1
|
Gennarini M, Canese R, Capuani S, Miceli V, Tomao F, Palaia I, Zecca V, Maiuro A, Balba I, Catalano C, Rizzo SMR, Manganaro L. Multi-model quantitative MRI of uterine cancers in precision medicine's era-a narrative review. Insights Imaging 2025; 16:113. [PMID: 40437300 PMCID: PMC12119420 DOI: 10.1186/s13244-025-01965-z] [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: 09/23/2024] [Accepted: 03/30/2025] [Indexed: 06/01/2025] Open
Abstract
PURPOSE This review aims to summarize the current applications of quantitative MRI biomarkers in the staging, treatment response evaluation, and prognostication of endometrial (EC) and cervical cancer (CC). By focusing on functional imaging techniques, we explore how these biomarkers enhance personalized cancer management beyond traditional morphological assessments. METHODS A structured search of the PubMed database from January to May 2024 was conducted to identify relevant studies on quantitative MRI in uterine cancers. We included studies examining MRI biomarkers like Dynamic Contrast-Enhanced MRI (DCE-MRI), Diffusion-Weighted Imaging (DWI), and Magnetic Resonance Spectroscopy (MRS), emphasizing their roles in assessing tumor physiology, microstructure, and metabolic changes. RESULTS DCE-MRI provides valuable quantitative biomarkers such as Ktrans and Ve, which reflect microvascular characteristics and tumor aggressiveness, outperforming T2-weighted imaging in detecting critical factors like myometrial and cervical invasion. DWI, including advanced models like Intravoxel Incoherent Motion (IVIM), distinguishes between normal and cancerous tissue and correlates with tumor grade and treatment response. MRS identifies metabolic alterations, such as elevated choline and lipid signals, which serve as prognostic markers in uterine cancers. CONCLUSION Quantitative MRI offers a noninvasive method to assess key biomarkers that inform prognosis and guide treatment decisions in uterine cancers. By providing insights into tumor biology, these imaging techniques represent a significant step forward in the precision medicine era, allowing for a more tailored therapeutic approach based on the unique pathological and molecular characteristics of each tumor. CRITICAL RELEVANCE STATEMENT Biomarkers obtained from MRI can provide useful quantitative information about the nature of uterine cancers and their prognosis, both at diagnosis and response assessment, allowing better therapeutic strategies to be prepared. KEY POINTS Quantitative MRI improves diagnosis and management of uterine cancers through advanced imaging biomarkers. Quantitative MRI biomarkers enhance staging, prognosis, and treatment response assessment in uterine cancers. Quantitative MRI biomarkers support personalized treatment strategies and improve patient management in uterine cancers.
Collapse
Affiliation(s)
- Marco Gennarini
- Department of Radiological, Oncological and Pathological Sciences, Umberto I Hospital, "Sapienza" University of Rome, Rome, Italy
| | - Rossella Canese
- Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy.
| | - Silvia Capuani
- National Research Council (CNR), Institute for Complex Systems (ISC) c/o Physics Department Sapienza University of Rome, Rome, Italy
| | - Valentina Miceli
- Department of Radiological, Oncological and Pathological Sciences, Umberto I Hospital, "Sapienza" University of Rome, Rome, Italy
| | - Federica Tomao
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Innocenza Palaia
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Valentina Zecca
- Core Facilities, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
- Department of Basic and Applied Sciences for Engineering, University of Rome Sapienza, Rome, Italy
| | - Alessandra Maiuro
- National Research Council (CNR), Institute for Complex Systems (ISC) c/o Physics Department Sapienza University of Rome, Rome, Italy
| | | | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences, Umberto I Hospital, "Sapienza" University of Rome, Rome, Italy
| | - Stefania Maria Rita Rizzo
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Lugano, CH, Switzerland
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Lugano, CH, Switzerland
| | - Lucia Manganaro
- Department of Radiological, Oncological and Pathological Sciences, Umberto I Hospital, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
2
|
Sonoda H, Iwasaki T, Ishihara S, Mori T, Nakashima Y, Oda Y. Impact of Tertiary Lymphoid Structure on Prognosis and Tumor Microenvironment in Undifferentiated Pleomorphic Sarcoma. Cancer Sci 2025; 116:1464-1473. [PMID: 40007136 PMCID: PMC12044655 DOI: 10.1111/cas.70018] [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: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Undifferentiated pleomorphic sarcoma (UPS) has a favorable objective response rate to anti-PD-1 drugs compared with other sarcomas. Tertiary lymphoid structure (TLS) is a favorable prognostic factor and a biomarker for immune checkpoint inhibitors (ICIs). Nevertheless, there are limited data on the tumor microenvironment (TME) to support a good response to ICIs in sarcoma. Therefore, this study was conducted to investigate the impact of TLS on prognosis and TME. A total of 52 of UPS with wide resection were divided into intratumoral TLS, extratumoral TLS, and without TLS groups. Survival analysis and immunohistochemistry were performed to evaluate immune cells and immune checkpoint molecules, and multiplexed immunofluorescence was conducted to evaluate T-cell exhaustion among the three groups. TLS was detected in 34 cases (65%), including 23 intratumoral TLS (44%) and 11 extratumoral TLS (21%) cases. Patients with TLS had significantly longer overall survival than those without TLS (log rank p = 0.020). The intratumoral TLS group had a significantly higher number of immune cells and higher expression of PD-L1 and IDO-1 than the without TLS group. Progenitor-exhausted T cells were also observed in patients with UPS. In conclusion, these findings could help predict prognosis in patients with UPS. TLS was demonstrated to be a favorable prognostic factor in patients with UPS. Intratumoral TLS may be a biomarker for the response to ICIs, especially anti-PD-1 drugs.
Collapse
Affiliation(s)
- Hiroki Sonoda
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Orthopaedic Surgery, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takeshi Iwasaki
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Shin Ishihara
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Orthopaedic Surgery, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Taro Mori
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
3
|
Kawaguchi M, Kato H, Furui T, Isobe M, Noda Y, Hyodo F, Miyazaki T, Matsuo M. Prediction of grading of ovarian endometrioid carcinoma using conventional MRI features. Jpn J Radiol 2025; 43:820-828. [PMID: 39730935 PMCID: PMC12052869 DOI: 10.1007/s11604-024-01727-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVE The purpose of this study was to evaluate MRI findings of ovarian endometrioid carcinoma (OEC) as a predictor of histological grade. MATERIALS AND METHODS This study included 60 patients with histopathologically confirmed OEC (20, 30, and 10 with grades 1, 2, and 3, respectively). Clinical and MRI results were retrospectively reviewed. We compared the following parameters between the three grades: age, tumor markers, presence of uterine corpus cancer, bilaterality, configuration, peritoneal dissemination, abnormal ascites, signal intensities of cystic and solid components, tumor size, and apparent diffusion coefficient (ADC) values of solid components. RESULTS T1-hyperintense cysts were more common in grade 1 than in grades 2-3 OEC (80% vs. 60%, vs. 40%, p < 0.05). The signal intensity ratio between the cystic components with the largest solid component and muscle (1.49 vs. 1.08 vs. 0.98, p < 0.05) was higher in grade 1 than in grades 2-3 OEC. Necrosis within solid components was less common in grade 1 than in grades 2-3 OEC (31% vs. 68% vs. 88%, p < 0.05), and the ADC values of solid components were higher in grade 1 than in grades 2-3 OEC (1.10 vs. 0.99 vs. 0.79 × 10-3 mm2/sec, p < 0.05). There were no significant differences in other factors. CONCLUSION On T1-weighted images, grade 1 OEC showed a higher signal intensity in the cystic components than grades 2-3 OEC. Necrosis and lower ADC values were more frequently observed in grades 2-3 than in grade 1 OEC.
Collapse
Affiliation(s)
- Masaya Kawaguchi
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan.
- Department of Radiology, Ogaki Municipal Hospital, 4-86 Minaminokawacho, Ogaki, 503-0864, Japan.
| | - Hiroki Kato
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Tatsuro Furui
- Department of Obstetrics and Gynecology, Gifu University, Gifu, Japan
| | - Masanori Isobe
- Department of Obstetrics and Gynecology, Gifu University, Gifu, Japan
- Enter for One Medicine Innovative Translational Research (COMIT), Institute for Advanced Study, Gifu University, Gifu, Japan
| | - Yoshifumi Noda
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Fuminori Hyodo
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | | | - Masayuki Matsuo
- Department of Radiology, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
4
|
Dora D, Revisnyei P, Pasic A, Galffy G, Dulka E, Mihucz A, Roskó B, Szincsak S, Iliuk A, Weiss GJ, Lohinai Z. Host and bacterial urine proteomics might predict treatment outcomes for immunotherapy in advanced non-small cell lung cancer patients. Front Immunol 2025; 16:1543817. [PMID: 40297587 PMCID: PMC12035445 DOI: 10.3389/fimmu.2025.1543817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Urine samples are non-invasive approaches to study potential circulating biomarkers from the host organism. Specific proteins cross the bloodstream through the intestinal barrier and may also derive from gut microbiota. In this study, we aimed to evaluate the predictive role of the host and bacterial urine extracellular vesicle (EV) proteomes in patients with non-small cell lung cancer (NSCLC) treated with anti-PD1 immunotherapy. Methods We analyzed the urine EV proteome of 33 advanced-stage NSCLC patients treated with anti-PD1 immunotherapy with LC-MS/MS, stratifying patients according to long (>6 months) and short (≤6 months) progression-free survival (PFS). Gut microbial communities on a subcohort of 23 patients were also analyzed with shotgun metagenomics. Internal validation was performed using the Random Forest (RF) machine learning (ML) algorithm. RF was validated with a non-linear Bayesian ML model. Gene enrichment, and pathway analysis of host urine proteins were analyzed using the Reactome and Gene Ontology databases. Results We identified human (n=3513), bacterial (n=2647), fungal (n=19), and viral (n=4) proteins. 186 human proteins showed differential abundance (p<0.05) according to PFS groups, 101 being significantly more abundant in patients with short PFS and n=85 in patients with long PFS. We found several pathways that were significantly enriched in patients with short PFS (vs long PFS). Multivariate Cox regression showed that human urine proteins MPP5, IGKV6-21, NT5E, and KRT27 were strongly associated with long PFS, and LMAN2, NUTF2, NID1, TNC, IGF1, BCR, GPHN, and PPBP showed the strongest association with short PFS. We revealed that an increased bacterial/host protein ratio in the urine is more frequent in patients with long PFS. Increased abundance of E. coli and E. faecalis proteins in the urine positively correlates with their gut metagenomic abundance. RF ML model supported the reliability in predicting PFS for critical human urine proteins (AUC=0.89), accuracy (95%) and Bacterial proteins (AUC=0.74). Conclusion To our knowledge, this is the first study to depict the predictive role of the host and bacterial urine proteome in anti-PD1-treated advanced NSCLC.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Peter Revisnyei
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
- HUN-REN-BME Information Systems Research Group, Budapest, Hungary
| | - Alija Pasic
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Edit Dulka
- County Hospital of Torokbalint, Torokbalint, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Sara Szincsak
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN, United States
| | - Glen J. Weiss
- Department of Medicine, UMass Chan Medical School, Worcester, MA, United States
| | - Zoltan Lohinai
- County Hospital of Torokbalint, Torokbalint, Hungary
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
Wang P, Zhang XP, Liu F, Wang W. Progressive Deactivation of Hydroxylases Controls Hypoxia-Inducible Factor-1α-Coordinated Cellular Adaptation to Graded Hypoxia. RESEARCH (WASHINGTON, D.C.) 2025; 8:0651. [PMID: 40171017 PMCID: PMC11960303 DOI: 10.34133/research.0651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 04/03/2025]
Abstract
Graded hypoxia is a common microenvironment in malignant solid tumors. As a central regulator in the hypoxic response, hypoxia-inducible factor-1 (HIF-1) can induce multiple cellular processes including glycolysis, angiogenesis, and necroptosis. How cells exploit the HIF-1 pathway to coordinate different processes to survive hypoxia remains unclear. We developed an integrated model of the HIF-1α network to elucidate the mechanism of cellular adaptation to hypoxia. By numerical simulations and bifurcation analysis, we found that HIF-1α is progressively activated with worsening hypoxia due to the sequential deactivation of the hydroxylases prolyl hydroxylase domain enzymes and factor inhibiting HIF (FIH). Bistable switches control the activation and deactivation processes. As a result, glycolysis, immunosuppression, angiogenesis, and necroptosis are orderly elicited in aggravating hypoxia. To avoid the excessive accumulation of lactic acid during glycolysis, HIF-1α induces monocarboxylate transporter and carbonic anhydrase 9 sequentially to export intracellular hydrogen ions, facilitating tumor cell survival. HIF-1α-induced miR-182 facilitates vascular endothelial growth factor production to promote angiogenesis under moderate hypoxia. The imbalance between accumulation and removal of lactic acid in severe hypoxia may result in acidosis and induce cell necroptosis. In addition, the deactivation of FIH results in the destabilization of HIF-1α in anoxia. Collectively, HIF-1α orchestrates the adaptation of tumor cells to hypoxia by selectively inducing its targets according to the severity of hypoxia. Our work may provide clues for tumor therapy by targeting the HIF-1 pathway.
