1
|
Leng Y, Kan A, Wang X, Li X, Xiao X, Wang Y, Liu L, Gong L. Contrast-enhanced CT radiomics for preoperative prediction of stage in epithelial ovarian cancer: a multicenter study. BMC Cancer 2024; 24:307. [PMID: 38448945 PMCID: PMC10916071 DOI: 10.1186/s12885-024-12037-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Preoperative prediction of International Federation of Gynecology and Obstetrics (FIGO) stage in patients with epithelial ovarian cancer (EOC) is crucial for determining appropriate treatment strategy. This study aimed to explore the value of contrast-enhanced CT (CECT) radiomics in predicting preoperative FIGO staging of EOC, and to validate the stability of the model through an independent external dataset. METHODS A total of 201 EOC patients from three centers, divided into a training cohort (n = 106), internal (n = 46) and external (n = 49) validation cohorts. The least absolute shrinkage and selection operator (LASSO) regression algorithm was used for screening radiomics features. Five machine learning algorithms, namely logistic regression, support vector machine, random forest, light gradient boosting machine (LightGBM), and decision tree, were utilized in developing the radiomics model. The optimal performing algorithm was selected to establish the radiomics model, clinical model, and the combined model. The diagnostic performances of the models were evaluated through receiver operating characteristic analysis, and the comparison of the area under curves (AUCs) were conducted using the Delong test or F-test. RESULTS Seven optimal radiomics features were retained by the LASSO algorithm. The five radiomics models demonstrate that the LightGBM model exhibits notable prediction efficiency and robustness, as evidenced by AUCs of 0.83 in the training cohort, 0.80 in the internal validation cohort, and 0.68 in the external validation cohort. The multivariate logistic regression analysis indicated that carcinoma antigen 125 and tumor location were identified as independent predictors for the FIGO staging of EOC. The combined model exhibited best diagnostic efficiency, with AUCs of 0.95 in the training cohort, 0.83 in the internal validation cohort, and 0.79 in the external validation cohort. The F-test indicated that the combined model exhibited a significantly superior AUC value compared to the radiomics model in the training cohort (P < 0.001). CONCLUSIONS The combined model integrating clinical characteristics and radiomics features shows potential as a non-invasive adjunctive diagnostic modality for preoperative evaluation of the FIGO staging status of EOC, thereby facilitating clinical decision-making and enhancing patient outcomes.
Collapse
Affiliation(s)
- Yinping Leng
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Minde Road No. 1, 330006, Nanchang, Jiangxi Province, China
| | - Ao Kan
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Minde Road No. 1, 330006, Nanchang, Jiangxi Province, China
| | - Xiwen Wang
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Minde Road No. 1, 330006, Nanchang, Jiangxi Province, China
| | - Xiaofen Li
- Department of Radiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| | - Xuan Xiao
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Minde Road No. 1, 330006, Nanchang, Jiangxi Province, China
| | - Yu Wang
- Clinical and Technical Support, Philips Healthcare, Shanghai, China
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China.
| | - Lianggeng Gong
- Department of Radiology, the Second Affiliated Hospital of Nanchang University, Minde Road No. 1, 330006, Nanchang, Jiangxi Province, China.
| |
Collapse
|
2
|
Antil N, Wang H, Kaffas AE, Desser TS, Folkins A, Longacre T, Berek J, Lutz AM. In Vivo Ultrasound Molecular Imaging in the Evaluation of Complex Ovarian Masses: A Practical Guide to Correlation with Ex Vivo Immunohistochemistry. Adv Biol (Weinh) 2023; 7:e2300091. [PMID: 37403275 DOI: 10.1002/adbi.202300091] [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: 02/24/2023] [Revised: 04/22/2023] [Indexed: 07/06/2023]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related deaths in women and the most lethal gynecologic cancer. It is curable when discovered at an early stage, but usually remains asymptomatic until advanced stages. It is crucial to diagnose the disease before it metastasizes to distant organs for optimal patient management. Conventional transvaginal ultrasound imaging offers limited sensitivity and specificity in the ovarian cancer detection. With molecularly targeted ligands addressing targets, such as kinase insert domain receptor (KDR), attached to contrast microbubbles, ultrasound molecular imaging (USMI) can be used to detect, characterize and monitor ovarian cancer at a molecular level. In this article, the authors propose a standardized protocol is proposed for the accurate correlation between in- vivo transvaginal KDR-targeted USMI and ex vivo histology and immunohistochemistry in clinical translational studies. The detailed procedures of in vivo USMI and ex vivo immunohistochemistry are described for four molecular markers, CD31 and KDR with a focus on how to enable the accurate correlation between in vivo imaging findings and ex vivo expression of the molecular markers, even if not the entire tumor could can be imaged by USMI, which is not an uncommon scenario in clinical translational studies. This work aims to enhance the workflow and the accuracy of characterization of ovarian masses on transvaginal USMI using histology and immunohistochemistry as reference standards, which involves sonographers, radiologists, surgeons, and pathologists in a highly collaborative research effort of USMI in cancer.
