancer by faithfully recapitulating key microenvironmental characteristics. Despite recent advances allowing tissue-mimetic stiffness and molecular composition to be replicated in vitro, approaches for reproducing the 3D fibre architectures found in tumour extracellular matrix (ECM) remain relatively unexplored. Although the precise influences of patient-specific fibre architecture are unclear, we summarize the known roles of tumour fibre architecture, underlining their implications in cell-matrix interactions and ultimately clinical outcome. We then explore the challenges in reproducing tissue-specific 3D fibre architecture(s) in vitro, highlighting relevant biomaterial fabrication techniques and their benefits and limitations. Finally, we discuss imaging and image analysis techniques (focussing on collagen I-optimized approaches) that could hold the key to mapping tumour-specific ECM into high-fidelity biomaterial models. We anticipate that an interdisciplinary approach, combining materials science, cancer research and image analysis, will elucidate the role of 3D fibre architecture in tumour development, leading to the next generation of patient-mimetic models for mechanistic studies and drug discovery.
Collapse
Affiliation(s)
- Jennifer C Ashworth
- School of Veterinary Medicine & Science, Sutton Bonington Campus, University of Nottingham, Leicestershire, UK.
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK.
- Cancer Ecosystems Program, The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
| | - Thomas R Cox
- Cancer Ecosystems Program, The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia.
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
39
|
Shender VO, Anufrieva KS, Shnaider PV, Arapidi GP, Pavlyukov MS, Ivanova OM, Malyants IK, Stepanov GA, Zhuravlev E, Ziganshin RH, Butenko IO, Bukato ON, Klimina KM, Veselovsky VA, Grigorieva TV, Malanin SY, Aleshikova OI, Slonov AV, Babaeva NA, Ashrafyan LA, Khomyakova E, Evtushenko EG, Lukina MM, Wang Z, Silantiev AS, Nushtaeva AA, Kharlampieva DD, Lazarev VN, Lashkin AI, Arzumanyan LK, Petrushanko IY, Makarov AA, Lebedeva OS, Bogomazova AN, Lagarkova MA, Govorun VM. Therapy-induced secretion of spliceosomal components mediates pro-survival crosstalk between ovarian cancer cells. Nat Commun 2024; 15:5237. [PMID: 38898005 PMCID: PMC11187153 DOI: 10.1038/s41467-024-49512-6] [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: 02/04/2022] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
Ovarian cancer often develops resistance to conventional therapies, hampering their effectiveness. Here, using ex vivo paired ovarian cancer ascites obtained before and after chemotherapy and in vitro therapy-induced secretomes, we show that molecules secreted by ovarian cancer cells upon therapy promote cisplatin resistance and enhance DNA damage repair in recipient cancer cells. Even a short-term incubation of chemonaive ovarian cancer cells with therapy-induced secretomes induces changes resembling those that are observed in chemoresistant patient-derived tumor cells after long-term therapy. Using integrative omics techniques, we find that both ex vivo and in vitro therapy-induced secretomes are enriched with spliceosomal components, which relocalize from the nucleus to the cytoplasm and subsequently into the extracellular vesicles upon treatment. We demonstrate that these molecules substantially contribute to the phenotypic effects of therapy-induced secretomes. Thus, SNU13 and SYNCRIP spliceosomal proteins promote therapy resistance, while the exogenous U12 and U6atac snRNAs stimulate tumor growth. These findings demonstrate the significance of spliceosomal network perturbation during therapy and further highlight that extracellular signaling might be a key factor contributing to the emergence of ovarian cancer therapy resistance.