Collapse
Affiliation(s)
- Ping Wang
- Kuang Yaming Honors School,
Nanjing University, Nanjing 210023, China
- Key Laboratory of High Performance Scientific Computation, School of Science,
Xihua University, Chengdu 610039, China
| | - Xiao-Peng Zhang
- Kuang Yaming Honors School,
Nanjing University, Nanjing 210023, China
- Institute of Brain Sciences,
Nanjing University, Nanjing 210093, China
| | - Feng Liu
- Institute of Brain Sciences,
Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructures and Department of Physics,
Nanjing University, Nanjing 210093, China
| | - Wei Wang
- Institute of Brain Sciences,
Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructures and Department of Physics,
Nanjing University, Nanjing 210093, China
| |
Collapse
|
6
|
Hong Q, Li C, Li Z, Guo Z, Ashraf N, Li K. Noninvasive prediction model for predicting spontaneous tumor necrosis in hepatocellular carcinoma and prognostic study. Eur J Gastroenterol Hepatol 2025:00042737-990000000-00507. [PMID: 40207508 DOI: 10.1097/meg.0000000000002967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
BACKGROUND AND OBJECTIVES In hepatocellular carcinoma (HCC), patients with spontaneous tumor necrosis have a high recurrence rate and poor prognosis. However, conventional preoperative imaging could not detect the presence of tumor necrosis. Accordingly, we developed and assessed a nomogram to forecast tumor necrosis. METHODS Clinical data were collected retrospectively from 495 patients with HCC who received a hepatectomy at Zhongnan Hospital of Wuhan University from 1 January 2015 to 31 May 2024. The patients (n = 495) were randomly divided in a 7 : 3 ratio into the training cohort (TC, n = 348) and the validation cohort (VC, n = 147). The logistic regression analyses were used to identify factors independently predicting tumor necrosis in the patients with TC. The Kaplan-Meier survival analysis was used for comparing and estimating survival rates. RESULTS Preoperative clinical tumor-node-metastasis stage, hemoglobin, systemic immune inflammation, alkaline phosphatase, and alpha-fetoprotein levels were identified as hazard factors for predicting tumor necrosis. The area under the receiver operating characteristic curve of the TC, VC, and the full cohort was 0.810, 0.758, and 0.795, respectively. The calibration curves demonstrated a high degree of concordance. The decision curve analysis showed the clinical significance of the nomogram. Both overall survival and recurrence-free survival of patients in the tumor necrosis group were poorer. CONCLUSION Our predictive model could effectively predict the risk of spontaneous tumor necrosis in patients with HCC, and tumor necrosis was related to a worse prognosis.
Collapse
Affiliation(s)
- Qingyong Hong
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University
| | - Chunmin Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Clinical College of Wuhan University, Zhongnan Hospital of Wuhan University
| | - Ziqiang Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University
| | - Zhidong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University
| | - Nadeem Ashraf
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University
| | - Kun Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Medicine Research Center for Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Molefi T, Mabonga L, Hull R, Mwazha A, Sebitloane M, Dlamini Z. The Histomorphology to Molecular Transition: Exploring the Genomic Landscape of Poorly Differentiated Epithelial Endometrial Cancers. Cells 2025; 14:382. [PMID: 40072110 PMCID: PMC11898822 DOI: 10.3390/cells14050382] [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: 12/06/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
The peremptory need to circumvent challenges associated with poorly differentiated epithelial endometrial cancers (PDEECs), also known as Type II endometrial cancers (ECs), has prompted therapeutic interrogation of the prototypically intractable and most prevalent gynecological malignancy. PDEECs account for most endometrial cancer-related mortalities due to their aggressive nature, late-stage detection, and poor response to standard therapies. PDEECs are characterized by heterogeneous histopathological features and distinct molecular profiles, and they pose significant clinical challenges due to their propensity for rapid progression. Regardless of the complexities around PDEECs, they are still being administered inefficiently in the same manner as clinically indolent and readily curable type-I ECs. Currently, there are no targeted therapies for the treatment of PDEECs. The realization of the need for new treatment options has transformed our understanding of PDEECs by enabling more precise classification based on genomic profiling. The transition from a histopathological to a molecular classification has provided critical insights into the underlying genetic and epigenetic alterations in these malignancies. This review explores the genomic landscape of PDEECs, with a focus on identifying key molecular subtypes and associated genetic mutations that are prevalent in aggressive variants. Here, we discuss how molecular classification correlates with clinical outcomes and can refine diagnostic accuracy, predict patient prognosis, and inform therapeutic strategies. Deciphering the molecular underpinnings of PDEECs has led to advances in precision oncology and protracted therapeutic remissions for patients with these untamable malignancies.
Collapse
Affiliation(s)
- Thulo Molefi
- Discipline of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4002, South Africa;
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
- Department of Medical Oncology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Lloyd Mabonga
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| | - Absalom Mwazha
- Department of Anatomical Pathology, National Health Laboratory Services, Durban 4058, South Africa
| | - Motshedisi Sebitloane
- Discipline of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4002, South Africa;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| |
Collapse
|
8
|
Fan J, Zhang M, Wu H, Ye Z, Wang L. Estrogen Promotes Endometrial Cancer Development by Modulating ZNF626, SLK, and RFWD3 Gene Expression and Inducing Immune Inflammatory Changes. Biomedicines 2025; 13:498. [PMID: 40002911 PMCID: PMC11853163 DOI: 10.3390/biomedicines13020498] [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: 12/23/2024] [Revised: 01/24/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Elevated estrogen has been found to contribute to the pathological development of endometrial cancer (EC), potentially through alterations in the tumor inflammatory immune microenvironment. However, the exact mechanisms underlying this process remain unclear. Methods: Bioinformatics was used to identify differentially expressed genes, analyze pathway enrichment, and assess their correlation with immune cell infiltration. Ishikawa cells and ECC-1 cells were stimulated with estradiol (E2) or the selective estrogen receptor modulator Arzoxifene, and qPCR was performed to measure gene expression changes. CCK8 and FACS assays were used to analyze cell cycle alterations, while Western blotting (WB) was used to evaluate apoptosis. Results: ZNF626 and SLK were highly expressed in EC tissues, whereas RFWD3 expression was downregulated. Immune cell infiltration analysis revealed a positive correlation between ZNF626 and M2 macrophages, while SLK was negatively correlated with M1 macrophages, memory B cells, and plasma cells. RFWD3 showed more complex correlations with multiple immune cell phenotypes, including T cells. E2 stimulation resulted in the increased expression of ZNF626 and SLK, while RFWD3 expression decreased. This was accompanied by enhanced cell proliferation and suppressed apoptosis. In contrast, Arzoxifene stimulation produced the opposite effects. Conclusions: Estrogen promotes cell proliferation and inhibits apoptosis by upregulating ZNF626 and SLK, while downregulating RFWD3. Furthermore, estrogen induces a shift in the tumor microenvironment, characterized by a reduction in memory CD4+ T cells and a transition from M1 to M2 macrophage phenotypes, thus facilitating the onset and progression of EC.
Collapse
Affiliation(s)
- Jiuming Fan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Mengyao Zhang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Huailiang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Zehua Ye
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Liyan Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| |
Collapse
|
9
|
Geetha AVS, Harithpriya K, Ganesan K, Ramkumar KM. Exploring the Role of Hypoxia and HIF-1α in the Intersection of Type 2 Diabetes Mellitus and Endometrial Cancer. Curr Oncol 2025; 32:106. [PMID: 39996906 PMCID: PMC11854729 DOI: 10.3390/curroncol32020106] [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: 12/22/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetes and Cancer are the most complex chronic diseases, accounting for significant global mortality and morbidity. The association between Type 2 DM (T2DM) and endometrial cancer (EC) is multifaced, sharing numerous risk factors, including insulin resistance, obesity, hypoxia, and oxidative stress. Hypoxia plays a vital role in T2DM pathogenesis by altering the insulin level and pancreatic β-cell failure through an imbalance between antioxidant enzymes and cellular oxidative levels, while chronic inflammation contributes to EC malignancy. HIF-1α is a potent transcription factor involved in modulating cellular responses to hypoxia within the disease environment. Targeting the HIF-1α signaling cascade, a major metabolic regulator may contribute to advanced therapeutic advances. This review focuses on the association between T2DM and EC, especially focusing on hypoxia and HIF signaling pathways. These intersect with key pathways involved in T2DM and EC pathology, such as insulin signaling, PI3K/AKT, mTOR pathway, MUC1/HIF-1α pathway, and hormonal imbalance. Understanding this complex relationship paves the way for future researchers to develop HIF-1α-targeted therapies that could lead to novel combination therapies to treat these comorbid conditions.
Collapse
Affiliation(s)
- Alagappan V. S. Geetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (A.V.S.G.); (K.H.)
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (A.V.S.G.); (K.H.)
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (A.V.S.G.); (K.H.)
| |
Collapse
|
10
|
Raza M, Rajan AR, Kennedy BB, Reznicek TE, Oruji F, Mirza S, Rowley MJ, Kristiansen G, Datta K, Mohapatra BC, Band H, Band V. ECD, a novel androgen receptor target promotes prostate cancer tumorigenesis by regulating glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635534. [PMID: 39975152 PMCID: PMC11838420 DOI: 10.1101/2025.01.30.635534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Androgen receptor (AR)-mediated signaling is essential for PC tumorigenesis. In TCGA database we observed a positive correlation between ECD and AR expression. Consistently, Dihydrotestosterone (DHT) treatment of PC cell lines increased ECD mRNA and protein levels, and AR knockdown (KD) reduced ECD expression. Bioinformatic analysis predicted three consensus androgen response elements in the ECD promoter, and DHT treatment increased AR occupancy at the ECD promoter, and enhanced ECD promoter activity. Enzalutamide treatment decreased ECD levels, and ECD knockout (KO) in PC cells reduced oncogenic traits, suggesting a functional role of ECD to maintain PC oncogenesis. ECD mRNA and protein are overexpressed in PC patient tissues, and its overexpression predicts shorter survival. Overexpression of ECD in PC cell lines enhanced the oncogenic traits in vitro and developed faster and larger highly proliferative xenograft tumors. RNA-seq analysis of mouse tumors revealed increase mRNA levels of several glycolytic genes. ECD associates with mRNA of several key glycolytic genes and is required for their stability, consistent with our recent demonstration of ECD as an RNA binding protein. Higher glucose uptake and glycolysis was seen upon ECD OE in PC cells. Together, we demonstrate role of a novel AR target gene ECD in PC tumorigenesis.
Collapse
|
11
|
Sachani P, Dhande R, Parihar P, Kasat PR, Bedi GN, Pradeep U, Kothari P, Mapari SA. Enhancing the Understanding of Breast Vascularity Through Insights From Dynamic Contrast-Enhanced Magnetic Resonance Imaging: A Comprehensive Review. Cureus 2024; 16:e70226. [PMID: 39463566 PMCID: PMC11512160 DOI: 10.7759/cureus.70226] [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: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Breast vascularity plays a crucial role in both physiological and pathological processes, particularly in the development and progression of breast cancer. Understanding vascular changes within breast tissue is essential for accurate diagnosis, treatment planning, and monitoring therapeutic response. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has emerged as a valuable tool for evaluating breast vascularity due to its ability to provide detailed functional and morphological insights. DCE-MRI utilizes contrast agents to highlight blood flow and vessel permeability, making it especially useful in differentiating between benign and malignant lesions. This review explores the significance of DCE-MRI in breast vascularity assessment, highlighting its principles, clinical applications, and role in detecting malignancy through vascular changes. We also examine its utility in monitoring treatment response and quantitative analysis of perfusion metrics such as Ktrans and extracellular-extravascular volume (Ve). While DCE-MRI offers remarkable diagnostic accuracy, challenges remain regarding its cost, accessibility, and potential overlap of enhancement patterns between benign and malignant conditions. The review further discusses emerging technologies and future directions for DCE-MRI, including advanced imaging techniques and machine learning-based quantification. Overall, DCE-MRI stands out as a powerful tool in the comprehensive evaluation of breast vascularity, with significant potential to improve patient outcomes in breast cancer management.
Collapse
Affiliation(s)
- Pratiksha Sachani
- Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Rajasbala Dhande
- Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Pratapsingh Parihar
- Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Paschyanti R Kasat
- Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Gautam N Bedi
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Utkarsh Pradeep
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | | | - Smruti A Mapari
- Obstetrics and Gynecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
12
|
Ye J, Wei B, Zhou G, Xu Y, He Y, Hu X, Chen X, Zhang G, Liu H. Multi-dimensional characterization of apoptosis in the tumor microenvironment and therapeutic relevance in melanoma. Cell Oncol (Dordr) 2024; 47:1333-1353. [PMID: 38502270 PMCID: PMC11322377 DOI: 10.1007/s13402-024-00930-0] [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] [Accepted: 02/23/2024] [Indexed: 03/21/2024] Open
Abstract
PURPOSE Melanoma is widely utilized as a prominent model for the development of immunotherapy, thought an inadequate immune response can occur. Moreover, the development of apoptosis-related therapies and combinations with other therapeutic strategies is impeded by the limited understanding of apoptosis's role within diverse tumor immune microenvironments (TMEs). METHODS Here, we constructed an apoptosis-related tumor microenvironment signature (ATM) and employ multi-dimensional analysis to understand the roles of apoptosis in tumor microenvironment. We further assessed the clinical applications of ATM in nine independent cohorts, and anticipated the impact of ATM on cellular drug response in cultured cells. RESULTS Our ATM model exhibits robust performance in survival prediction in multiple melanoma cohorts. Different ATM groups exhibited distinct molecular signatures and biological processes. The low ATM group exhibited significant enrichment in B cell activation-related pathways. What's more, plasma cells showed the lowest ATM score, highlighting their role as pivotal contributors in the ATM model. Mechanistically, the analysis of the interplay between plasma cells and other immune cells elucidated their crucial role in orchestrating an effective anti-tumor immune response. Significantly, the ATM signature exhibited associations with therapeutic efficacy of immune checkpoint blockade and the drug sensitivity of various agents, including FDA-approved and clinically utilized drugs targeting the VEGF signaling pathway. Finally, ATM was associated with tertiary lymphoid structures (TLS), exhibiting stronger patient stratification ability compared to classical "hot tumors". CONCLUSION Our findings indicate that ATM is a prognostic factor and is associated with the immune response and drug sensitivity in melanoma.
Collapse
Affiliation(s)
- Jing Ye
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Benliang Wei
- Big Data Institute, Central South University, Changsha, Hunan, 410008, China
| | - Guowei Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Yi He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Xiheng Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Furong Laboratory, Changsha, Hunan, China.
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China.