Collapse
Affiliation(s)
- Neha Antil
- Department of Radiology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Huaijun Wang
- Department of Radiology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Ahmed El Kaffas
- Department of Radiology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Terry S Desser
- Department of Radiology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Ann Folkins
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Teri Longacre
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Jonathan Berek
- Stanford Women's Cancer Center, Stanford Cancer Institute, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| | - Amelie M Lutz
- Department of Radiology, Stanford University, School of Medicine, Stanford, CA, 94304, USA
| |
Collapse
|
3
|
Luo J, Zhang Y, Zheng T, Jing Y, Cao R, Wu M, Fan D, Tao Y, Zhao M. Application of long non-coding RNA RBAT1 in improving diagnosis and prognosis of ovarian carcinoma. Anticancer Drugs 2023; 34:9-14. [PMID: 36206099 PMCID: PMC9760461 DOI: 10.1097/cad.0000000000001315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022]
Abstract
Tumorigenesis of bladder cancer and retinoblastoma is correlated with long non-coding RNA (lncRNA) RBAT1. However, the role of RBAT1 in ovarian carcinoma (OC) is unclear. Thus, the study explored the role of RBAT1 in OC. This research enrolled patients with OC ( n = 68), irritable bowel disease (IBD, n = 68, females), digestive tract inflammation (DTI, n = 68, females), urinary tract infection (UTI, n = 68, females), endometriosis (EM, n = 68, females), and healthy controls (HCs, n = 68) to collect plasma sampled. OC and paired non-tumor tissues were collected from patients with OC. RBAT1 accumulation in all samples was analyzed using RT-qPCR. The role of plasma RBAT1 in OC diagnosis was examined using the ROC curves with OC patients as the true positive cases and other patients and HCs as the true negative cases. The role of RBAT1 in predicting the survival of OC patients was analyzed using the survival curve study. RBAT1 was overexpressed in both OC plasma and tissues. Plasma RBAT1 levels were correlated with RBAT1 levels in OC tissues but not in non-tumor tissues. Plasma RBAT1 could distinguish OC patients from other patients and HCs. Patients with high plasma RBAT1 levels had a shorter survival. RBAT1 is overexpressed in OC and might be applied to improve the diagnosis and prognosis of OC.
Collapse
Affiliation(s)
- Jie Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Yuqing Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Ting Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Yongping Jing
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Rongyu Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Minmin Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Die Fan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Ying Tao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| | - Mandan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou City, Guangdong Province, China
| |
Collapse
|
4
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [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: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
5
|
Giusti I, Di Francesco M, Poppa G, Esposito L, D’Ascenzo S, Dolo V. Tumor-Derived Extracellular Vesicles Activate Normal Human Fibroblasts to a Cancer-Associated Fibroblast-Like Phenotype, Sustaining a Pro-Tumorigenic Microenvironment. Front Oncol 2022; 12:839880. [PMID: 35280782 PMCID: PMC8905682 DOI: 10.3389/fonc.2022.839880] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Fibroblasts in the tumor microenvironment have been proven to actively participate in tumor progression; they can be "educated" by cancer cells acquiring an activated state and, as such, are identified as cancer-associated fibroblasts (CAFs); CAFs, in turn, remodel tumor stroma to be more advantageous for cancer progression by modulating several processes, including angiogenesis, immunosuppression, and drug access, presumably driving the chemoresistance. That is why they are believed to hamper the response to clinical therapeutic options. The communication between cancer cells and fibroblasts can be mediated by extracellular vesicles (EVs), composed of both exosomes (EXOs) and microvesicles (MVs). To verify the role of different subpopulations of EVs in this cross-talk, a nearly pure subpopulation of EXO-like EVs and the second one of mixed EXO- and MV-like EVs were isolated from ovarian cancer cells and administered to fibroblasts. It turned out that EVs can activate fibroblasts to a CAF-like state, supporting their proliferation, motility, invasiveness, and enzyme expression; EXO-like EV subpopulation seems to be more efficient in some of those processes, suggesting different roles for different EV subpopulations. Moreover, the secretome of these "activated" fibroblasts, composed of both soluble and EV-associated molecules, was, in turn, able to modulate the response of bystander cells (fibroblasts, tumor, and endothelial cells), supporting the idea that EVs sustain the mutual cross-talk between tumor cells and CAFs.