Collapse
Affiliation(s)
- Victoria O Shender
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russian Federation.
| | - Ksenia S Anufrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Polina V Shnaider
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
- Faculty of Biology; Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Georgij P Arapidi
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russian Federation
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, 141701, Russian Federation
| | - Marat S Pavlyukov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russian Federation
| | - Olga M Ivanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Irina K Malyants
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
- Faculty of Chemical-Pharmaceutical Technologies and Biomedical Drugs, Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation
| | - Grigory A Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, 630090, Russian Federation
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Evgenii Zhuravlev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, 630090, Russian Federation
| | - Rustam H Ziganshin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russian Federation
| | - Ivan O Butenko
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Olga N Bukato
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Ksenia M Klimina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Vladimir A Veselovsky
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | | | | | - Olga I Aleshikova
- National Medical Scientific Centre of Obstetrics, Gynaecology and Perinatal Medicine named after V.I. Kulakov, Moscow, 117198, Russian Federation
- Russian Research Center of Roentgenology and Radiology, Moscow, 117997, Russian Federation
| | - Andrey V Slonov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Nataliya A Babaeva
- National Medical Scientific Centre of Obstetrics, Gynaecology and Perinatal Medicine named after V.I. Kulakov, Moscow, 117198, Russian Federation
- Russian Research Center of Roentgenology and Radiology, Moscow, 117997, Russian Federation
| | - Lev A Ashrafyan
- National Medical Scientific Centre of Obstetrics, Gynaecology and Perinatal Medicine named after V.I. Kulakov, Moscow, 117198, Russian Federation
- Russian Research Center of Roentgenology and Radiology, Moscow, 117997, Russian Federation
| | | | - Evgeniy G Evtushenko
- Faculty of Chemistry; Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Maria M Lukina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Zixiang Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University; Jinan, 250012, Shandong, China
| | - Artemiy S Silantiev
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Anna A Nushtaeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk, 630090, Russian Federation
| | - Daria D Kharlampieva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Vassili N Lazarev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Arseniy I Lashkin
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Lorine K Arzumanyan
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Irina Yu Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russian Federation
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russian Federation
| | - Olga S Lebedeva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Alexandra N Bogomazova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, 119435, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, 119435, Russian Federation
| | - Vadim M Govorun
- Research Institute for Systems Biology and Medicine, Moscow, 117246, Russian Federation
| |
Collapse
|
40
|
Baniasadi A, Das JP, Prendergast CM, Beizavi Z, Ma HY, Jaber MY, Capaccione KM. Imaging at the nexus: how state of the art imaging techniques can enhance our understanding of cancer and fibrosis. J Transl Med 2024; 22:567. [PMID: 38872212 PMCID: PMC11177383 DOI: 10.1186/s12967-024-05379-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Both cancer and fibrosis are diseases involving dysregulation of cell signaling pathways resulting in an altered cellular microenvironment which ultimately leads to progression of the condition. The two disease entities share common molecular pathophysiology and recent research has illuminated the how each promotes the other. Multiple imaging techniques have been developed to aid in the early and accurate diagnosis of each disease, and given the commonalities between the pathophysiology of the conditions, advances in imaging one disease have opened new avenues to study the other. Here, we detail the most up-to-date advances in imaging techniques for each disease and how they have crossed over to improve detection and monitoring of the other. We explore techniques in positron emission tomography (PET), magnetic resonance imaging (MRI), second generation harmonic Imaging (SGHI), ultrasound (US), radiomics, and artificial intelligence (AI). A new diagnostic imaging tool in PET/computed tomography (CT) is the use of radiolabeled fibroblast activation protein inhibitor (FAPI). SGHI uses high-frequency sound waves to penetrate deeper into the tissue, providing a more detailed view of the tumor microenvironment. Artificial intelligence with the aid of advanced deep learning (DL) algorithms has been highly effective in training computer systems to diagnose and classify neoplastic lesions in multiple organs. Ultimately, advancing imaging techniques in cancer and fibrosis can lead to significantly more timely and accurate diagnoses of both diseases resulting in better patient outcomes.
Collapse
Affiliation(s)
- Alireza Baniasadi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA.
