1
|
Pecci V, Pierconti F, Carlino A, Pinto F, Gradilone U, De Martino S, Rotili D, Grassi C, Pontecorvi A, Gaetano C, Strigari L, Farsetti A, Nanni S. Prognostic Impact of H19/Cell Adhesion Molecules Circuitry on Prostate Cancer Biopsy. Biomedicines 2024; 12:2322. [PMID: 39457633 PMCID: PMC11504315 DOI: 10.3390/biomedicines12102322] [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: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION Metastatic prostate cancer (PCa) presents a significant challenge in oncology due to its high mortality rate and the absence of effective biomarkers for predicting patient outcomes. Building on previous research that highlighted the critical role of the long noncoding RNA (lncRNA) H19 and cell adhesion molecules in promoting tumor progression under hypoxia and estrogen stimulation, this study aimed to assess the potential of these components as prognostic biomarkers for PCa at the biopsy stage. METHODS This research utilized immunohistochemistry and droplet digital PCR to analyze formalin-fixed paraffin-embedded (FFPE) biopsies, focusing on specific markers within the H19/cell adhesion molecules pathway. RESULTS A novel multivariate analysis led to a "BioScore", a composite biomarker score to predict disease progression. This score is based on evaluating five key markers: the expression levels of Hypoxia-Inducible Factor 2 Alpha (HIF-2α), endothelial Nitric Oxide Synthase (eNOS), β4 integrin, E-cadherin transcript (CDH1), and lncRNA H19. The criteria for the "BioScore" involve identifying three out of these five markers, combining elevated levels of HIF-2α, eNOS, β4 integrin, and CDH1 with reduced H19 expression. CONCLUSIONS This finding suggests the possibility of identifying, at the time of biopsy, PCa patients at higher risk of metastasis based on dysregulation in the H19/cell adhesion molecules circuitry. This study provides a valuable opportunity for early intervention in managing PCa, potentially contributing to personalized treatment strategies.
Collapse
Affiliation(s)
- Valeria Pecci
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (F.P.); (U.G.); (A.P.)
| | - Francesco Pierconti
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (F.P.); (A.C.); (C.G.)
- Department of Woman, Child and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Angela Carlino
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (F.P.); (A.C.); (C.G.)
| | - Francesco Pinto
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (F.P.); (U.G.); (A.P.)
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (F.P.); (A.C.); (C.G.)
| | - Ugo Gradilone
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (F.P.); (U.G.); (A.P.)
| | - Sara De Martino
- National Research Council (CNR)–Istituto di Analisi dei Sistemi ed Informatica “Antonio Ruberti” (IASI), 00185 Rome, Italy;
| | - Dante Rotili
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Claudio Grassi
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (F.P.); (A.C.); (C.G.)
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alfredo Pontecorvi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (F.P.); (U.G.); (A.P.)
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (F.P.); (A.C.); (C.G.)
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Lidia Strigari
- Department of Medical Physics, S. Orsola Malpighi University Hospital, 40138 Bologna, Italy;
| | - Antonella Farsetti
- National Research Council (CNR)–Istituto di Analisi dei Sistemi ed Informatica “Antonio Ruberti” (IASI), 00185 Rome, Italy;
| | - Simona Nanni
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.P.); (F.P.); (U.G.); (A.P.)
- Fondazione “Policlinico Universitario A. Gemelli IRCCS”, 00168 Rome, Italy; (F.P.); (A.C.); (C.G.)