- Big Data Institute, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
13
|
Han YJ, Liu S, Hardeman A, Rajagopal PS, Mueller J, Khramtsova G, Sanni A, Ajani M, Clayton W, Hurley IW, Yoshimatsu TF, Zheng Y, Parker J, Perou CM, Olopade OI. The VEGF-Hypoxia Signature Is Upregulated in Basal-like Breast Tumors from Women of African Ancestry and Associated with Poor Outcomes in Breast Cancer. Clin Cancer Res 2024; 30:2609-2618. [PMID: 38564595 DOI: 10.1158/1078-0432.ccr-23-1526] [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: 05/19/2023] [Revised: 11/21/2023] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Black women experience the highest breast cancer mortality rate compared with women of other racial/ethnic groups. To gain a deeper understanding of breast cancer heterogeneity across diverse populations, we examined a VEGF-hypoxia gene expression signature in breast tumors from women of diverse ancestry. EXPERIMENTAL DESIGN We developed a NanoString nCounter gene expression panel and applied it to breast tumors from Nigeria (n = 182) and the University of Chicago (Chicago, IL; n = 161). We also analyzed RNA sequencing data from Nigeria (n = 84) and The Cancer Genome Atlas (TCGA) datasets (n = 863). Patient prognosis was analyzed using multiple datasets. RESULTS The VEGF-hypoxia signature was highest in the basal-like subtype compared with other subtypes, with greater expression in Black women compared with White women. In TCGA dataset, necrotic breast tumors had higher scores for the VEGF-hypoxia signature compared with non-necrosis tumors (P < 0.001), with the highest proportion in the basal-like subtype. Furthermore, necrotic breast tumors have higher scores for the proliferation signature, suggesting an interaction between the VEGF-hypoxia signature, proliferation, and necrosis. T-cell gene expression signatures also correlated with the VEGF-hypoxia signature when testing all tumors in TCGA dataset. Finally, we found a significant association of the VEGF-hypoxia profile with poor outcomes when using all patients in the METABRIC (P < 0.0001) and SCAN-B datasets (P = 0.002). CONCLUSIONS These data provide further evidence for breast cancer heterogeneity across diverse populations and molecular subtypes. Interventions selectively targeting VEGF-hypoxia and the immune microenvironment have the potential to improve overall survival in aggressive breast cancers that disproportionately impact Black women in the African Diaspora.
Collapse
Affiliation(s)
- Yoo Jane Han
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Siyao Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Ashley Hardeman
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Padma Sheila Rajagopal
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Jeffrey Mueller
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Galina Khramtsova
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Ayodele Sanni
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Mustapha Ajani
- Department of Pathology, College of Medicine, University of Ibadan/University College Hospital, Ibadan, Oyo, Nigeria
| | - Wendy Clayton
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Ian W Hurley
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Toshio F Yoshimatsu
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Joel Parker
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
14
|
Martínez-López MF, Muslin C, Kyriakidis NC. STINGing Defenses: Unmasking the Mechanisms of DNA Oncovirus-Mediated Immune Escape. Viruses 2024; 16:574. [PMID: 38675916 PMCID: PMC11054469 DOI: 10.3390/v16040574] [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: 02/27/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
DNA oncoviruses represent an intriguing subject due to their involvement in oncogenesis. These viruses have evolved mechanisms to manipulate the host immune response, facilitating their persistence and actively contributing to carcinogenic processes. This paper describes the complex interactions between DNA oncoviruses and the innate immune system, with a particular emphasis on the cGAS-STING pathway. Exploring these interactions highlights that DNA oncoviruses strategically target and subvert this pathway, exploiting its vulnerabilities for their own survival and proliferation within the host. Understanding these interactions lays the foundation for identifying potential therapeutic interventions. Herein, we sought to contribute to the ongoing efforts in advancing our understanding of the innate immune system in oncoviral pathogenesis.
Collapse
Affiliation(s)
- Mayra F Martínez-López
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de las Américas, Quito 170503, Ecuador;
| | - Claire Muslin
- One Health Research Group, Faculty of Health Sciences, Universidad de las Américas, Quito 170503, Ecuador;
| | - Nikolaos C. Kyriakidis
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de las Américas, Quito 170503, Ecuador;
| |
Collapse
|
15
|
Nishida H, Albero AB, Onoue K, Ikegawa Y, Sulekh S, Sakizli U, Minami Y, Yonemura S, Wang YC, Yoo SK. Necrosensor: a genetically encoded fluorescent sensor for visualizing necrosis in Drosophila. Biol Open 2024; 13:bio060104. [PMID: 38156558 PMCID: PMC10836653 DOI: 10.1242/bio.060104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/15/2023] [Indexed: 12/30/2023] Open
Abstract
Historically, necrosis has been considered a passive process, which is induced by extreme stress or damage. However, recent findings of necroptosis, a programmed form of necrosis, shed a new light on necrosis. It has been challenging to detect necrosis reliably in vivo, partly due to the lack of genetically encoded sensors to detect necrosis. This is in stark contrast with the availability of many genetically encoded biosensors for apoptosis. Here we developed Necrosensor, a genetically encoded fluorescent sensor that detects necrosis in Drosophila, by utilizing HMGB1, which is released from the nucleus as a damage-associated molecular pattern (DAMP). We demonstrate that Necrosensor is able to detect necrosis induced by various stresses in multiple tissues in both live and fixed conditions. Necrosensor also detects physiological necrosis that occurs during spermatogenesis in the testis. Using Necrosensor, we discovered previously unidentified, physiological necrosis of hemocyte progenitors in the hematopoietic lymph gland of developing larvae. This work provides a new transgenic system that enables in vivo detection of necrosis in real time without any intervention.
Collapse
Affiliation(s)
- Hiroshi Nishida
- Division of Cell Physiology, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, 650-0047, Japan
| | | | - Kenta Onoue
- Laboratory for Ultrastructural Research, RIKEN BDR, Kobe, 650-0047, Japan
| | - Yuko Ikegawa
- Laboratory of Molecular Cell Biology and Development, Kyoto University, Kobe, 650-0047, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, 650-0047, Japan
| | - Shivakshi Sulekh
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, 650-0047, Japan
- Division of Developmental Biology and Regenerative Medicine, Graduate School of Medicine, Kobe University, Kobe, 650-0047, Japan
| | - Ugurcan Sakizli
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, 650-0047, Japan
- Division of Developmental Biology and Regenerative Medicine, Graduate School of Medicine, Kobe University, Kobe, 650-0047, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Shigenobu Yonemura
- Laboratory for Ultrastructural Research, RIKEN BDR, Kobe, 650-0047, Japan
- Department of Cell Biology, Tokushima University Graduate School of Medicine, Tokushima, 770-8503, Japan
| | - Yu-Chiun Wang
- Laboratory for Epithelial Morphogenesis, RIKEN BDR, Kobe, 650-0047, Japan
| | - Sa Kan Yoo
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, 650-0047, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, 650-0047, Japan
- Division of Developmental Biology and Regenerative Medicine, Graduate School of Medicine, Kobe University, Kobe, 650-0047, Japan
| |
Collapse
|
16
|
Wakonig KM, Dommerich S, Fischer T, Arens P, Hamm B, Olze H, Lerchbaumer MH. The Diagnostic Performance of Multiparametric Ultrasound in the Qualitative Assessment of Inconclusive Cervical Lymph Nodes. Cancers (Basel) 2023; 15:5035. [PMID: 37894402 PMCID: PMC10605624 DOI: 10.3390/cancers15205035] [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: 09/01/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Enlarged cervical lymph nodes (CLNs) can result from infection or malignancies, and a definitive diagnosis requires histological examination. Ultrasound (US) remains the first-line imaging modality for detection, and new US techniques may improve characterization. The aim of our study was to investigate whether the qualitative assessment of multiparametric US (mpUS) can improve diagnostic performance in the differentiation of benign and malignant CLNs. METHODS 107 CLNs in 105 patients were examined by preoperative mpUS consisting of B-mode US, color-coded duplex sonography (CCDS), shear wave elastography (SWE) and contrast-enhanced US (CEUS). US images were evaluated in consensus by two experienced US operators. Histopathological examination was used as reference standard. RESULTS SWE and CEUS combined showed the highest overall diagnostic performance (91% sensitivity, 77% specificity, 87% positive predictive value (PPV), 83% negative predictive value (NPV), 90% accuracy, χ2 (1) = 51.485, p < 0.001) compared to B-mode US and CCDS (87% sensitivity, 44% specificity, 73% PPV, 65% NPV, 73% accuracy χ2 (1) = 12.415, p < 0.001). In terms of individual techniques, SWE had higher specificity than B-mode and CCDS (71% sensitivity, 90% specificity, 92% PPV, 64% NPV, 78% accuracy, χ2 (1) = 36.115, p < 0.001), while qualitative CEUS showed the best diagnostic performance of all investigated US techniques (93% sensitivity, 85% specificity, 91% PPV, 87% NPV, 90% accuracy, χ2 (1) = 13.219, p < 0.001). Perfusion patterns, homogeneity, presence of necrosis, and malignancy differed significantly between malignant and benign CLNs (p < 0.001). CONCLUSIONS SWE and CEUS can facilitate the differentiation of inconclusive CLNs when performed to supplement B-mode US and CCDS. MpUS may thus aid the decision between surgery and a watch-and-scan strategy in enlarged CLNs.
Collapse
Affiliation(s)
- Katharina Margherita Wakonig
- Department of Otorhinolaryngology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum and Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (S.D.); (P.A.); (H.O.)
| | - Steffen Dommerich
- Department of Otorhinolaryngology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum and Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (S.D.); (P.A.); (H.O.)
| | - Thomas Fischer
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (T.F.); (B.H.); (M.H.L.)
| | - Philipp Arens
- Department of Otorhinolaryngology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum and Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (S.D.); (P.A.); (H.O.)
| | - Bernd Hamm
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (T.F.); (B.H.); (M.H.L.)
| | - Heidi Olze
- Department of Otorhinolaryngology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum and Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (S.D.); (P.A.); (H.O.)
| | - Markus Herbert Lerchbaumer
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (T.F.); (B.H.); (M.H.L.)
| |
Collapse
|
17
|
Li J, Zheng H, Vega AA, Beverly LJ, Gray BD, Pak KY, Ng CK. Evaluation of 2-deoxy-2-[ 18F]fluoro glucaric acid (FGA) as a potential PET tracer for tumor necrosis. Appl Radiat Isot 2023; 200:110988. [PMID: 37633190 DOI: 10.1016/j.apradiso.2023.110988] [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: 03/20/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
In this study, [18F]FGA was obtained by a one-step oxidation of [18F]FDG using sodium hypochlorite. The conversion from [18F]FDG to [18F]FGA was confirmed by HPLC to be over 95% using the optimal condition. A549-luciferase NSCLC xenografted mice was used for in vivo PET imaging. Prior to either saline or cisplatin treatment, there was no significant difference on tumor uptake of [18F]FGA in all mice, with an average uptake of (0.21 ± 0.16) %ID/g. After treatment, tumor uptake of [18F]FGA was not changed significantly for saline-treated mice, whereas the tumor uptake of [18F]FGA drastically increased for cisplatin-treated mice, with an average uptake of (1.63 ± 0.16) %ID/g. The ratio of tumor uptake between cisplatin-treated vs. saline-treated mice was 7.8 ± 0.2 within one week of treatment. PET imaging results were consistent with ex vivo biodistribution data. BLI showed significant light intensity suppression after treatment, indicating necrosis. Our data indicate that [18F]FGA uptake was related to tumor necrosis. [18F]FGA PET/CT imaging might be a useful tool to monitor treatment response to chemotherapy by imaging tumor necrosis.
Collapse
Affiliation(s)
- Junling Li
- Department of Radiology, University of Louisville, Louisville, KY, USA
| | - Huaiyu Zheng
- Department of Radiology, University of Louisville, Louisville, KY, USA
| | - Alexis A Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, USA; Brown Cancer Center, University of Louisville School of Medicine, 505 S. Hancock St. Rm 204, Louisville, KY, 40202, USA
| | - Levi J Beverly
- Department of Medicine and James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Brian D Gray
- Molecular Targeting Technologies, Inc., West Chester, PA, USA
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc., West Chester, PA, USA
| | - Chin K Ng
- Department of Radiology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
18
|
Sufyan M, Shokat Z, Ashfaq UA. Artificial intelligence in cancer diagnosis and therapy: Current status and future perspective. Comput Biol Med 2023; 165:107356. [PMID: 37688994 DOI: 10.1016/j.compbiomed.2023.107356] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/21/2023] [Accepted: 08/12/2023] [Indexed: 09/11/2023]
Abstract
Artificial intelligence (AI) in healthcare plays a pivotal role in combating many fatal diseases, such as skin, breast, and lung cancer. AI is an advanced form of technology that uses mathematical-based algorithmic principles similar to those of the human mind for cognizing complex challenges of the healthcare unit. Cancer is a lethal disease with many etiologies, including numerous genetic and epigenetic mutations. Cancer being a multifactorial disease is difficult to be diagnosed at an early stage. Therefore, genetic variations and other leading factors could be identified in due time through AI and machine learning (ML). AI is the synergetic approach for mining the drug targets, their mechanism of action, and drug-organism interaction from massive raw data. This synergetic approach is also facing several challenges in data mining but computational algorithms from different scientific communities for multi-target drug discovery are highly helpful to overcome the bottlenecks in AI for drug-target discovery. AI and ML could be the epicenter in the medical world for the diagnosis, treatment, and evaluation of almost any disease in the near future. In this comprehensive review, we explore the immense potential of AI and ML when integrated with the biological sciences, specifically in the context of cancer research. Our goal is to illuminate the many ways in which AI and ML are being applied to the study of cancer, from diagnosis to individualized treatment. We highlight the prospective role of AI in supporting oncologists and other medical professionals in making informed decisions and improving patient outcomes by examining the intersection of AI and cancer control. Although AI-based medical therapies show great potential, many challenges must be overcome before they can be implemented in clinical practice. We critically assess the current hurdles and provide insights into the future directions of AI-driven approaches, aiming to pave the way for enhanced cancer interventions and improved patient care.