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Conor M Prendergast
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Zahra Beizavi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Hong Y Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | | | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| |
Collapse
|
41
|
Adi W, Perez BER, Liu Y, Runkle S, Eliceiri KW, Yesilkoy F. Machine learning assisted mid-infrared spectrochemical fibrillar collagen imaging in clinical tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595393. [PMID: 38826188 PMCID: PMC11142197 DOI: 10.1101/2024.05.22.595393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Significance Label-free multimodal imaging methods that can provide complementary structural and chemical information from the same sample are critical for comprehensive tissue analyses. These methods are specifically needed to study the complex tumor-microenvironment where fibrillar collagen's architectural changes are associated with cancer progression. To address this need, we present a multimodal computational imaging method where mid-infrared spectral imaging (MIRSI) is employed with second harmonic generation (SHG) microscopy to identify fibrillar collagen in biological tissues. Aim To demonstrate a multimodal approach where a morphology-specific contrast mechanism guides a mid-infrared spectral imaging method to detect fibrillar collagen based on its chemical signatures. Approach We trained a supervised machine learning (ML) model using SHG images as ground truth collagen labels to classify fibrillar collagen in biological tissues based on their mid-infrared hyperspectral images. Five human pancreatic tissue samples (sizes are in the order of millimeters) were imaged by both MIRSI and SHG microscopes. In total, 2.8 million MIRSI spectra were used to train a random forest (RF) model. The remaining 68 million spectra were used to validate the collagen images generated by the RF-MIRSI model in terms of collagen segmentation, orientation, and alignment. Results Compared to the SHG ground truth, the generated MIRSI collagen images achieved a high average boundary F-score (0.8 at 4 pixels threshold) in the collagen distribution, high correlation (Pearson's R 0.82) in the collagen orientation, and similarly high correlation (Pearson's R 0.66) in the collagen alignment. Conclusions We showed the potential of ML-aided label-free mid-infrared hyperspectral imaging for collagen fiber and tumor microenvironment analysis in tumor pathology samples.
Collapse
Affiliation(s)
- Wihan Adi
- Department of Biomedical Engineering University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Bryan E. Rubio Perez
- Department of Electrical and Computer Engineering University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yuming Liu
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sydney Runkle
- Department of Computer Science University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering University of Wisconsin-Madison, Madison, WI, 53705, USA
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53706, USA
| | - Filiz Yesilkoy
- Department of Biomedical Engineering University of Wisconsin-Madison, Madison, WI, 53705, USA
| |
Collapse
|
42
|
Apa L, Martire MV, Carraro S, Cosentino M, Del Prete Z, Peruzzi B, Rizzuto E. Development of an Optical System for Strain Drop Measurement of Osteosarcoma Cells on Substrates with Different Stiffness. SENSORS (BASEL, SWITZERLAND) 2024; 24:3383. [PMID: 38894171 PMCID: PMC11175146 DOI: 10.3390/s24113383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024]
Abstract
Adherent cells perceive mechanical feedback from the underlying matrix and convert it into biochemical signals through a process known as mechanotransduction. The response to changes in the microenvironment relies on the cell's mechanical properties, including elasticity, which was recently identified as a biomarker for various diseases. Here, we propose the design, development, and characterization of a new system for the measurement of adherent cells' strain drop, a parameter correlated with cells' elasticity. To consider the interplay between adherent cells and the host extracellular matrix, cell stretching was combined with adhesion on substrates with different stiffnesses. The technique is based on the linear stretching of silicone chambers, high-speed image acquisition, and feedback for image centering. The system was characterized in terms of the strain homogeneity, impact of collagen coating, centering capability, and sensitivity. Subsequently, it was employed to measure the strain drop of two osteosarcoma cell lines, low-aggressive osteoblast-like SaOS-2 and high-aggressive 143B, cultured on two different substrates to recall the stiffness of the bone and lung extracellular matrices. Results demonstrated good substrate homogeneity, a negligible effect of the collagen coating, and an accurate image centering. Finally, the experimental results showed an average strain drop that was lower in the 143B cells in comparison with the SaOS-2 cells in all the tested conditions.