| |
Collapse
|
2
|
Zeng G, Shen Y, Sun W, Lu H, Liang Y, Wu J, Liao G. Phenotype remodelling of HNSCC cells in the muscle invasion environment. J Transl Med 2024; 22:909. [PMID: 39375763 PMCID: PMC11457420 DOI: 10.1186/s12967-024-05607-8] [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: 03/12/2024] [Accepted: 08/18/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Tumour invading muscle in head and neck squamous cell carcinoma (HNSCC) is often associated with destructive growth and poor prognosis. However, the phenotypic functions and pathological mechanisms of muscle-invasive cancer cells in tumour progress remains unknown. In this study, we aimed to investigate the phenotypic functions of muscle-invasive cancer cells of HNSCC and their potential crosstalk with tumour microenvironment. METHODS We obtained scRNA-seq data (SC) from GSE103322 (N = 18) and GSE181919 (N = 37), spatial RNA-seq data (ST) from GSE208253 and GSE181300 (N = 4), transcriptomics of human HNSCC samples from GSE42743 (N = 12) and GSE41613 (N = 97). Utilizing the TCGA-HNSC dataset, we conducted univariate and multivariate Cox analyses to investigate the prognostic impact of muscle-invasion in HNSCC, with validation in an additional cohort. Through Stutility and AUCell approaches, we identified and characterized muscle-invasive cancer cell clusters, including their functional phenotypes and gene-specific profiles. Integration of SC and ST data was achieved using Seurat analysis, multimodal intersection analysis, and spatial deconvolution. The results were further validated via in vitro and in vivo experiments. RESULTS Our analyses of the TCGA-HNSC cohort revealed the presence of muscle-invasion was associated with a poor prognosis. By combining ST and SC, we identified muscle-invasive cancer cells exhibiting epithelial-to-mesenchymal transition (EMT) and myoepithelial-like transcriptional programs, which were correlated with a poor prognosis. Furthermore, we identified G0S2 as a novel marker of muscle-invasive malignant cells that potentially promotes EMT and the acquisition of myoepithelium-like phenotypes. These findings were validated through in vitro assays and chorioallantoic membranes experiments. Additionally, we demonstrated that G0S2-overexpressing cancer cells might attract human ECs via VEGF signalling. Subsequent in vitro and in vivo experiments revealed G0S2 plays key roles in promoting the proliferation and invasion of cancer cells. CONCLUSIONS In this study, we profiled the transcriptional programs of muscle-invasive HNSCC cell populations and characterized their EMT and myoepithelial-like phenotypes. Furthermore, our findings highlight the presence of muscle-invasion as a predictive marker for HNSCC patients. G0S2 as one of the markers of muscle-invasive cancer cells is involved in HNSCC intravasation, probably via VEGF signalling.
Collapse
Affiliation(s)
- Guozhong Zeng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yi Shen
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Huanzi Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yujie Liang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Jiashun Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Guiqing Liao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Szewczyk K, Jiang L, Khawaja H, Miranti CK, Zohar Y. Microfluidic Applications in Prostate Cancer Research. MICROMACHINES 2024; 15:1195. [PMID: 39459070 PMCID: PMC11509716 DOI: 10.3390/mi15101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Prostate cancer is a disease in which cells in the prostate, a gland in the male reproductive system below the bladder, grow out of control and, among men, it is the second-most frequently diagnosed cancer (other than skin cancer). In recent years, prostate cancer death rate has stabilized and, currently, it is the second-most frequent cause of cancer death in men (after lung cancer). Most deaths occur due to metastasis, as cancer cells from the original tumor establish secondary tumors in distant organs. For a long time, classical cell cultures and animal models have been utilized in basic and applied scientific research, including clinical applications for many diseases, such as prostate cancer, since no better alternatives were available. Although helpful in dissecting cellular mechanisms, these models are poor predictors of physiological behavior mainly because of the lack of appropriate microenvironments. Microfluidics has emerged in the last two decades as a technology that could lead to a paradigm shift in life sciences and, in particular, controlling cancer. Microfluidic systems, such as organ-on-chips, have been assembled to mimic the critical functions of human organs. These microphysiological systems enable the long-term maintenance of cellular co-cultures in vitro to reconstitute in vivo tissue-level microenvironments, bridging the gap between traditional cell cultures and animal models. Several reviews on microfluidics for prostate cancer studies have been published focusing on technology advancement and disease progression. As metastatic castration-resistant prostate cancer remains a clinically challenging late-stage cancer, with no curative treatments, we expanded this review to cover recent microfluidic applications related to prostate cancer research. The review includes discussions of the roles of microfluidics in modeling the human prostate, prostate cancer initiation and development, as well as prostate cancer detection and therapy, highlighting potentially major contributions of microfluidics in the continuous march toward eradicating prostate cancer.