Collapse
Affiliation(s)
- Muhammad Sufyan
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan.
| | - Zeeshan Shokat
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan.
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan.
| |
Collapse
|
19
|
Samaratunga H, Egevad L, Delahunt B. Not all tumour necrosis is granular necrosis. Pathology 2023; 55:903-904. [PMID: 37543492 DOI: 10.1016/j.pathol.2023.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/15/2023] [Indexed: 08/07/2023]
Affiliation(s)
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Brett Delahunt
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
20
|
Song B, Hwang SI, Lee HJ, Lee H, Oh JJ, Lee S, Hong SK, Byun SS, Kim JK. Computer tomography-based shape of tumor contour and texture of tumor heterogeneity are independent prognostic indicators for clinical T1b-T2 renal cell carcinoma. World J Urol 2023; 41:2723-2734. [PMID: 37530807 DOI: 10.1007/s00345-023-04543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 07/19/2023] [Indexed: 08/03/2023] Open
Abstract
PURPOSE To evaluate association between computer tomography (CT)-based features of renal cell carcinoma (RCC) and survival outcomes. METHODS Data of 958 patients with clinical T1b-T2 RCC who underwent partial/radical nephrectomy from June 2003 to March 2022 were retrospectively evaluated. CT images of patients were reviewed by two radiologists for texture analysis of tumor heterogeneity and shape analysis of tumor contour. Patients were divided into three groups according to patterns of CT-based features: (1) favorable feature group (n = 117); (2) intermediate feature group (n = 606); and (3) unfavorable feature group (n = 235). Kaplan-Meier survival analysis and multivariate Cox regression analysis were performed to evaluate overall survival (OS), cancer-specific survival (CSS), and recurrence-free survival (RFS). RESULTS RCCs with unfavorable CT-based feature showed larger size on CT, higher nuclear grade, higher rate of histologic necrosis, and higher rate of capsular invasion than those in the other two groups (all p < 0.001). Unfavorable feature was associated with poorer OS (p = 0.001), CSS (p < 0.001), and RFS (p < 0.001) on Kaplan-Meier analysis. In multivariate analysis, intermediate and unfavorable features were independent predictors for recurrence (hazard ratio [HR] 2.51, 95% confidence interval [CI] 1.09-5.79, p = 0.031 and HR 3.71, 95% CI 1.58-8.73, p = 0.003, respectively), but not for overall death or RCC-specific death. CONCLUSIONS A combination of irregular tumor contour feature with heterogeneous tumor texture feature on CT is associated with poor RFS in clinical T1b-T2 RCC preoperatively.
Collapse
Affiliation(s)
- Byeongdo Song
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Sung Il Hwang
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam-si, South Korea
| | - Hak Jong Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam-si, South Korea
| | - Hakmin Lee
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jong Jin Oh
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sangchul Lee
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sung Kyu Hong
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, South Korea
| | - Seok-Soo Byun
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jung Kwon Kim
- Department of Urology, Seoul National University Bundang Hospital, 166, Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
- Department of Urology, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
21
|
de Alencar MVOB, Islam MT, da Mata AMOF, Dos Reis AC, de Lima RMT, de Oliveira Ferreira JR, de Castro E Sousa JM, Ferreira PMP, de Carvalho Melo-Cavalcante AA, Rauf A, Hemeg HA, Alsharif KF, Khan H. Anticancer effects of phytol against Sarcoma (S-180) and Human Leukemic (HL-60) cancer cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:80996-81007. [PMID: 37308630 DOI: 10.1007/s11356-023-28036-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/29/2023] [Indexed: 06/14/2023]
Abstract
Phytol (Pyt), a diterpenoid, possesses many important bioactivities. This study evaluates the anticancer effects of Pyt on sarcoma 180 (S-180) and human leukemia (HL-60) cell lines. For this purpose, cells were treated with Pyt (4.72, 7.08, or 14.16 μM) and a cell viability assay was performed. Additionally, the alkaline comet assay and micronucleus test with cytokinesis were also performed using doxorubicin (6 μM) and hydrogen peroxide (10 mM) as positive controls and stressors, respectively. Results revealed that Pyt significantly reduced the viability and rate of division in S-180 and HL-60 cells with IC50 values of 18.98 ± 3.79 and 1.17 ± 0.34 μM, respectively. Pyt at 14.16 μM exerted aneugenic and/or clastogenic effects in S-180 and HL-60 cells, where the number of micronuclei and other nuclear abnormalities (e.g., nucleoplasmic bridges and nuclear buds) were frequently observed. Moreover, Pyt at all concentrations induced apoptosis and showed necrosis at 14.16 μM, suggesting its anticancer effects on the tested cancer cell lines. Taken together, Pyt showed promising anticancer effects, possibly through inducing apoptosis and necrosis mechanisms, and it exerted aneugenic and/or clastogenic effects on the S-180 and HL-60 cell lines.
Collapse
Affiliation(s)
- Marcus Vinícius Oliveira Barros de Alencar
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Biomedical Sciences Research and Innovation Laboratory, Postgraduate Program in Biotechnology, INTA University Center, Sobral, 62.011-230, Brazil
- Laboratory of Toxicological Genetics, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil
| | - Muhammad Torequl Islam
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Ana Maria Oliveira Ferreira da Mata
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Laboratory of Toxicological Genetics, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil
| | - Antonielly Campinho Dos Reis
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Laboratory of Toxicological Genetics, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil
| | - Rosália Maria Torres de Lima
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Laboratory of Toxicological Genetics, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil
| | | | - João Marcelo de Castro E Sousa
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Laboratory of Toxicological Genetics, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Laboratory of Experimental Cancerology, Department of Biophysics and Physiology, Federal University of Piauí, Teresina, 64.049-550, Brazil
| | - Ana Amélia de Carvalho Melo-Cavalcante
- Postgraduate Program in Biotechnology (RENORBIO), Federal University of Piauí, Teresina, 64.049-550, Brazil
- Laboratory of Toxicological Genetics, Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, Swabi, Khyber Pakhtunkhwa, 23430, Pakistan
| | - Hassan A Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Al-Medinah Al-Monawara, 41411, Saudi Arabia
| | - Khalaf F Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, Taif, 21944, Saudi Arabia
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
| |
Collapse
|
22
|
Wang H, Yan R, Li Z, Wang B, Jin X, Guo Z, Liu W, Zhang M, Wang K, Guo J, Han D. Quantitative dynamic contrast-enhanced parameters and intravoxel incoherent motion facilitate the prediction of TP53 status and risk stratification of early-stage endometrial carcinoma. Radiol Oncol 2023; 57:257-269. [PMID: 37341203 DOI: 10.2478/raon-2023-0023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/06/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND The aim of the study was to investigate the value of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and intravoxel incoherent motion (IVIM) in differentiating TP53-mutant from wild type, low-risk from non-low-risk early-stage endometrial carcinoma (EC). PATIENTS AND METHODS A total of 74 EC patients underwent pelvic MRI. Parameters volume transfer constant (Ktrans), rate transfer constant (Kep), the volume of extravascular extracellular space per unit volume of tissue (Ve), true diffusion coefficient (D), pseudo-diffusion coefficient (D*), and microvascular volume fraction (f) were compared. The combination of parameters was investigated by logistic regression and evaluated by bootstrap (1000 samples), receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA). RESULTS In the TP53-mutant group, Ktrans and Kep were higher and D was lower than in the TP53-wild group; Ktrans, Ve, f, and D were lower in the non-low-risk group than in the low-risk group (all P < 0.05). In the identification of TP53-mutant and TP53-wild early-stage EC, Ktrans and D were independent predictors, and the combination of them had an optimal diagnostic efficacy (AUC, 0.867; sensitivity, 92.00%; specificity, 80.95%), which was significantly better than D (Z = 2.169, P = 0.030) and Ktrans (Z = 2.572, P = 0.010). In the identification of low-risk and non-low-risk early-stage EC, Ktrans, Ve, and f were independent predictors, and the combination of them had an optimal diagnostic efficacy (AUC, 0.947; sensitivity, 83.33%; specificity, 93.18%), which was significantly better than D (Z = 3.113, P = 0.002), f (Z = 4.317, P < 0.001), Ktrans (Z = 2.713, P = 0.007), and Ve (Z = 3.175, P = 0.002). The calibration curves showed that the above two combinations of independent predictors, both have good consistency, and DCA showed that these combinations were reliable clinical prediction tools. CONCLUSIONS Both DCE-MRI and IVIM facilitate the prediction of TP53 status and risk stratification in early-stage EC. Compare with each single parameter, the combination of independent predictors provided better predictive power and may serve as a superior imaging marker.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Ruifang Yan
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Zhong Li
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Beiran Wang
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Xingxing Jin
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Zhenfang Guo
- Department of Neurology, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Wangyi Liu
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Meng Zhang
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Kaiyu Wang
- MR Research China, GE Healthcare, Beijing, China
| | - Jinxia Guo
- MR Research China, GE Healthcare, Beijing, China
| | - Dongming Han
- Department of MR, the First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| |
Collapse
|
23
|
Kastinen M, Sirniö P, Elomaa H, Ahtiainen M, Väyrynen SA, Herzig KH, Meriläinen S, Aro R, Häivälä R, Rautio T, Saarnio J, Wirta EV, Helminen O, Seppälä TT, Kuopio T, Böhm J, Tuomisto A, Mecklin JP, Mäkinen MJ, Väyrynen JP. Immunological and prognostic significance of tumour necrosis in colorectal cancer. Br J Cancer 2023; 128:2218-2226. [PMID: 37031328 PMCID: PMC10241859 DOI: 10.1038/s41416-023-02258-2] [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: 11/03/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/10/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) causes the second most cancer deaths worldwide, but the disease course varies according to tumour characteristics and immunological factors. Our objective was to examine the associations of tumour necrosis with tumour characteristics, immune cell infiltrates, serum cytokine concentrations, as well as prognosis in CRC. METHODS Three independent CRC cohorts, including 1413 patients, were analysed. Associations of the areal percentage of tumour necrosis with clinicopathologic parameters, tumour infiltrating immune cells, cytokine concentrations in systemic and mesenteric vein blood, and survival were examined. RESULTS Higher tumour necrosis percentage associated with shorter colorectal cancer-specific survival independent of tumour grade, T, N or M-class, mismatch repair status, BRAF status, and other possible confounding factors. In the largest cohort (N = 1100), the HR for high tumour necrosis percentage (≥40% vs. <3%) was 3.22 (95% CI 1.68-6.17, Ptrend < 0.0001). Tumour necrosis percentage positively correlated with peripheral serum levels of CXCL8, a proinflammatory chemokine, and negatively correlated with mesenteric serum levels of CXCL10 and mast cell densities in the invasive margin of the tumour. CONCLUSIONS Our results support the value of tumour necrosis as a prognostic factor in colorectal cancer. CXCL8 may have a role in the systemic effects of tumour necrosis.
Collapse
Affiliation(s)
- Meeri Kastinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Päivi Sirniö
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Hanna Elomaa
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Education and Research, Wellbeing services county of Central Finland, Jyväskylä, Finland
| | - Maarit Ahtiainen
- Department of Pathology, Wellbeing services county of Central Finland, Jyväskylä, Finland
| | - Sara A Väyrynen
- Department of Internal Medicine, Oulu University Hospital, Oulu, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center Oulu, University of Oulu, Oulu University Hospital, Oulu, Finland
- Department of Pediatric Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
| | - Sanna Meriläinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Raila Aro
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Reetta Häivälä
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Tero Rautio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Juha Saarnio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Erkki-Ville Wirta
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Olli Helminen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Toni T Seppälä
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics, Research Program Unit, University of Helsinki, Helsinki, Finland
| | - Teijo Kuopio
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Pathology, Wellbeing services county of Central Finland, Jyväskylä, Finland
| | - Jan Böhm
- Department of Pathology, Wellbeing services county of Central Finland, Jyväskylä, Finland
| | - Anne Tuomisto
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Jukka-Pekka Mecklin
- Department of Education and Research, Wellbeing services county of Central Finland, Jyväskylä, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Markus J Mäkinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Juha P Väyrynen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland.
| |
Collapse
|
24
|
Nandi S, Mondal A, Ghosh A, Mukherjee S, Das C. Lnc-ing epigenetic mechanisms with autophagy and cancer drug resistance. Adv Cancer Res 2023; 160:133-203. [PMID: 37704287 DOI: 10.1016/bs.acr.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Long noncoding RNAs (lncRNAs) comprise a diverse class of RNA molecules that regulate various physiological processes and have been reported to be involved in several human pathologies ranging from neurodegenerative disease to cancer. Therapeutic resistance is a major hurdle for cancer treatment. Over the past decade, several studies has emerged on the role of lncRNAs in cancer drug resistance and many trials have been conducted employing them. LncRNAs also regulate different cell death pathways thereby maintaining a fine balance of cell survival and death. Autophagy is a complex cell-killing mechanism that has both cytoprotective and cytotoxic roles. Similarly, autophagy can lead to the induction of both chemosensitization and chemoresistance in cancer cells upon therapeutic intervention. Recently the role of lncRNAs in the regulation of autophagy has also surfaced. Thus, lncRNAs can be used in cancer therapeutics to alleviate the challenges of chemoresistance by targeting the autophagosomal axis. In this chapter, we discuss about the role of lncRNAs in autophagy-mediated cancer drug resistance and its implication in targeted cancer therapy.