Collapse
Affiliation(s)
- Ludovica Apa
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy; (L.A.); (M.V.M.); (S.C.); (Z.D.P.)
| | - Maria Vittoria Martire
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy; (L.A.); (M.V.M.); (S.C.); (Z.D.P.)
| | - Serena Carraro
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy; (L.A.); (M.V.M.); (S.C.); (Z.D.P.)
| | - Marianna Cosentino
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy;
| | - Zaccaria Del Prete
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy; (L.A.); (M.V.M.); (S.C.); (Z.D.P.)
| | - Barbara Peruzzi
- Bone Physiopathology Research Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, 00184 Rome, Italy; (L.A.); (M.V.M.); (S.C.); (Z.D.P.)
| |
Collapse
|
43
|
Ferraresi A, Girone C, Maheshwari C, Vallino L, Dhanasekaran DN, Isidoro C. Ovarian Cancer Cell-Conditioning Medium Induces Cancer-Associated Fibroblast Phenoconversion through Glucose-Dependent Inhibition of Autophagy. Int J Mol Sci 2024; 25:5691. [PMID: 38891879 PMCID: PMC11171902 DOI: 10.3390/ijms25115691] [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: 03/19/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
One aspect of ovarian tumorigenesis which is still poorly understood is the tumor-stroma interaction, which plays a major role in chemoresistance and tumor progression. Cancer-associated fibroblasts (CAFs), the most abundant stromal cell type in the tumor microenvironment, influence tumor growth, metabolism, metastasis, and response to therapy, making them attractive targets for anti-cancer treatment. Unraveling the mechanisms involved in CAFs activation and maintenance is therefore crucial for the improvement of therapy efficacy. Here, we report that CAFs phenoconversion relies on the glucose-dependent inhibition of autophagy. We show that ovarian cancer cell-conditioning medium induces a metabolic reprogramming towards the CAF-phenotype that requires the autophagy-dependent glycolytic shift. In fact, 2-deoxy-D-glucose (2DG) strongly hampers such phenoconversion and, most importantly, induces the phenoreversion of CAFs into quiescent fibroblasts. Moreover, pharmacological inhibition (by proline) or autophagy gene knockdown (by siBECN1 or siATG7) promotes, while autophagy induction (by either 2DG or rapamycin) counteracts, the metabolic rewiring induced by the ovarian cancer cell secretome. Notably, the nutraceutical resveratrol (RV), known to inhibit glucose metabolism and to induce autophagy, promotes the phenoreversion of CAFs into normal fibroblasts even in the presence of ovarian cancer cell-conditioning medium. Overall, our data support the view of testing autophagy inducers for targeting the tumor-promoting stroma as an adjuvant strategy to improve therapy success rates, especially for tumors with a highly desmoplastic stroma, like ovarian cancer.
Collapse
Affiliation(s)
- Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Carlo Girone
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Chinmay Maheshwari
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| |
Collapse
|
44
|
Jalil SMA, Henry JC, Cameron AJM. Targets in the Tumour Matrisome to Promote Cancer Therapy Response. Cancers (Basel) 2024; 16:1847. [PMID: 38791926 PMCID: PMC11119821 DOI: 10.3390/cancers16101847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The extracellular matrix (ECM) is composed of complex fibrillar proteins, proteoglycans, and macromolecules, generated by stromal, immune, and cancer cells. The components and organisation of the matrix evolves as tumours progress to invasive disease and metastasis. In many solid tumours, dense fibrotic ECM has been hypothesised to impede therapy response by limiting drug and immune cell access. Interventions to target individual components of the ECM, collectively termed the matrisome, have, however, revealed complex tumour-suppressor, tumour-promoter, and immune-modulatory functions, which have complicated clinical translation. The degree to which distinct components of the matrisome can dictate tumour phenotypes and response to therapy is the subject of intense study. A primary aim is to identify therapeutic opportunities within the matrisome, which might support a better response to existing therapies. Many matrix signatures have been developed which can predict prognosis, immune cell content, and immunotherapy responses. In this review, we will examine key components of the matrisome which have been associated with advanced tumours and therapy resistance. We have primarily focussed here on targeting matrisome components, rather than specific cell types, although several examples are described where cells of origin can dramatically affect tumour roles for matrix components. As we unravel the complex biochemical, biophysical, and intracellular transduction mechanisms associated with the ECM, numerous therapeutic opportunities will be identified to modify tumour progression and therapy response.