Collapse
Affiliation(s)
- Kailie Szewczyk
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA;
| | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
4
|
Paxson AI, Chang LH, Gard JMC, Harryman WL, Nelson CS, Salmon SB, Marr KD, Wachsmuth LM, Ramanathan A, Ran J, Kapoor A, Marugan JJ, Henderson MJ, Sanchez TW, Cress AE. Phenotype plasticity and altered sensitivity to chemotherapeutic agents in aggressive prostate cancer cells. Front Cell Dev Biol 2023; 11:1285372. [PMID: 38046670 PMCID: PMC10690371 DOI: 10.3389/fcell.2023.1285372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/24/2023] [Indexed: 12/05/2023] Open
Abstract
In 2023, approximately 288,300 new diagnoses of prostate cancer will occur, with 34,700 disease-related deaths. Death from prostate cancer is associated with metastasis, enabled by progression of tumor phenotypes and successful extracapsular extension to reach Batson's venous plexus, a specific route to the spine and brain. Using a mouse-human tumor xenograft model, we isolated an aggressive muscle invasive cell population of prostate cancer, called DU145J7 with a distinct biophysical phenotype, elevated histone H3K27, and increased matrix metalloproteinase 14 expression as compared to the non-aggressive parent cell population called DU145WT. Our goal was to determine the sensitivities to known chemotherapeutic agents of the aggressive cells as compared to the parent population. High-throughput screening was performed with 5,578 compounds, comprising of approved and investigational drugs for oncology. Eleven compounds were selected for additional testing, which revealed that vorinostat, 5-azacitidine, and fimepinostat (epigenetic inhibitors) showed 2.6-to-7.5-fold increases in lethality for the aggressive prostate cancer cell population as compared to the parent, as judged by the concentration of drug to inhibit 50% cell growth (IC50). On the other hand, the DU145J7 cells were 2.2-to-4.0-fold resistant to mitoxantrone, daunorubicin, and gimatecan (topoisomerase inhibitors) as compared to DU145WT. No differences in sensitivities between cell populations were found for docetaxel or pirarubicin. The increased sensitivity of DU145J7 prostate cancer cells to chromatin modifying agents suggests a therapeutic vulnerability occurs after tumor cells invade into and through muscle. Future work will determine which epigenetic modifiers and what combinations will be most effective to eradicate early aggressive tumor populations.
Collapse
Affiliation(s)
- Allan I. Paxson
- Partnership for Native American Cancer Prevention, University of Arizona, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Loren H. Chang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Jaime M. C. Gard
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - William L. Harryman
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Colin S. Nelson
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Stella B. Salmon
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Kendra D. Marr
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
- Medical Scientist Training MD/PhD Program, College of Medicine Tucson, University of Arizona, Tucson, AZ, United States
| | - Leah M. Wachsmuth
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Anita Ramanathan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Jing Ran
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Abhijeet Kapoor
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Juan J. Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Mark J. Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Tino W. Sanchez
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Anne E. Cress
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
5
|
Marr KD, Gard JMC, Harryman WL, Keeswood EJ, Paxson AI, Wolgemuth C, Knudsen BS, Nagle RB, Hazlehurst L, Sorbellini M, Cress AE. Biophysical phenotype mixtures reveal advantages for tumor muscle invasion in vivo. Biophys J 2023; 122:4194-4206. [PMID: 37766428 PMCID: PMC10645557 DOI: 10.1016/j.bpj.2023.09.016] [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: 03/13/2023] [Revised: 08/23/2023] [Accepted: 09/25/2023] [Indexed: 09/29/2023] Open
Abstract
Bladder, colon, gastric, prostate, and uterine cancers originate in organs surrounded by laminin-coated smooth muscle. In human prostate cancer, tumors that are organ confined, without extracapsular extension through muscle, have an overall cancer survival rate of up to 97% compared with 32% for metastatic disease. Our previous work modeling extracapsular extension reported the blocking of tumor invasion by mutation of a laminin-binding integrin called α6β1. Expression of the α6AA mutant resulted in a biophysical switch from cell-ECM (extracellular matrix) to cell-cell adhesion with drug sensitivity properties and an inability to invade muscle. Here we used different admixtures of α6AA and α6WT cells to test the cell heterogeneity requirements for muscle invasion. Time-lapse video microscopy revealed that tumor mixtures self-assembled into invasive networks in vitro, whereas α6AA cells assembled only as cohesive clusters. Invasion of α6AA cells into and through live muscle occurred using a 1:1 mixture of α6AA and α6WT cells. Electric cell-substrate impedance sensing measurements revealed that compared with α6AA cells, invasion-competent α6WT cells were 2.5-fold faster at closing a cell-ECM or cell-cell wound, respectively. Cell-ECM rebuilding kinetics show that an increased response occurred in mixtures since the response was eightfold greater compared with populations containing only one cell type. A synthetic cell adhesion cyclic peptide called MTI-101 completely blocked electric cell-substrate impedance sensing cell-ECM wound recovery that persisted in vitro up to 20 h after the wound. Treatment of tumor-bearing animals with 10 mg/kg MTI-101 weekly resulted in a fourfold decrease of muscle invasion by tumor and a decrease of the depth of invasion into muscle comparable to the α6AA cells. Taken together, these data suggest that mixed biophysical phenotypes of tumor cells within a population can provide functional advantages for tumor invasion into and through muscle that can be potentially inhibited by a synthetic cell adhesion molecule.