Collapse
Affiliation(s)
- Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India
| | - Aritra Ghosh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Indian Institute of Science Education and Research, Kolkata, India
| | - Shravanti Mukherjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
25
|
Prashantha K, Krishnappa A, Muthappa M. 3D bioprinting of gastrointestinal cancer models: A comprehensive review on processing, properties, and therapeutic implications. Biointerphases 2023; 18:020801. [PMID: 36963961 DOI: 10.1116/6.0002372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023] Open
Abstract
Gastrointestinal tract (GIT) malignancies are an important public health problem considering the increased incidence in recent years and the high morbidity and mortality associated with it. GIT malignancies constitute 26% of the global cancer incidence burden and 35% of all cancer-related deaths. Gastrointestinal cancers are complex and heterogenous diseases caused by the interplay of genetic and environmental factors. The tumor microenvironment (TME) of gastrointestinal tract carcinomas is dynamic and complex; it cannot be recapitulated in the basic two-dimensional cell culture systems. In contrast, three-dimensional (3D) in vitro models can mimic the TME more closely, enabling an improved understanding of the microenvironmental cues involved in the various stages of cancer initiation, progression, and metastasis. However, the heterogeneity of the TME is incompletely reproduced in these 3D culture models, as they fail to regulate the orientation and interaction of various cell types in a complex architecture. To emulate the TME, 3D bioprinting has emerged as a useful technique to engineer cancer tissue models. Bioprinted cancer tissue models can potentially recapitulate cancer pathology and increase drug resistance in an organ-mimicking 3D environment. In this review, we describe the 3D bioprinting methods, bioinks, characterization of 3D bioprinted constructs, and their application in developing gastrointestinal tumor models that integrate their microenvironment with different cell types and substrates, as well as bioprinting modalities and their application in therapy and drug screening. We review prominent studies on the 3D bioprinted esophageal, hepatobiliary, and colorectal cancer models. In addition, this review provides a comprehensive understanding of the cancer microenvironment in printed tumor models, highlights current challenges with respect to their clinical translation, and summarizes future perspectives.
Collapse
Affiliation(s)
- Kalappa Prashantha
- Centre for Research and Innovation, Adichunchanagiri School of Natural Sciences, Adichunchanagiri University, BGSIT, B.G. Nagara, Mandya District 571448, Karnataka, India
| | - Amita Krishnappa
- Department of Pathology, Adichunchanagiri Institute of Medicinal Sciences Adichunchanagiri University, B.G. Nagara, Mandya District 571448, Karnataka, India
| | - Malini Muthappa
- Department of Physiology, Adichunchanagiri Institute of Medicinal Sciences Adichunchanagiri University, B.G. Nagara, Mandya District 571448, Karnataka, India
| |
Collapse
|
26
|
Yamamoto A, Huang Y, Krajina BA, McBirney M, Doak AE, Qu S, Wang CL, Haffner MC, Cheung KJ. Metastasis from the tumor interior and necrotic core formation are regulated by breast cancer-derived angiopoietin-like 7. Proc Natl Acad Sci U S A 2023; 120:e2214888120. [PMID: 36853945 PMCID: PMC10013750 DOI: 10.1073/pnas.2214888120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/06/2023] [Indexed: 03/01/2023] Open
Abstract
Necrosis in the tumor interior is a common feature of aggressive cancers that is associated with poor clinical prognosis and the development of metastasis. How the necrotic core promotes metastasis remains unclear. Here, we report that emergence of necrosis inside the tumor is correlated temporally with increased tumor dissemination in a rat breast cancer model and in human breast cancer patients. By performing spatially focused transcriptional profiling, we identified angiopoietin-like 7 (Angptl7) as a tumor-specific factor localized to the perinecrotic zone. Functional studies showed that Angptl7 loss normalizes central necrosis, perinecrotic dilated vessels, metastasis, and reduces circulating tumor cell counts to nearly zero. Mechanistically, Angptl7 promotes vascular permeability and supports vascular remodeling in the perinecrotic zone. Taken together, these findings show that breast tumors actively produce factors controlling central necrosis formation and metastatic dissemination from the tumor core.
Collapse
Affiliation(s)
- Ami Yamamoto
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA98195
| | - Yin Huang
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Brad A. Krajina
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Margaux McBirney
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Andrea E. Doak
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA98195
| | - Sixuan Qu
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Carolyn L. Wang
- Department of Radiology, University of Washington School of Medicine, Seattle, WA98195
| | - Michael C. Haffner
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA98109
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Kevin J. Cheung
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| |
Collapse
|
27
|
Burdett NL, Willis MO, Alsop K, Hunt AL, Pandey A, Hamilton PT, Abulez T, Liu X, Hoang T, Craig S, Fereday S, Hendley J, Garsed DW, Milne K, Kalaria S, Marshall A, Hood BL, Wilson KN, Conrads KA, Pishas KI, Ananda S, Scott CL, Antill Y, McNally O, Mileshkin L, Hamilton A, Au-Yeung G, Devereux L, Thorne H, Bild A, Bateman NW, Maxwell GL, Chang JT, Conrads TP, Nelson BH, Bowtell DDL, Christie EL. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer. Nat Genet 2023; 55:437-450. [PMID: 36849657 DOI: 10.1038/s41588-023-01320-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/26/2023] [Indexed: 03/01/2023]
Abstract
High-grade serous ovarian cancer (HGSC) is frequently characterized by homologous recombination (HR) DNA repair deficiency and, while most such tumors are sensitive to initial treatment, acquired resistance is common. We undertook a multiomics approach to interrogate molecular diversity in end-stage disease, using multiple autopsy samples collected from 15 women with HR-deficient HGSC. Patients had polyclonal disease, and several resistance mechanisms were identified within most patients, including reversion mutations and HR restoration by other means. We also observed frequent whole-genome duplication and global changes in immune composition with evidence of immune escape. This analysis highlights diverse evolutionary changes within HGSC that evade therapy and ultimately overwhelm individual patients.
Collapse
Affiliation(s)
- Nikki L Burdett
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia
| | | | - Kathryn Alsop
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Allison L Hunt
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, Annandale, Victoria, USA
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Ahwan Pandey
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Tamara Abulez
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Xuan Liu
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
| | - Therese Hoang
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Stuart Craig
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sian Fereday
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joy Hendley
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Dale W Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Katy Milne
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Shreena Kalaria
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Ashley Marshall
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Brian L Hood
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Katlin N Wilson
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Kelly A Conrads
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Kathleen I Pishas
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Sumitra Ananda
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Medical Oncology, Western Health, St Albans, Victoria, Australia
- Department of Medicine, Western Health, The University of Melbourne, St Albans, Victoria, Australia
- Epworth Healthcare, East Melbourne, Victoria, Australia
| | - Clare L Scott
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Yoland Antill
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
- Cabrini Health, Malvern, Victoria, Australia
- Department of Medical Oncology, Peninsula health, Frankston, Victoria, Australia
| | - Orla McNally
- The Royal Women's Hospital, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Linda Mileshkin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Anne Hamilton
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- The Royal Women's Hospital, Parkville, Victoria, Australia
| | - George Au-Yeung
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Lisa Devereux
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Heather Thorne
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Andrea Bild
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- The John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University, Bethesda, MD, USA
| | - G Larry Maxwell
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, Annandale, Victoria, USA
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University, Bethesda, MD, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, TX, USA
| | - Thomas P Conrads
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University and Walter Reed National Military Medical Center, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- The John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University, Bethesda, MD, USA
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
28
|
Alves ÂVF, Melo CR, Chagas-Neto JL, Amaral RG, Ambrósio SR, Moreira MR, Veneziani RCS, Cardoso JC, Severino P, Gondak RO, Souto EB, de Albuquerque-Júnior RLC. Ent-kaurenoic acid-enriched Mikania glomerata leaves-complexed β-cyclodextrin: Pharmaceutical development and in vivo antitumor activity in a sarcoma 180 mouse model. Int J Pharm 2023; 631:122497. [PMID: 36529360 DOI: 10.1016/j.ijpharm.2022.122497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
The extract obtained from Mikania glomerata leaves rich in ent-kaurenoic acid (ERKA) shows cytotoxic activity in vitro, but its hydrophobic nature and thermosensitivity are issues to be solved prior to in vivo antitumor studies. The purpose of this study was to investigate the antitumor activity of inclusion complexes formed between ERKA and β-cyclodextrin (ERKA:β-CD) in rodents. ERKA:β-CD complexes obtained by malaxation (MX) and co-evaporation (CE) methods were firstly characterized regarding their physical properties, encapsulation efficiency, and cytotoxicity againts L929 cells. The antitumor activity study was then performed in mice with sarcoma 180 treated with saline, 5-fluouracil (5FU) and ERKA:β-CD at 30, 100 and 300 µg/kg. The weight, volume, percentage of inhibition growth, gross and pathological features and positivity for TUNEL, ki67, NFκB and NRF2 in the tumors were assessed. Serum lactate-dehydrogenase activity (LDH), white blood cells count (WBC) and both gross and pathological features of the liver, kidneys and spleen were also evaluated. The formation of the inclusion complexes was confirmed by thermal analysis and FTIR, and they were non-toxic for L929 cells. The MX provided a better complexation efficiency. ERKA:β-CD300 promoted significant tumor growth inhibition, and attenuated the tumor mitotic activity and necrosis content, comparable to 5-fluorouracil. ERKA:β-CD300 also increased TUNEL-detected cell death, reduced Ki67 and NF-kB immunoexpression, and partially inhibited the serum LDH activity. No side effect was observed in ERKA:β-CD300-treated animals. The ERKA:β-CD inclusion complexes at 300 µg/kg displays antitumour activity in mice with low systemic toxicity, likely due to inhibition on the NF-kB signaling pathway and LDH activity.
Collapse
Affiliation(s)
- Ângela V F Alves
- Institute of Technology and Research, University of Tiradentes, Av. Murilo Dantas, 300, Bairro Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - Carlisson R Melo
- Institute of Technology and Research, University of Tiradentes, Av. Murilo Dantas, 300, Bairro Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - José L Chagas-Neto
- School of Dentistry, University of Tiradentes, Av. Murilo Dantas, 300, Bairro Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - Ricardo G Amaral
- Department of Physiology, Federal University of Sergipe, 49100-000 São Cristóvão, Sergipe, Brazil
| | - Sérgio R Ambrósio
- Research Group in Exact and Technological, University of Franca, Av. Dr. Armando de Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Monique R Moreira
- Research Group in Exact and Technological, University of Franca, Av. Dr. Armando de Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Rodrigo C S Veneziani
- Research Group in Exact and Technological, University of Franca, Av. Dr. Armando de Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Juliana C Cardoso
- Institute of Technology and Research, University of Tiradentes, Av. Murilo Dantas, 300, Bairro Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - Patricia Severino
- Institute of Technology and Research, University of Tiradentes, Av. Murilo Dantas, 300, Bairro Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - Rogério O Gondak
- Department of Pathology, Federal University of Santa Catarina, R. Delfino Conti, S/N, 88040-370 Florianópolis, Santa Catarina, Brazil
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; REQUIMTE/UCIBIO, Faculty of Pharmacy of University of Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - Ricardo L C de Albuquerque-Júnior
- Department of Pathology, Federal University of Santa Catarina, R. Delfino Conti, S/N, 88040-370 Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
29
|
Stejskal P, Goodarzi H, Srovnal J, Hajdúch M, van ’t Veer LJ, Magbanua MJM. Circulating tumor nucleic acids: biology, release mechanisms, and clinical relevance. Mol Cancer 2023; 22:15. [PMID: 36681803 PMCID: PMC9862574 DOI: 10.1186/s12943-022-01710-w] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/29/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Despite advances in early detection and therapies, cancer is still one of the most common causes of death worldwide. Since each tumor is unique, there is a need to implement personalized care and develop robust tools for monitoring treatment response to assess drug efficacy and prevent disease relapse. MAIN BODY Recent developments in liquid biopsies have enabled real-time noninvasive monitoring of tumor burden through the detection of molecules shed by tumors in the blood. These molecules include circulating tumor nucleic acids (ctNAs), comprising cell-free DNA or RNA molecules passively and/or actively released from tumor cells. Often highlighted for their diagnostic, predictive, and prognostic potential, these biomarkers possess valuable information about tumor characteristics and evolution. While circulating tumor DNA (ctDNA) has been in the spotlight for the last decade, less is known about circulating tumor RNA (ctRNA). There are unanswered questions about why some tumors shed high amounts of ctNAs while others have undetectable levels. Also, there are gaps in our understanding of associations between tumor evolution and ctNA characteristics and shedding kinetics. In this review, we summarize current knowledge about ctNA biology and release mechanisms and put this information into the context of tumor evolution and clinical utility. CONCLUSIONS A deeper understanding of the biology of ctDNA and ctRNA may inform the use of liquid biopsies in personalized medicine to improve cancer patient outcomes.
Collapse
Affiliation(s)
- Pavel Stejskal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital in Olomouc, Olomouc, 779 00 Czech Republic
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158 USA
- Department of Urology, University of California San Francisco, San Francisco, CA 94158 USA
| | - Josef Srovnal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital in Olomouc, Olomouc, 779 00 Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital in Olomouc, Olomouc, 779 00 Czech Republic
| | - Laura J. van ’t Veer
- Department of Laboratory Medicine, University of California San Francisco, 2340 Sutter Street, San Francisco, CA USA
| | - Mark Jesus M. Magbanua
- Department of Laboratory Medicine, University of California San Francisco, 2340 Sutter Street, San Francisco, CA USA
| |
Collapse
|
30
|
Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers. Cancers (Basel) 2022; 15:cancers15010087. [PMID: 36612084 PMCID: PMC9817756 DOI: 10.3390/cancers15010087] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Lactate mediates multiple cell-intrinsic effects in cancer metabolism in terms of development, maintenance, and metastasis and is often correlated with poor prognosis. Its functions are undertaken as an energy source for neighboring carcinoma cells and serve as a lactormone for oncogenic signaling pathways. Indeed, two isomers of lactate are produced in the Warburg effect: L-lactate and D-lactate. L-lactate is the main end-production of glycolytic fermentation which catalyzes glucose, and tiny D-lactate is fabricated through the glyoxalase system. Their production inevitably affects cancer development and therapy. Here, we systematically review the mechanisms of lactate isomers production, and highlight emerging evidence of the carcinogenic biological effects of lactate and its isomers in cancer. Accordingly, therapy that targets lactate and its metabolism is a promising approach for anticancer treatment.