Collapse
Affiliation(s)
| | | | - Angus J. M. Cameron
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK; (S.M.A.J.); (J.C.H.)
| |
Collapse
|
45
|
Sneider A, Liu Y, Starich B, Du W, Nair PR, Marar C, Faqih N, Ciotti GE, Kim JH, Krishnan S, Ibrahim S, Igboko M, Locke A, Lewis DM, Hong H, Karl MN, Vij R, Russo GC, Gómez-de-Mariscal E, Habibi M, Muñoz-Barrutia A, Gu L, Eisinger-Mathason TK, Wirtz D. Small Extracellular Vesicles Promote Stiffness-mediated Metastasis. CANCER RESEARCH COMMUNICATIONS 2024; 4:1240-1252. [PMID: 38630893 PMCID: PMC11080964 DOI: 10.1158/2767-9764.crc-23-0431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/13/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
Tissue stiffness is a critical prognostic factor in breast cancer and is associated with metastatic progression. Here we show an alternative and complementary hypothesis of tumor progression whereby physiologic matrix stiffness affects the quantity and protein cargo of small extracellular vesicles (EV) produced by cancer cells, which in turn aid cancer cell dissemination. Primary patient breast tissue released by cancer cells on matrices that model human breast tumors (25 kPa; stiff EVs) feature increased adhesion molecule presentation (ITGα2β1, ITGα6β4, ITGα6β1, CD44) compared with EVs from softer normal tissue (0.5 kPa; soft EVs), which facilitates their binding to extracellular matrix proteins including collagen IV, and a 3-fold increase in homing ability to distant organs in mice. In a zebrafish xenograft model, stiff EVs aid cancer cell dissemination. Moreover, normal, resident lung fibroblasts treated with stiff and soft EVs change their gene expression profiles to adopt a cancer-associated fibroblast phenotype. These findings show that EV quantity, cargo, and function depend heavily on the mechanical properties of the extracellular microenvironment. SIGNIFICANCE Here we show that the quantity, cargo, and function of breast cancer-derived EVs vary with mechanical properties of the extracellular microenvironment.
Collapse
Affiliation(s)
- Alexandra Sneider
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Ying Liu
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Bartholomew Starich
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Wenxuan Du
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Praful R. Nair
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Carolyn Marar
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Najwa Faqih
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Gabrielle E. Ciotti
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joo Ho Kim
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Sejal Krishnan
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Salma Ibrahim
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Muna Igboko
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Alexus Locke
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Daniel M. Lewis
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Hanna Hong
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Michelle N. Karl
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Raghav Vij
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Gabriella C. Russo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Estibaliz Gómez-de-Mariscal
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Mehran Habibi
- Johns Hopkins Breast Center, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Arrate Muñoz-Barrutia
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, Leganés, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Luo Gu
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - T.S. Karin Eisinger-Mathason
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences–Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
- Department of Materials Science and Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
46
|
Domenech J, Villacorta A, Ferrer JF, Llorens-Chiralt R, Marcos R, Hernández A, Catalán J. In vitro cell-transforming potential of secondary polyethylene terephthalate and polylactic acid nanoplastics. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:134030. [PMID: 38493621 DOI: 10.1016/j.jhazmat.2024.134030] [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: 12/08/2023] [Revised: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Continuous exposure to plastic pollutants may have serious consequences on human health. However, most toxicity assessments focus on non-environmentally relevant particles and rarely investigate long-term effects such as cancer induction. The present study assessed the carcinogenic potential of two secondary nanoplastics: polyethylene terephthalate (PET) particles generated from plastic bottles, and a biodegradable polylactic acid material, as respective examples of environmentally existing particles and new bioplastics. Pristine polystyrene nanoplastics were also included for comparison. A broad concentration range (6.25-200 μg/mL) of each nanoplastic was tested in both the initiation and promotion conditions of the regulatory assessment-accepted in vitro Bhas 42 cell transformation assay. Parallel cultures allowed confirmation of the efficient cellular internalisation of the three nanoplastics. Cell growth was enhanced by polystyrene in the initiation assay, and by PET in both conditions. Moreover, the number of transformed foci was significantly increased only by the highest PET concentration in the promotion assay, which also showed dose-dependency, indicating that nano PET can act as a non-genotoxic tumour promotor. Together, these findings support the carcinogenic risk assessment of nanoplastics and raise concerns regarding whether real-life co-exposure of PET nanoplastics and other environmental pollutants may result in synergistic transformation capacities.