Collapse
Affiliation(s)
- Kendra D Marr
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona; Medical Scientist Training Program, College of Medicine, University of Arizona, Tucson, Arizona
| | | | | | - Elijah J Keeswood
- University of Arizona Cancer Center, Tucson, Arizona; Partnership for Native American Cancer Prevention, University of Arizona, Tucson, Arizona
| | - Allan I Paxson
- Partnership for Native American Cancer Prevention, University of Arizona, Tucson, Arizona
| | | | - Beatrice S Knudsen
- Department of Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Raymond B Nagle
- Department of Pathology, University of Arizona Cancer Center, Tucson, Arizona
| | - Lori Hazlehurst
- Associate Director of Basic Research, Co-Leader Alexander B. Osborn Hematopoietic Malignancy and Transplantation, West Virginia University, Morgantown, West Virginia
| | | | - Anne E Cress
- University of Arizona Cancer Center, Tucson, Arizona; Department of Cellular and Molecular Medicine and Department of Radiation Oncology, College of Medicine, University of Arizona, Tucson, Arizona.
| |
Collapse
|
6
|
Dautle M, Zhang S, Chen Y. scTIGER: A Deep-Learning Method for Inferring Gene Regulatory Networks from Case versus Control scRNA-seq Datasets. Int J Mol Sci 2023; 24:13339. [PMID: 37686146 PMCID: PMC10488287 DOI: 10.3390/ijms241713339] [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: 08/06/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Inferring gene regulatory networks (GRNs) from single-cell RNA-seq (scRNA-seq) data is an important computational question to find regulatory mechanisms involved in fundamental cellular processes. Although many computational methods have been designed to predict GRNs from scRNA-seq data, they usually have high false positive rates and none infer GRNs by directly using the paired datasets of case-versus-control experiments. Here we present a novel deep-learning-based method, named scTIGER, for GRN detection by using the co-differential relationships of gene expression profiles in paired scRNA-seq datasets. scTIGER employs cell-type-based pseudotiming, an attention-based convolutional neural network method and permutation-based significance testing for inferring GRNs among gene modules. As state-of-the-art applications, we first applied scTIGER to scRNA-seq datasets of prostate cancer cells, and successfully identified the dynamic regulatory networks of AR, ERG, PTEN and ATF3 for same-cell type between prostatic cancerous and normal conditions, and two-cell types within the prostatic cancerous environment. We then applied scTIGER to scRNA-seq data from neurons with and without fear memory and detected specific regulatory networks for BDNF, CREB1 and MAPK4. Additionally, scTIGER demonstrates robustness against high levels of dropout noise in scRNA-seq data.
Collapse
Affiliation(s)
- Madison Dautle
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Shaoqiang Zhang
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Yong Chen
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA;
| |
Collapse
|
7
|
Kohno S, Ikegami M, Yamamoto SR, Aoki H, Ogawa M, Yano F, Eto K. A rare case of colorectal metastasis found 8 years and 10 months after gastrectomy for advanced gastric cancer: A case report and literature review. Oncol Lett 2023; 25:203. [PMID: 37123025 PMCID: PMC10131265 DOI: 10.3892/ol.2023.13789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/24/2023] [Indexed: 05/02/2023] Open
Abstract
Colorectal metastasis from gastric cancer is rare and may develop several years after gastric cancer surgery. Therefore, colonoscopic findings provide useful diagnostic information. The present report describes a case of gastric cancer colon metastasis diagnosed 8 years and 10 months after gastrectomy for advanced gastric cancer. A 64-year-old male patient underwent gastrectomy in December 2010 and received chemotherapy for 4 years and 10 months after the surgery. Subsequently, the patient was diagnosed as having colorectal cancer by computed tomography in February 2019. Colonoscopy revealed linitis plastica-like colon stenosis; however, biopsy pathology did not reveal any findings indicating malignancy. Right hemicolectomy was performed, and pathological examination revealed colon metastasis from gastric cancer. The patient received chemotherapy but died of peritoneal carcinomatosis 1 year and 8 months after the colectomy. According to literature, colorectal metastasis from gastric cancer is often attributed to hematogenous metastasis and often exhibits characteristic macroscopic features. Treatments, such as chemotherapy for gastric cancer and/or colorectal resection, are considered effective for gastric cancer colorectal metastasis.