Collapse
|
31
|
deSouza NM, Choudhury A, Greaves M, O’Connor JPB, Hoskin PJ. Imaging hypoxia in endometrial cancer: How and why should it be done? Front Oncol 2022; 12:1020907. [PMID: 36439503 PMCID: PMC9682004 DOI: 10.3389/fonc.2022.1020907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/21/2022] [Indexed: 11/07/2023] Open
Affiliation(s)
- Nandita M. deSouza
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Imaging, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Ananya Choudhury
- Radiation Oncology, The Christie National Health Service (NHS) Foundation Trust Manchester, Manchester, United Kingdom
- The Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - James P. B. O’Connor
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Department of Imaging, The Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- The Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Peter J. Hoskin
- The Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Radiation Oncology, Mount Vernon Cancer Centre, Northwood, United Kingdom
| |
Collapse
|
32
|
Predarska I, Saoud M, Drača D, Morgan I, Komazec T, Eichhorn T, Mihajlović E, Dunđerović D, Mijatović S, Maksimović-Ivanić D, Hey-Hawkins E, Kaluđerović GN. Mesoporous Silica Nanoparticles Enhance the Anticancer Efficacy of Platinum(IV)-Phenolate Conjugates in Breast Cancer Cell Lines. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3767. [PMID: 36364539 PMCID: PMC9659259 DOI: 10.3390/nano12213767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 05/15/2023]
Abstract
The main reasons for the limited clinical efficacy of the platinum(II)-based agent cisplatin include drug resistance and significant side effects. Due to their better stability, as well as the possibility to introduce biologically active ligands in their axial positions constructing multifunctional prodrugs, creating platinum(IV) complexes is a tempting strategy for addressing these limitations. Another strategy for developing chemotherapeutics with lower toxicity relies on the ability of nanoparticles to accumulate in greater quantities in tumor tissues through passive targeting. To combine the two approaches, three platinum(IV) conjugates based on a cisplatin scaffold containing in the axial positions derivatives of caffeic and ferulic acid were prepared and loaded into SBA-15 to produce the corresponding mesoporous silica nanoparticles (MSNs). The free platinum(IV) conjugates demonstrated higher or comparable activity with respect to cisplatin against different human breast cancer cell lines, while upon immobilization, superior antiproliferative activity with markedly increased cytotoxicity (more than 1000-fold lower IC50 values) compared to cisplatin was observed. Mechanistic investigations with the most potent conjugate, cisplatin-diacetyl caffeate (1), and the corresponding MSNs (SBA-15|1) in a 4T1 mouse breast cancer cell line showed that these compounds induce apoptotic cell death causing strong caspase activation. In vivo, in BALB/c mice, 1 and SBA-15|1 inhibited the tumor growth while decreasing the necrotic area and lowering the mitotic rate.
Collapse
Affiliation(s)
- Ivana Predarska
- Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Universität Leipzig, Johannisallee 29, 04103 Leipzig, Germany
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Mohamad Saoud
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany
| | - Dijana Drača
- Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Ibrahim Morgan
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany
| | - Teodora Komazec
- Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Thomas Eichhorn
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Ekatarina Mihajlović
- Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Duško Dunđerović
- Institute of Pathology, School of Medicine, University of Belgrade, dr Subotića 1, 11000 Belgrade, Serbia
| | - Sanja Mijatović
- Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Evamarie Hey-Hawkins
- Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Universität Leipzig, Johannisallee 29, 04103 Leipzig, Germany
| | - Goran N. Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle (Saale), Germany
| |
Collapse
|
33
|
Mohsen MO, Heath M, Kramer MF, Velazquez TC, Bullimore A, Skinner MA, Speiser DE, Bachmann MF. In situ delivery of nanoparticles formulated with micron-sized crystals protects from murine melanoma. J Immunother Cancer 2022; 10:jitc-2022-004643. [PMID: 36100311 PMCID: PMC9472128 DOI: 10.1136/jitc-2022-004643] [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] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction Intratumoral injections of novel therapeutics can activate tumor antigen-specific T cells for locoregional tumor control and may even induce durable systemic protection (against distant metastases) via recirculating T cells. Here we explored the possibility of a universal immunotherapy that promotes T-cell responses in situ and beyond, upon intratumoral injection of nanoparticles formulated with micron-sized crystals. Methods Cucumber mosaic virus-like particles containing a tetanus toxin peptide (CuMVTT) were formulated with microcrystalline tyrosine (MCT) adjuvant and injected directly in B16F10 melanoma tumors. To further enhance immunogenicity, we loaded the nanoparticles with a TLR7/8 ligand and incorporated a universal tetanus toxin T-helper cell peptide. We assessed therapeutic efficacy and induction of local and systemic immune responses, including RNA sequencing, providing broad insight into the tumor microenvironment and correlates of protection. Results MCT crystals were successfully decorated with CuMVTT nanoparticles. This ‘immune-enhancer’ formed immunogenic depots in injected tumors, enhanced polyfunctional CD8+ and CD4+ T cells, and inhibited B16F10 tumor growth locally and systemically. Local inflammation and immune responses were associated with upregulation of genes involved in complement activation and collagen formation. Conclusions Our new immune-enhancer turned immunologically cold tumors into hot ones and inhibited local and distant tumor growth. This type of immunotherapy does not require the identification of (patient–individual) relevant tumor antigens. It is well tolerated, non-infectious, and affordable, and can readily be upscaled for future clinical testing and broad application in melanoma and likely other solid tumors.
Collapse
Affiliation(s)
- Mona O Mohsen
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | | - Daniel E Speiser
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Martin F Bachmann
- Department of BioMedical Research, University of Bern, Bern, Switzerland.,Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Iazzolino G, Mendibil U, Arnaiz B, Ruiz-de-Angulo A, Azkargorta M, Uribe KB, Khatami N, Elortza F, Olalde B, Gomez-Vallejo V, Llop J, Abarrategi A. Decellularization of xenografted tumors provides cell-specific in vitro 3D environment. Front Oncol 2022; 12:956940. [PMID: 36059712 PMCID: PMC9434107 DOI: 10.3389/fonc.2022.956940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
In vitro cell culture studies are common in the cancer research field, and reliable biomimetic 3D models are needed to ensure physiological relevance. In this manuscript, we hypothesized that decellularized xenograft tumors can serve as an optimal 3D substrate to generate a top-down approach for in vitro tumor modeling. Multiple tumor cell lines were xenografted and the formed solid tumors were recovered for their decellularization by several techniques and further characterization by histology and proteomics techniques. Selected decellularized tumor xenograft samples were seeded with the HCC1806 human triple-negative breast cancer (TNBC) basal-like subtype cell line, and cell behavior was compared among them and with other control 2D and 3D cell culture methods. A soft treatment using Freeze-EDTA-DNAse allows proper decellularization of xenografted tumor samples. Interestingly, proteomic data show that samples decellularized from TNBC basal-like subtype xenograft models had different extracellular matrix (ECM) compositions compared to the rest of the xenograft tumors tested. The in vitro recellularization of decellularized ECM (dECM) yields tumor-type–specific cell behavior in the TNBC context. Data show that dECM derived from xenograft tumors is a feasible substrate for reseeding purposes, thereby promoting tumor-type–specific cell behavior. These data serve as a proof-of-concept for further potential generation of patient-specific in vitro research models.
Collapse
Affiliation(s)
- Gaia Iazzolino
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Unai Mendibil
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
- TECNALIA, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Blanca Arnaiz
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Ane Ruiz-de-Angulo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Kepa B. Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Neda Khatami
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Beatriz Olalde
- TECNALIA, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Vanessa Gomez-Vallejo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Jordi Llop
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
| | - Ander Abarrategi
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- *Correspondence: Ander Abarrategi,
| |
Collapse
|
35
|
Ghanem NH, El-Badawy N, El Din SS, Hewedi I, Shash L. Transforming Growth Factor Beta1 Expression in Cancer- Associated Fibroblasts of Urinary Bladder Cancer: Crucial Applications and Deep Insights. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Urinary bladder carcinoma (UBC) is one of the most common malignancies in Egypt and all over the world. TGFB levels in plasma and urine were proved to connote predictive and prognostic attributes in UBC patients. Furthermore, Cancer associated fibroblasts (CAFs) are now recognized as a key player in carcinogenesis. Yet, TGFΒ1 expression in CAFs of UBC had not been elucidated. Moreover, TGFB1 targeted therapy is now emerging with potential benefits for TGFB1 expressing cancers.
Aim of the study: we dedicated this study to explore potential implications of TGFB1 immunohistochemical expression in CAFs of UBC by correlating it to relevant clinical and pathological data.
Material and methods: This retrospective study included 48 UBC specimens. Different tumor grades were presented in balanced groups. TGFB1 immunohistochemical expression was evaluated, categorized as low or high and compared in CAFs among different UBC grades, statistical analysis of the results was then followed.
Results: TGFB1 expression in CAFs was significantly different among tumor histologic types (P=0.01), high tumor grade (P=<0.01), presence of muscle invasion (P=<0.001), higher tumor stage (P=0.01), presence of preceding bilharziasis (P=0.003), and necrosis (P=0.03). There was a highly significant difference between TGFB1 expression in both tumor cells and CAFs (P=0.002). Intense CAFs TGFB1 staining was also strikingly observed along the muscle invading frontside UBC cells further emphasizing the pivotal role of CAFs expressing TGFB1 in invasion.
Conclusion: This study demonstrates significant predictive implications of TGFB1 in UBC, thus emphasizing its potential benefits in management and therapy.
Collapse
|
36
|
Mohammad Mirzaei N, Tatarova Z, Hao W, Changizi N, Asadpoure A, Zervantonakis IK, Hu Y, Chang YH, Shahriyari L. A PDE Model of Breast Tumor Progression in MMTV-PyMT Mice. J Pers Med 2022; 12:807. [PMID: 35629230 PMCID: PMC9145520 DOI: 10.3390/jpm12050807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
The evolution of breast tumors greatly depends on the interaction network among different cell types, including immune cells and cancer cells in the tumor. This study takes advantage of newly collected rich spatio-temporal mouse data to develop a data-driven mathematical model of breast tumors that considers cells' location and key interactions in the tumor. The results show that cancer cells have a minor presence in the area with the most overall immune cells, and the number of activated immune cells in the tumor is depleted over time when there is no influx of immune cells. Interestingly, in the case of the influx of immune cells, the highest concentrations of both T cells and cancer cells are in the boundary of the tumor, as we use the Robin boundary condition to model the influx of immune cells. In other words, the influx of immune cells causes a dominant outward advection for cancer cells. We also investigate the effect of cells' diffusion and immune cells' influx rates in the dynamics of cells in the tumor micro-environment. Sensitivity analyses indicate that cancer cells and adipocytes' diffusion rates are the most sensitive parameters, followed by influx and diffusion rates of cytotoxic T cells, implying that targeting them is a possible treatment strategy for breast cancer.
Collapse
Affiliation(s)
- Navid Mohammad Mirzaei
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (Y.H.)
| | - Zuzana Tatarova
- Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Wenrui Hao
- Department of Mathematics, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Navid Changizi
- Department of Civil and Environmental Engineering, University of Massachusetts, Dartmouth, MA 02747, USA; (N.C.); (A.A.)
| | - Alireza Asadpoure
- Department of Civil and Environmental Engineering, University of Massachusetts, Dartmouth, MA 02747, USA; (N.C.); (A.A.)
| | - Ioannis K. Zervantonakis
- Department of Bioengineering, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15219, USA;
| | - Yu Hu
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (Y.H.)
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA; (N.M.M.); (Y.H.)
| |
Collapse
|
37
|
Neutrophil infiltration combined with necrosis in the primary tumor is a useful prognostic indicator for three‑year disease‑free survival time in patients with colorectal cancer. Oncol Lett 2022; 23:199. [PMID: 35572490 PMCID: PMC9100607 DOI: 10.3892/ol.2022.13320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/11/2022] [Indexed: 11/30/2022] Open
Abstract
Histopathological evaluation plays a key role in the diagnosis of colorectal cancer (CRC). Tumor-related local inflammation is regarded as a novel prognostic parameter. Neutrophils constitute one of the main types of inflammatory cells. The aim of the present study was to evaluate the prognostic value of intratumoral tumor-associated neutrophils (intraTANs), stromal TANs (stromaTANs) and necrosis, as well as their combined parametric value in formalin-fixed paraffin-embedded tissue sections from patients with CRC. For this purpose, a retrospective study of 160 patients with CRC who underwent surgery was conducted. The association of intraTANs, stromaTANs, necrosis and their combined parametric value with the clinicopathological features of patients with CRC was examined. The Kaplan-Meier method and the log-rank test were used to compare survival curves. To identify independent prognostic factors, uni- and multivariate Cox proportional hazards regression models were used. StromaTANs were associated with lymph node metastasis (P=0.049) and tumor deposits (P=0.041). In addition, necrosis was found to be associated with venous (P=0.003), lymphatic (P=0.007) and perineural (P=0.015) invasion, as well as with lymph node metastasis (P=0.033), the number of invaded lymph nodes (P=0.012), and lymph node pouch invasion (P=0.043). Furthermore, necrosis was found to be associated with the white blood cell count (P=0.030), neutrophil count (P=0.011), the combined neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (NLR-PLR) (P=0.038), and the combined platelet and NLR (PLT-NLR status) (P=0.030), as well as with the serum carcinoembryonic antigen (CEA) levels following surgery (P=0.011) and the monocyte-to-lymphocyte ratio (P=0.023). The combined parametric value was found to be associated with pT stage (P=0.049), venous (P=0.034) and lymphatic (P=0.026) invasion, and with serum CEA levels prior to surgery (P=0.029). The analysis of the 3-year disease-free survival (DFS) time revealed that tumor growth [hazard ratio (HR), 2.070; 95% CI, 1.837-3.808; P=0.003] and the combined parametric value (intraTANs, stromaTANs and necrosis, HR, 1.577; 95% CI, 1.372-3.032; P=0.028) were independent factors for patients with CRC. Taken together, the findings of the present study demonstrated that the combined value of neutrophils and necrosis examined in the cancerous tissue may be used as a prognostic factor for the 3-year DFS time in patients with CRC.