Collapse
Affiliation(s)
- Josefa Domenech
- Finnish Institute of Occupational Health, Box 40, Työterveyslaitos, 00032 Helsinki, Finland
| | - Aliro Villacorta
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Facultad de Recursos Naturales Renovables, Universidad Arturo Prat, Iquique, Chile
| | | | | | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Alba Hernández
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Julia Catalán
- Finnish Institute of Occupational Health, Box 40, Työterveyslaitos, 00032 Helsinki, Finland; Department of Anatomy, Embryology and Genetics, University of Zaragoza, 50013 Zaragoza, Spain.
| |
Collapse
|
47
|
Xiao T, Lee J, Gauntner TD, Velegraki M, Lathia JD, Li Z. Hallmarks of sex bias in immuno-oncology: mechanisms and therapeutic implications. Nat Rev Cancer 2024; 24:338-355. [PMID: 38589557 DOI: 10.1038/s41568-024-00680-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/10/2024]
Abstract
Sex differences are present across multiple non-reproductive organ cancers, with male individuals generally experiencing higher incidence of cancer with poorer outcomes. Although some mechanisms underlying these differences are emerging, the immunological basis is not well understood. Observations from clinical trials also suggest a sex bias in conventional immunotherapies with male individuals experiencing a more favourable response and female individuals experiencing more severe adverse events to immune checkpoint blockade. In this Perspective article, we summarize the major biological hallmarks underlying sex bias in immuno-oncology. We focus on signalling from sex hormones and chromosome-encoded gene products, along with sex hormone-independent and chromosome-independent epigenetic mechanisms in tumour and immune cells such as myeloid cells and T cells. Finally, we highlight opportunities for future studies on sex differences that integrate sex hormones and chromosomes and other emerging cancer hallmarks such as ageing and the microbiome to provide a more comprehensive view of how sex differences underlie the response in cancer that can be leveraged for more effective immuno-oncology approaches.
Collapse
Affiliation(s)
- Tong Xiao
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy D Gauntner
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Maria Velegraki
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
- Rose Ella Burkhardt Brain Tumour Center, Cleveland Clinic, Cleveland, OH, USA.
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA.
| |
Collapse
|
48
|
Liu J, Zou Q. Supramolecular Peptide‐basedNanomaterials for the Treatment of Fibrosis. PEPTIDE SELF‐ASSEMBLY AND ENGINEERING 2024:479-503. [DOI: 10.1002/9783527841264.ch20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
49
|
Sneider A, Liu Y, Starich B, Du W, Marar C, Faqih N, Ciotti GE, Kim JH, Krishnan S, Ibrahim S, Igboko M, Locke A, Lewis DM, Hong H, Karl M, Vij R, Russo GC, Nair P, Gómez-de-Mariscal E, Habibi M, Muñoz-Barrutia A, Gu L, Eisinger-Mathason TSK, Wirtz D. Small extracellular vesicles promote stiffness-mediated metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.545937. [PMID: 37425743 PMCID: PMC10327142 DOI: 10.1101/2023.07.01.545937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Tissue stiffness is a critical prognostic factor in breast cancer and is associated with metastatic progression. Here we show an alternative and complementary hypothesis of tumor progression whereby physiological matrix stiffness affects the quantity and protein cargo of small EVs produced by cancer cells, which in turn drive their metastasis. Primary patient breast tissue produces significantly more EVs from stiff tumor tissue than soft tumor adjacent tissue. EVs released by cancer cells on matrices that model human breast tumors (25 kPa; stiff EVs) feature increased adhesion molecule presentation (ITGα 2 β 1 , ITGα 6 β 4 , ITGα 6 β 1 , CD44) compared to EVs from softer normal tissue (0.5 kPa; soft EVs), which facilitates their binding to extracellular matrix (ECM) protein collagen IV, and a 3-fold increase in homing ability to distant organs in mice. In a zebrafish xenograft model, stiff EVs aid cancer cell dissemination through enhanced chemotaxis. Moreover, normal, resident lung fibroblasts treated with stiff and soft EVs change their gene expression profiles to adopt a cancer associated fibroblast (CAF) phenotype. These findings show that EV quantity, cargo, and function depend heavily on the mechanical properties of the extracellular microenvironment.