Collapse
Affiliation(s)
- Shuzo Kohno
- Department of Surgery, The Jikei University Katsushika Medical Center, Tokyo 125-8061, Japan
- Correspondence to: Dr Shuzo Kohno, Department of Surgery, The Jikei University Katsushika Medical Center, 6-41-2 Aoto, Katsushika-ku, Tokyo 125-8061, Japan, E-mail:
| | - Masahiro Ikegami
- Department of Pathology, The Jikei University Katsushika Medical Center, Tokyo 125-8061, Japan
| | - Se Ryung Yamamoto
- Department of Surgery, The Jikei University Katsushika Medical Center, Tokyo 125-8061, Japan
| | - Hiroaki Aoki
- Department of Surgery, The Jikei University Katsushika Medical Center, Tokyo 125-8061, Japan
| | - Masaichi Ogawa
- Department of Surgery, The Jikei University Katsushika Medical Center, Tokyo 125-8061, Japan
| | - Fumiaki Yano
- Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Ken Eto
- Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| |
Collapse
|
8
|
Tu H, Liu H, Zhang L, Tan Z, Wang H, Jiang Y, Xia Z, Guo L, Xia X, Gu P, Liu X. A novel prognostic model based on three integrin subunit genes-related signature for bladder cancer. Front Oncol 2022; 12:970576. [PMID: 36267977 PMCID: PMC9577111 DOI: 10.3389/fonc.2022.970576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Presently, a comprehensive analysis of integrin subunit genes (ITGs) in bladder cancer (BLCA) is absent. This study endeavored to thoroughly analyze the utility of ITGs in BLCA through computer algorithm-based bioinformatics. Methods BLCA-related materials were sourced from reputable databases, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). R software-based bioinformatics analyses included limma-differential expression analysis, survival-Cox analysis, glmnet-Least absolute shrinkage and selection operator (LASSO), clusterProfiler-functional annotation, and gsva-estimate-immune landscape analysis. The expression difference of key genes was verified by quantitative real-time polymerase chain reaction (qRT-PCR). Results Among the 11 ITGs that were abnormally expressed in BLCA, ITGA7, ITGA5, and ITGB6 were categorized as the optimal variables for structuring the risk model. The high-risk subcategories were typified by brief survival, abysmal prognosis, prominent immune and stromal markers, and depressed tumor purity. The risk model was also an isolated indicator of the impact of clinical outcomes in BLCA patients. Moreover, the risk model, specifically the high-risk subcategory with inferior prognosis, became heavily interlinked with the immune-inflammatory response and smooth muscle contraction and relaxation. Conclusion This study determined three ITGs with prognostic values (ITGA7, ITGA5, and ITGB6), composed a novel (ITG-associated) prognostic gene signature, and preliminarily probed the latent molecular mechanisms of the model.