Collapse
|
38
|
Xia Y, Ge M, Xia L, Shan G, Qian H. CTSV (cathepsin V) promotes bladder cancer progression by increasing NF-κB activity. Bioengineered 2022; 13:10180-10190. [PMID: 35443863 PMCID: PMC9162008 DOI: 10.1080/21655979.2022.2061278] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chronic inflammation is positively associated with the development of urinary bladder cancer. However, its detailed regulatory mechanism remains elusive. The quantitative real-time polymerase chain reaction was used to measure mRNA levels of relative genes. The protein levels were monitored by western blotting. Cell proliferation and viability were evaluated by the cell counting Kit 8 (CCK8) and colony formation assays, respectively. The dual-luciferase reporter assay was performed to assay the transcriptional activity. In vivo experiments were implemented in nude mice as well. The TCGA database analysis suggested that the aberrant expression of cathepsin V (CTSV) was related to a poor outcome in bladder cancer patients. CTSV boosted the inflammation reaction, which facilitated the development of bladder cancer. The overexpression of CTSV increased the proliferation and viability of bladder cancer cells. On the contrary, the deletion of CTSV significantly inhibited the proliferation and viability of bladder cancer cells. The tumor repression resulting from CTSV deficiency in vitro was also verified in vivo. Moreover, multiple cancer-associated luciferase screening showed that the overexpression of CTSV triggered the inflammatory signaling pathway, which could be restored by introducing the NF-κB inhibitor. CTSV is upregulated and promotes proliferation through the NF-κB pathway in bladder cancer and may be a potential target in inflammation-associated bladder cancer.
Collapse
Affiliation(s)
- Yue Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Minghuan Ge
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ling Xia
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guang Shan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huijun Qian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
39
|
Controlling Cancer Cell Death Types to Optimize Anti-Tumor Immunity. Biomedicines 2022; 10:biomedicines10050974. [PMID: 35625711 PMCID: PMC9138898 DOI: 10.3390/biomedicines10050974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/16/2022] [Accepted: 04/17/2022] [Indexed: 11/17/2022] Open
Abstract
Over several decades, cell biology research has characterized distinct forms of regulated cell death, identified master regulators such as nuclear factor kappa B (NFκB), and contributed to translating these findings in order to improve anti-cancer therapies. In the era of immunotherapy, however, the field warrants a new appraisal-the targeted induction of immunogenic cell death may offer personalized strategies to optimize anti-tumor immunity. Once again, the spotlight is on NFκB, which is not only a master regulator of cancer cell death, survival, and inflammation, but also of adaptive anti-tumor immune responses that are triggered by dying tumor cells.
Collapse
|
40
|
Buck E, Lee S, Gao Q, Tran SD, Tamimi F, Stone LS, Cerruti M. The Role of Surface Chemistry in the Osseointegration of PEEK Implants. ACS Biomater Sci Eng 2022; 8:1506-1521. [PMID: 35290033 DOI: 10.1021/acsbiomaterials.1c01434] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Poly(etheretherketone) (PEEK) implants suffer from poor osseointegration because of chronic inflammation. In this study, we hypothesized that adding NH2 and COOH groups to the surface of PEEK could modulate macrophage responses by altering protein adsorption and improve its osseointegration. NH2 and COOH-functionalized PEEK surfaces induced pro- and anti-inflammatory macrophage responses, respectively, and differences in protein adsorption patterns on these surfaces were related to the varied inflammatory responses. The macrophage responses to NH2 surfaces significantly reduced the osteogenic differentiation of mesenchymal stem cells (MSCs). MSCs cultured on NH2 surfaces differentiated less than those on COOH surfaces even though NH2 surfaces promoted the most mineralization in simulated body fluid solutions. After 14 days in rat tibia unicortical defects, the bone around NH2 surfaces had thinner trabeculae and higher specific bone surface than the bone around unmodified implants; surprisingly, the NH2 implants significantly increased bone-binding over the unmodified implants, while COOH implants only showed a trend for increasing bone-binding. Taken together, these results suggest that both mineral-binding and immune responses play a role in osseointegration, and PEEK implant integration may be improved with mixtures of these two functional groups to harness the ability to reduce inflammation and bind bone strongly.
Collapse
Affiliation(s)
- Emily Buck
- Mining and Materials Engineering, McGill University, 3610 University Street, Montreal, Quebec H3A 0C5, Canada
| | - Seunghwan Lee
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada.,Alan Edwards Center for Research on Pain, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec H3A 0G1, Canada
| | - Qiman Gao
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada
| | - Simon D Tran
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada
| | - Faleh Tamimi
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada
| | - Laura S Stone
- Faculty of Dentistry, McGill University, 2001 McGill College Avenue #500, Montreal, Quebec H3A 1G1, Canada.,Alan Edwards Center for Research on Pain, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec H3A 0G1, Canada
| | - Marta Cerruti
- Mining and Materials Engineering, McGill University, 3610 University Street, Montreal, Quebec H3A 0C5, Canada
| |
Collapse
|
41
|
Meng F, Zhang K, Yang C, Zhang K, Xu Q, Ren R, Zhou Y, Sun Y, Peng Y, Li Y, Guo H, Ren Y, Zhao Z. Prognostic Pathways Guide Drug Indications in Pan-Cancers. Front Oncol 2022; 12:849552. [PMID: 35372084 PMCID: PMC8964428 DOI: 10.3389/fonc.2022.849552] [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: 01/06/2022] [Accepted: 02/07/2022] [Indexed: 11/18/2022] Open
Abstract
Pathway-level analysis is a powerful approach enabling the interpretation of post-genomic data at a higher level than that of individual molecules. Molecular-targeted therapy focusing on cascade signaling pathways has become a new paradigm in anticancer therapy, instead of a single protein. However, the approaches to narrowing down the long list of biological pathways are limited. Here, we proposed a strategy for in silico Drug Prescription on biological pathways across pan-Cancers (CDP), by connecting drugs to candidate pathways. Applying on a list of 120 traditional Chinese medicines (TCM), we especially identified the “TCM–pathways–cancers” triplet and constructed it into a heterogeneous network across pan-cancers. Applying them into TCMs, the computational prescribing methods deepened the understanding of the efficacy of TCM at the molecular level. Further applying them into Western medicines, CDP could promote drug reposition avoiding time-consuming developments of new drugs.
Collapse
Affiliation(s)
- Fanlin Meng
- Marketing and Management Department, CapitalBio Technology, Beijing, China.,National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Kenan Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Changlin Yang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ke Zhang
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Quan Xu
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Ruifang Ren
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yiming Zhou
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yimin Sun
- Marketing and Management Department, CapitalBio Technology, Beijing, China.,National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yan Peng
- Marketing and Management Department, CapitalBio Technology, Beijing, China
| | - Yanze Li
- Marketing and Management Department, CapitalBio Technology, Beijing, China
| | - Hongyan Guo
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yonghong Ren
- Marketing and Management Department, CapitalBio Technology, Beijing, China
| | - Zheng Zhao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
42
|
Krstic A, Pavic A, Avdovic E, Markovic Z, Stevanovic M, Petrovic I. Coumarin-Palladium(II) Complex Acts as a Potent and Non-Toxic Anticancer Agent against Pancreatic Carcinoma Cells. Molecules 2022; 27:2115. [PMID: 35408514 PMCID: PMC9000835 DOI: 10.3390/molecules27072115] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/30/2022] Open
Abstract
Pancreatic carcinoma still represents one of the most lethal malignant diseases in the world although some progress has been made in treating the disease in the past decades. Current multi-agent treatment options have improved the overall survival of patients, however, more effective treatment strategies are still needed. In this paper we have characterized the anticancer potential of coumarin-palladium(II) complex against pancreatic carcinoma cells. Cells viability, colony formation and migratory potential of pancreatic carcinoma cells were assessed in vitro, followed by evaluation of apoptosis induction and in vivo testing on zebrafish. Presented results showed remarkable reduction in pancreatic carcinoma cells growth both in vitro and in vivo, being effective at micromolar concentrations (0.5 μM). Treatments induced apoptosis, increased BAX/BCL-2 ratio and suppressed the expression of SOX9 and SOX18, genes shown to be significantly up-regulated in pancreatic ductal adenocarcinoma. Importantly, treatments of the zebrafish-pancreatic adenocarcinoma xenografts resulted in significant reduction in tumor mass, without provoking any adverse toxic effects including hepatotoxicity. Presented results indicate the great potential of the tested compound and the perspective of its further development towards pancreatic cancer therapy.
Collapse
Affiliation(s)
- Aleksandra Krstic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
| | - Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
| | - Edina Avdovic
- Department of Science, Institute of Information Technologies, University of Kragujevac, Jovana Cvijica bb, 34000 Kragujevac, Serbia; (E.A.); (Z.M.)
| | - Zoran Markovic
- Department of Science, Institute of Information Technologies, University of Kragujevac, Jovana Cvijica bb, 34000 Kragujevac, Serbia; (E.A.); (Z.M.)
| | - Milena Stevanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
- Department of Chemical and Biological Sciences, Serbian Academy of Sciences and Arts, Kneza Mihaila 35, 11000 Belgrade, Serbia
| | - Isidora Petrovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
| |
Collapse
|
43
|
Tsilimigras DI, Ejaz A, Cloyd J, Guglielmi A, Aldrighetti L, Weiss M, Bauer TW, Alexandrescu S, Poultsides GA, Maithel SK, Marques HP, Martel G, Pulitano C, Shen F, Soubrane O, Koerkamp BG, Endo I, Pawlik TM. Tumor Necrosis Impacts Prognosis of Patients Undergoing Resection for T1 Intrahepatic Cholangiocarcinoma. Ann Surg Oncol 2022; 29:10.1245/s10434-022-11462-y. [PMID: 35298762 DOI: 10.1245/s10434-022-11462-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/05/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND The prognostic impact of tumor necrosis among patients undergoing resection for intrahepatic cholangiocarcinoma (ICC) remains ill-defined. METHODS Patients who underwent curative-intent resection for ICC between 2000 and 2017 were identified using a multi-institutional database. The association of pathologic tumor necrosis with overall survival (OS) and recurrence-free survival (RFS) was examined. RESULTS Among 757 patients who underwent resection for ICC, tumor necrosis was present in 384 (50.7%) patients (no necrosis: n = 373, 49.3%; <50% necrosis: n = 291, 38.4%; ≥50% necrosis: n = 93, 12.3%). Tumor necrosis was associated with worse OS (5-year OS: no necrosis 39.3% vs. <50% necrosis 34.7% and ≥50% necrosis 24.0%; p = 0.03) and RFS (5-year RFS: no necrosis 25.7% vs. <50% necrosis 13.9% and ≥50% necrosis 18.8%; p < 0.001). After stratifying by T stage, tumor necrosis was able to further stratify prognosis among patients with T1a ICC (5-year RFS: T1a and no necrosis 46.7% vs. T1a and necrosis 36.1%; p = 0.02), and T1b ICC (5-year RFS: T1b and no necrosis 31.1% vs. T1b and necrosis 11.2%; p = 0.006), but was not associated with outcomes among patients with more advanced T2-T3 disease. Patients with T1a ICC and tumor necrosis had similar 5-year RFS as individuals with T1b ICC and no tumor necrosis (36.1% vs. 31.1%; p = 0.66). CONCLUSION Tumor necrosis was associated with worse prognosis among patients with T1 ICC. Tumor necrosis for T1 ICC should be considered as an important factor to further stratify outcomes of patients with early T-stage ICC.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Aslam Ejaz
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Jordan Cloyd
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | | | | | - Matthew Weiss
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - Hugo P Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | | | - Carlo Pulitano
- Department of Surgery, Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia
| | - Feng Shen
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Olivier Soubrane
- Department of Hepatobiliopancreatic Surgery and Liver Transplantation, AP-HP, Beaujon Hospital, Clichy, France
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, 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, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
44
|
In Vitro and In Vivo Evaluation of a Cyclic LyP-1-Modified Nanosystem for Targeted Endostatin Delivery in a KYSE-30 Cell Xenograft Athymic Nude Mice Model. Pharmaceuticals (Basel) 2022; 15:ph15030353. [PMID: 35337150 PMCID: PMC8955112 DOI: 10.3390/ph15030353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/01/2023] Open
Abstract
This work investigated the use of LyP-1 as a homing peptide for p32 receptor targeting on the surface of an endostatin (ENT)-loaded chitosan-grafted nanosystem intended for intracellular delivery of ENT and mitochondrial targeting in a squamous cell carcinoma (SCC) cell line (KYSE-30) model. The angiogenic factors for VEGF-C and MMP2 were assessed with in vivo evaluation of the nanosystem upon ENT release and tumor necrosis in nude mice with a KYSE-30 cell xenograft. The LyP-1-modified nanosystem revealed a three-fold decrease in proliferation at 1000 µg/mL compared with the control and facilitated receptor-mediated cellular uptake and internalization. In addition, targeting of the Lyp-1-functionalized nanosystem to mitochondrial and nuclear proteins in vitro and in vivo was achieved. Up to 60% inhibition of KYSE-30 cell migration was observed and the expressions of VEGF-C and MMP-2 as angiogenic markers were reduced 3- and 2-fold, respectively. A marked reduction in tumor mass was recorded (43.25%) with the control, a 41.36% decrease with the nanoparticles and a 61.01% reduction with the LyP-1-modified nanosystem following treatment in mice. The LyP-1-functionalized nanosystem targeted tumor lymphatics, instigated nuclear rupture and mitochondrial distortion, and decreased cell proliferation and migration with inhibition of VEGF-C and MMP2 expression.