Collapse
|
50
|
Colgrave EM, Keast JR, Nowell CJ, Healey M, Rogers PAW, Holdsworth-Carson SJ, Girling JE. Distribution of smooth muscle actin and collagen in superficial peritoneal endometriotic lesions varies from the surrounding microenvironment. Reprod Biomed Online 2024; 48:103610. [PMID: 38241767 DOI: 10.1016/j.rbmo.2023.103610] [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: 07/02/2023] [Revised: 09/26/2023] [Accepted: 10/09/2023] [Indexed: 01/21/2024]
Abstract
RESEARCH QUESTION Do different subtypes of superficial peritoneal endometriotic lesions exist, based on the presence and morphology of smooth muscle, collagen fibres and immune cell populations? DESIGN A retrospective cohort study of 24 patients, from across the menstrual cycle, with surgically and histologically confirmed endometriosis. Immunofluorescence was used to delineate the CD10 stromal area of lesions (n = 271 lesions from 67 endometriotic biopsies), and then smooth muscle actin (SMA) positive tissue and immune cell populations (CD45+ and CD68+) were quantified within and adjacent to these lesions. Second harmonic generation microscopy was used to evaluate the presence and morphology of type-1 collagen fibres within and surrounding lesions. RESULTS Overall, immune cell numbers and the area of SMA and collagen within endometriotic lesions tended to be low, but a spectrum of presentations significantly varied, particularly in the adjacent tissue microenvironment, based on lesion locations, the morphology of endometriotic gland profiles, or both. Lesions in which collagen fibres formed well aligned capsules around the CD10+ stromal border were identified compared with lesions in which collagen fibre distribution was random. Considerable inter- and intra-patient variability in the morphology of SMA and collagen was observed within and surrounding lesions. CONCLUSION These data demonstrate considerable diversity in the presence of immune cells and morphology of SMA and collagen within, but even more so, surrounding endometriotic lesions, even within individual patients. This heterogeneity, especially within individual patients, presents a challenge to incorporating these cell and tissue types into any new endometriosis classification systems or prognostic approaches.
Collapse
Affiliation(s)
- Eliza Morgan Colgrave
- Department of Obstetrics and Gynaecology, The University of Melbourne and Gynaecology Research Centre, Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Janet R Keast
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Cameron J Nowell
- Imaging, FACS and Analysis Core, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Martin Healey
- Department of Obstetrics and Gynaecology, The University of Melbourne and Gynaecology Research Centre, Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Peter A W Rogers
- Department of Obstetrics and Gynaecology, The University of Melbourne and Gynaecology Research Centre, Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Sarah J Holdsworth-Carson
- Department of Obstetrics and Gynaecology, The University of Melbourne and Gynaecology Research Centre, Royal Women's Hospital, Melbourne, Victoria, Australia; The Julia Argyrou Endometriosis Centre, Epworth HealthCare, Richmond, Victoria, Australia
| | - Jane E Girling
- Department of Obstetrics and Gynaecology, The University of Melbourne and Gynaecology Research Centre, Royal Women's Hospital, Melbourne, Victoria, Australia; Department of Anatomy, School of Biomedical Sciences, The University of Otago, Dunedin, Aotearoa New Zealand.
| |
Collapse
|