Collapse
Affiliation(s)
- Hongtao Tu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, China
| | - Haolin Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Longfei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhiyong Tan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hai Wang
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, China
| | - Yongming Jiang
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhongyou Xia
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, China
| | - Liwei Guo
- Department of Urology, The Dazhu County People’s Hospital, Dazhou, China
| | - Xiaodong Xia
- Department of Urology, The Dazhu County People’s Hospital, Dazhou, China
| | - Peng Gu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, China
- *Correspondence: Xiaodong Liu, ; Peng Gu,
| | - Xiaodong Liu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, China
- *Correspondence: Xiaodong Liu, ; Peng Gu,
| |
Collapse
|
9
|
Harryman WL, Marr KD, Nagle RB, Cress AE. Integrins and Epithelial-Mesenchymal Cooperation in the Tumor Microenvironment of Muscle-Invasive Lethal Cancers. Front Cell Dev Biol 2022; 10:837585. [PMID: 35300411 PMCID: PMC8921537 DOI: 10.3389/fcell.2022.837585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/04/2022] [Indexed: 11/18/2022] Open
Abstract
Muscle-invasive lethal carcinomas traverse into and through this specialized biophysical and growth factor enriched microenvironment. We will highlight cancers that originate in organs surrounded by smooth muscle, which presents a barrier to dissemination, including prostate, bladder, esophageal, gastric, and colorectal cancers. We propose that the heterogeneity of cell-cell and cell-ECM adhesion receptors is an important driver of aggressive tumor networks with functional consequences for progression. Phenotype heterogeneity of the tumor provides a biophysical advantage for tumor network invasion through the tensile muscle and survival of the tumor network. We hypothesize that a functional epithelial-mesenchymal cooperation (EMC)exists within the tumor invasive network to facilitate tumor escape from the primary organ, invasion and traversing of muscle, and navigation to metastatic sites. Cooperation between specific epithelial cells within the tumor and stromal (mesenchymal) cells interacting with the tumor is illustrated using the examples of laminin-binding adhesion molecules—especially integrins—and their response to growth and inflammatory factors in the tumor microenvironment. The cooperation between cell-cell (E-cadherin, CDH1) and cell-ECM (α6 integrin, CD49f) expression and growth factor receptors is highlighted within poorly differentiated human tumors associated with aggressive disease. Cancer-associated fibroblasts are examined for their role in the tumor microenvironment in generating and organizing various growth factors. Cellular structural proteins are potential utility markers for future spatial profiling studies. We also examine the special characteristics of the smooth muscle microenvironment and how invasion by a primary tumor can alter this environment and contribute to tumor escape via cooperation between epithelial and stromal cells. This cooperative state allows the heterogenous tumor clusters to be shaped by various growth factors, co-opt or evade immune system response, adapt from hypoxic to normoxic conditions, adjust to varying energy sources, and survive radiation and chemotherapeutic interventions. Understanding the epithelial-mesenchymal cooperation in early tumor invasive networks holds potential for both identifying early biomarkers of the aggressive transition and identification of novel agents to prevent the epithelial-mesenchymal cooperation phenotype. Epithelial-mesenchymal cooperation is likely to unveil new tumor subtypes to aid in selection of appropriate therapeutic strategies.
Collapse
Affiliation(s)
- William L Harryman
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States
| | - Kendra D Marr
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States.,Medical Scientist Training Program, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Ray B Nagle
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Pathology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Anne E Cress
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Cellular and Molecular Medicine and Department of Radiation Oncology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
10
|
Paulsen JD, Polydorides AD. Pathology and Prognosis of Colonic Adenocarcinomas With Intermediate Primary Tumor Stage Between pT2 and pT3. Arch Pathol Lab Med 2021; 146:591-602. [PMID: 34473229 DOI: 10.5858/arpa.2021-0109-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Primary tumor stage (pT) is an important prognostic indicator in colonic adenocarcinomas; however, cases that have no muscle fibers beyond the advancing tumor edge but also show no extension beyond the apparent outer border of muscularis propria (termed pT2int), have not been previously studied. OBJECTIVE.— To address the clinicopathologic characteristics and prognosis of pT2int tumors. DESIGN.— We recharacterized 168 colon carcinomas and compared pT2int cases to bona fide pT2 and pT3 tumors. RESULTS.— In side-by-side analysis, 21 pT2int cases diverged from 29 pT2 tumors only in terms of larger size (P = .03), but they were less likely to show high-grade (P = .03), lymphovascular (P < .001), and extramural venous invasion (P = .04); discontinuous tumor deposits (P = .02); lymph node involvement (P = .001); and advanced stage (P = .001), compared with 118 pT3 tumors. Combining pT2int with pT2 cases (versus pT3) was a better independent predictor of negative lymph nodes in multivariate analysis (P = .04; odds ratio [OR], 3.96; CI, 1.09-14.42) and absent distant metastasis in univariate analysis (P = .04), compared with sorting pT2int with pT3 cases (versus pT2). Proportional hazards regression showed that pT2 and pT2int cases together were associated with better disease-free survival compared with pT3 tumors (P = .04; OR, 3.65; CI, 1.05-12.70). Kaplan-Meier analysis demonstrated that when pT2int were grouped with pT2 tumors, they were significantly less likely to show disease progression compared with pT3 (P = .002; log-rank test) and showed a trend toward better disease-specific survival (P = .06), during a mean patient follow-up of 44.9 months. CONCLUSIONS.— These data support the conclusion that pT2int carcinomas have clinicopathologic characteristics and are associated with patient outcomes more closely aligned with pT2 rather than pT3 tumors.
Collapse
Affiliation(s)
- John D Paulsen
- From the Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexandros D Polydorides
- From the Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|