Collapse
|
45
|
Davis L, Recktenwald M, Hutt E, Fuller S, Briggs M, Goel A, Daringer N. Targeting HIF-2α in the Tumor Microenvironment: Redefining the Role of HIF-2α for Solid Cancer Therapy. Cancers (Basel) 2022; 14:1259. [PMID: 35267567 PMCID: PMC8909461 DOI: 10.3390/cancers14051259] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 02/26/2022] [Indexed: 02/06/2023] Open
Abstract
Inadequate oxygen supply, or hypoxia, is characteristic of the tumor microenvironment and correlates with poor prognosis and therapeutic resistance. Hypoxia leads to the activation of the hypoxia-inducible factor (HIF) signaling pathway and stabilization of the HIF-α subunit, driving tumor progression. The homologous alpha subunits, HIF-1α and HIF-2α, are responsible for mediating the transcription of a multitude of critical proteins that control proliferation, angiogenic signaling, metastasis, and other oncogenic factors, both differentially and sequentially regulating the hypoxic response. Post-translational modifications of HIF play a central role in its behavior as a mediator of transcription, as well as the temporal transition from HIF-1α to HIF-2α that occurs in response to chronic hypoxia. While it is evident that HIF-α is highly dynamic, HIF-2α remains vastly under-considered. HIF-2α can intensify the behaviors of the most aggressive tumors by adapting the cell to oxidative stress, thereby promoting metastasis, tissue remodeling, angiogenesis, and upregulating cancer stem cell factors. The structure, function, hypoxic response, spatiotemporal dynamics, and roles in the progression and persistence of cancer of this HIF-2α molecule and its EPAS1 gene are highlighted in this review, alongside a discussion of current therapeutics and future directions.
Collapse
Affiliation(s)
- Leah Davis
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Matthias Recktenwald
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Evan Hutt
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Schuyler Fuller
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Madison Briggs
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Arnav Goel
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Nichole Daringer
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| |
Collapse
|
46
|
Stroet MCM, de Blois E, de Jong M, Seimbille Y, Mezzanotte L, Löwik CWGM, Panth KM. Improved Multimodal Tumor Necrosis Imaging with IRDye800CW-DOTA Conjugated to an Albumin-Binding Domain. Cancers (Basel) 2022; 14:cancers14040861. [PMID: 35205609 PMCID: PMC8870237 DOI: 10.3390/cancers14040861] [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: 12/10/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Anti-tumor treatment efficacy is determined by tumor shrinkage, which takes valuable time to become apparent and poses a risk of unnecessary treatment with severe side effects. Therefore, there is an unmet need for more reliable and specific methods to monitor treatment efficacy. We explore radiolabeled cyanines for imaging tumor necrosis as a unique marker for therapy efficacy. Moreover, spontaneous tumor necrosis is a hallmark for aggressively growing tumor types with poor prognosis. We improved the binding properties of a previously reported necrosis-avid contrast agent (NACA) and successfully detected spontaneous and therapy-induced tumor necrosis in mice using radioactivity and fluorescence imaging modalities. This NACA may pave the way to in vivo detection of tumor necrosis for early-stage determination of tumor aggressiveness and therapy efficacy. Abstract Purpose: To assess our improved NACA for the detection of tumor necrosis. Methods: We increased the blood circulation time of our NACA by adding an albumin-binding domain to the molecular structure. We tested the necrosis avidity on dead or alive cultured cells and performed SPECT and fluorescence imaging of both spontaneous and treatment-induced necrosis in murine breast cancer models. We simultaneously recorded [18F]FDG-PET and bioluminescence images for complementary detection of tumor viability. Results: We generated two albumin-binding IRDye800CW derivatives which were labeled with indium-111 with high radiochemical purity. Surprisingly, both albumin-binding NACAs had >10x higher in vitro binding towards dead cells. We selected [111In]3 for in vivo experiments which showed higher dead cell binding in vitro and in vivo stability. The doxorubicin-treated tumors showed increased [111In]3-uptake (1.74 ± 0.08%ID/g after saline treatment, 2.25 ± 0.16%ID/g after doxorubicin treatment, p = 0.044) and decreased [18F]FDG-uptake (3.02 ± 0.51%ID/g after saline treatment, 1.79 ± 0.11%ID/g after doxorubicin treatment, p = 0.040), indicating therapy efficacy. Moreover, we detected increased [111In]3-uptake and tumor necrosis in more rapidly growing EMT6 tumors. Conclusions: Our albumin-binding NACA based on IRDye800CW facilitates tumor-necrosis imaging for assessment of therapy efficacy and aggressiveness in solid tumors using both fluorescence and SPECT imaging.
Collapse
Affiliation(s)
- Marcus C. M. Stroet
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Erik de Blois
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
| | - Marion de Jong
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
| | - Yann Seimbille
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
| | - Laura Mezzanotte
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Clemens W. G. M. Löwik
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- CHUV Department of Oncology, University of Lausanne, CH-1066 Lausanne, Switzerland
- Correspondence: (C.W.G.M.L.); (K.M.P.)
| | - Kranthi M. Panth
- Erasmus MC, Department of Radiology & Nuclear Medicine, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (M.C.M.S.); (E.d.B.); (Y.S.); (L.M.)
- Erasmus MC, Department of Molecular Genetics, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Correspondence: (C.W.G.M.L.); (K.M.P.)
| |
Collapse
|
47
|
Radiomic Phenotypes for Improving Early Prediction of Survival in Stage III Non-Small Cell Lung Cancer Adenocarcinoma after Chemoradiation. Cancers (Basel) 2022; 14:cancers14030700. [PMID: 35158971 PMCID: PMC8833400 DOI: 10.3390/cancers14030700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
We evaluate radiomic phenotypes derived from CT scans as early predictors of overall survival (OS) after chemoradiation in stage III primary lung adenocarcinoma. We retrospectively analyzed 110 thoracic CT scans acquired between April 2012-October 2018. Patients received a median radiation dose of 66.6 Gy at 1.8 Gy/fraction delivered with proton (55.5%) and photon (44.5%) beam treatment, as well as concurrent chemotherapy (89%) with carboplatin-based (55.5%) and cisplatin-based (36.4%) doublets. A total of 56 death events were recorded. Using manual tumor segmentations, 107 radiomic features were extracted. Feature harmonization using ComBat was performed to mitigate image heterogeneity due to the presence or lack of intravenous contrast material and variability in CT scanner vendors. A binary radiomic phenotype to predict OS was derived through the unsupervised hierarchical clustering of the first principal components explaining 85% of the variance of the radiomic features. C-scores and likelihood ratio tests (LRT) were used to compare the performance of a baseline Cox model based on ECOG status and age, with a model integrating the radiomic phenotype with such clinical predictors. The model integrating the radiomic phenotype (C-score = 0.69, 95% CI = (0.62, 0.77)) significantly improved (p<0.005) upon the baseline model (C-score = 0.65, CI = (0.57, 0.73)). Our results suggest that harmonized radiomic phenotypes can significantly improve OS prediction in stage III NSCLC after chemoradiation.
Collapse
|
48
|
Salinas-Vera YM, Gallardo-Rincón D, Ruíz-García E, Silva-Cázares MB, de la Peña-Cruz CS, López-Camarillo C. The Role of Hypoxia in Endometrial Cancer. Curr Pharm Biotechnol 2022; 23:221-234. [PMID: 33655827 DOI: 10.2174/1389201022666210224130022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 12/24/2022]
Abstract
Endometrial cancer represents the most frequent neoplasia from the corpus uteri and comprises the 14th leading cause of death in women worldwide. Risk factors that contribute to the disease include early menarche, late menopause, nulliparity, and menopausal hormone use, as well as hypertension and obesity comorbidities. The clinical effectiveness of chemotherapy is variable, suggesting that novel molecular targeted therapies against specific cellular processes associated with the maintenance of cancer cell survival and therapy resistance ameliorate the rates of success in endometrial cancer treatment. In the course of tumor growth, cancer cells must adapt to decreased oxygen availability in the microenvironment by upregulation of hypoxia-inducible factors, which orchestrate the activation of a transcriptional program leading to cell survival. During this adaptative process, the hypoxic cancer cells may acquire invasive and metastatic properties as well as increased cell proliferation and resistance to chemotherapy, enhanced angiogenesis, vasculogenic mimicry, and maintenance of cancer cell stemness, which contribute to more aggressive cancer phenotypes. Several studies have shown that hypoxia-inducible factor 1 alpha (HIF-1α) protein is aberrantly overexpressed in many solid tumors of the breast, prostate, ovarian, bladder, colon, brain, and pancreas. Thus, it has been considered an important therapeutic target. Here, we reviewed the current knowledge of the relevant roles of cellular hypoxia mechanisms and HIF-1α functions in diverse processes associated with endometrial cancer progression. In addition, we also summarize the role of microRNAs in the posttranscriptional regulation of protein-encoding genes involved in the hypoxia response in endometrial cancer. Finally, we pointed out the need for urgent targeted therapies to impair the cellular processes activated by hypoxia in the tumor microenvironment.
Collapse
Affiliation(s)
| | - Dolores Gallardo-Rincón
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastrointestinales, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Erika Ruíz-García
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastrointestinales, Instituto Nacional de Cancerología, Ciudad de México, Mexico
| | - Macrina B Silva-Cázares
- Doctorado Institucional en Ingeniería y Ciencia de Materiales, Universidad Autónoma de San Luis Potosí, México
| | | | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México
| |
Collapse
|
49
|
Recapitulating the Angiogenic Switch in a Hydrogel-Based 3D In Vitro Tumor-Stroma Model. Bioengineering (Basel) 2021; 8:bioengineering8110186. [PMID: 34821752 PMCID: PMC8614676 DOI: 10.3390/bioengineering8110186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
To ensure nutrient and oxygen supply, tumors beyond a size of 1–2 mm3 need a connection to the vascular system. Thus, tumor cells modify physiological tissue homeostasis by secreting inflammatory and angiogenic cytokines. This leads to the activation of the tumor microenvironment and the turning of the angiogenic switch, resulting in tumor vascularization and growth. To inhibit tumor growth by developing efficient anti-angiogenic therapies, an in depth understanding of the molecular mechanism initiating angiogenesis is essential. Yet so far, predominantly 2D cell cultures or animal models have been used to clarify the interactions within the tumor stroma, resulting in poor transferability of the data obtained to the in vivo situation. Consequently, there is an abundant need for complex, humanized, 3D models in vitro. We established a dextran-hydrogel-based 3D organotypic in vitro model containing microtumor spheroids, macrophages, neutrophils, fibroblasts and endothelial cells, allowing for the analysis of tumor–stroma interactions in a controlled and modifiable environment. During the cultivation period of 21 days, the microtumor spheroids in the model grew in size and endothelial cells formed elongated tubular structures resembling capillary vessels, that appeared to extend towards the tumor spheroids. The tubular structures exhibited complex bifurcations and expanded without adding external angiogenic factors such as VEGF to the culture. To allow high-throughput screening of therapeutic candidates, the 3D cell culture model was successfully miniaturized to a 96-well format, while still maintaining the same level of tumor spheroid growth and vascular sprouting. The quantification of VEGF in the conditioned medium of these cultures showed a continuous increase during the cultivation period, suggesting the contribution of endogenous VEGF to the induction of the angiogenic switch and vascular sprouting. Thus, this model is highly suitable as a testing platform for novel anticancer therapeutics targeting the tumor as well as the vascular compartment.
Collapse
|
50
|
Muñoz R, Girotti A, Hileeto D, Arias FJ. Metronomic Anti-Cancer Therapy: A Multimodal Therapy Governed by the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13215414. [PMID: 34771577 PMCID: PMC8582362 DOI: 10.3390/cancers13215414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Metronomic chemotherapy with different mechanisms of action against cancer cells and their microenvironment represents an exceptional holistic cancer treatment. Each type of tumor has its own characteristics, including each individual tumor in each patient. Understanding the complexity of the dynamic interactions that take place between tumor and stromal cells and the microenvironment in tumor progression and metastases, as well as the response of the host and the tumor itself to anticancer therapy, will allow therapeutic actions with long-lasting effects to be implemented using metronomic regimens. This study aims to highlight the complexity of cellular interactions in the tumor microenvironment and summarize some of the preclinical and clinical results that explain the multimodality of metronomic therapy, which, together with its low toxicity, supports an inhibitory effect on the primary tumor and metastases. We also highlight the possible use of nano-therapeutic agents as good partners for metronomic chemotherapy. Abstract The concept of cancer as a systemic disease, and the therapeutic implications of this, has gained special relevance. This concept encompasses the interactions between tumor and stromal cells and their microenvironment in the complex setting of primary tumors and metastases. These factors determine cellular co-evolution in time and space, contribute to tumor progression, and could counteract therapeutic effects. Additionally, cancer therapies can induce cellular and molecular responses in the tumor and host that allow them to escape therapy and promote tumor progression. In this study, we describe the vascular network, tumor-infiltrated immune cells, and cancer-associated fibroblasts as sources of heterogeneity and plasticity in the tumor microenvironment, and their influence on cancer progression. We also discuss tumor and host responses to the chemotherapy regimen, at the maximum tolerated dose, mainly targeting cancer cells, and a multimodal metronomic chemotherapy approach targeting both cancer cells and their microenvironment. In a combination therapy context, metronomic chemotherapy exhibits antimetastatic efficacy with low toxicity but is not exempt from resistance mechanisms. As such, a better understanding of the interactions between the components of the tumor microenvironment could improve the selection of drug combinations and schedules, as well as the use of nano-therapeutic agents against certain malignancies.
Collapse
Affiliation(s)
- Raquel Muñoz
- Department of Biochemistry, Physiology and Molecular Biology, University of Valladolid, Paseo de Belén, 47011 Valladolid, Spain
- Smart Biodevices for NanoMed Group, University of Valladolid, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
- Correspondence:
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
| | - Denise Hileeto
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON N2L 361, Canada;
| | - Francisco Javier Arias
- Smart Biodevices for NanoMed Group, University of Valladolid, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
| |
Collapse
|