Collapse
Affiliation(s)
| | | | | | | | | | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
6
|
Chen F, Sun C, Yue Z, Zhang Y, Xu W, Shabbir S, Zou L, Lu W, Wang W, Xie Z, Zhou L, Lu Y, Yu J. Screening ovarian cancers with Raman spectroscopy of blood plasma coupled with machine learning data processing. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 265:120355. [PMID: 34530200 DOI: 10.1016/j.saa.2021.120355] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 06/13/2023]
Abstract
The mortality of ovarian cancer is closely related to its poor rate of early detection. In the search of an efficient diagnosis method, Raman spectroscopy of blood features as a promising technique allowing simple, rapid, minimally-invasive and cost-effective detection of cancers, in particular ovarian cancer. Although Raman spectroscopy has been demonstrated to be effective to detect ovarian cancers with respect to normal controls, a binary classification remains idealized with respect to the real clinical practice. This work considered a population of 95 woman patients initially suspected of an ovarian cancer and finally fixed with a cancer or a cyst. Additionally, 79 normal controls completed the ensemble of samples. Such sample collection proposed us a study case where a ternary classification should be realized with Raman spectroscopy of the collected blood samples coupled with suitable spectroscopic data treatment algorithms. In the medical as well as data points of view, the appearance of the cyst case considerably reduces the distances among the different populations and makes their distinction much more difficult, since the intermediate cyst case can share the specific features of the both cancer and normal cases. After a proper spectrum pretreatment, we first demonstrated the evidence of different behaviors among the Raman spectra of the 3 types of samples. Such difference was further visualized in a high dimensional space, where the data points of the cancer and the normal cases are separately clustered, whereas the data of the cyst case were scattered into the areas respectively occupied by the cancer and normal cases. We finally developed and tested an ensemble of models for a ternary classification with 2 consequent steps of binary classifications, based on machine learning algorithms, allowing identification with sensitivity and specificity of 81.0% and 97.3% for cancer samples, 63.6% and 91.5% for cyst samples, 100% and 90.6% for normal samples.
Collapse
Affiliation(s)
- Fengye Chen
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chen Sun
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zengqi Yue
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqing Zhang
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weijie Xu
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sahar Shabbir
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Long Zou
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weiguo Lu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women's Reproductive Health Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310011, China; Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310011, China
| | - Wei Wang
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Zhenwei Xie
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women's Reproductive Health Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310011, China; Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310011, China
| | - Lanyun Zhou
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women's Reproductive Health Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310011, China; Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310011, China
| | - Yan Lu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women's Reproductive Health Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310011, China; Department of Gynecologic Oncology, Women's Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310011, China.
| | - Jin Yu
- School of Physics and Astronomy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
7
|
Xun L, Zhai L, Xu H. Comparison of conventional, doppler and contrast-enhanced ultrasonography in differential diagnosis of ovarian masses: a systematic review and meta-analysis. BMJ Open 2021; 11:e052830. [PMID: 34952878 PMCID: PMC8710872 DOI: 10.1136/bmjopen-2021-052830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES To assess the value of conventional, Doppler and contrast-enhanced ultrasonography (CEUS) (conventional ultrasonography (US), Doppler US and CEUS) for diagnosing ovarian cancer. DESIGN Systematic review and meta-analysis. DATA SOURCES PubMed, Embase and the Cochrane Library were conducted for studies published until October 2021. ELIGIBILITY CRITERIA Studies assessed the diagnostic value of conventional US, Doppler US or CEUS for detecting ovarian cancer, with no restrictions placed on published language and status. DATA EXTRACTION AND SYNTHESIS The study selection and data extraction were performed by two independent authors. The sensitivity, specificity, positive and negative likelihood ratio (PLR and NLR), diagnostic OR (DOR) and area under the receiver operating characteristic curve (AUC) were pooled using the bivariate generalised linear mixed model and random effects model. RESULTS The meta-analysis included 72 studies and involved 9296 women who presented with ovarian masses. The pooled sensitivity, specificity, PLR, NLR, DOR and AUC for conventional US were 0.91 (95% CI: 0.87 to 0.94) and 0.87 (95% CI: 0.82 to 0.91), 6.87 (95% CI: 4.98 to 9.49) and 0.10 (95% CI: 0.07 to 0.15), 57.52 (95% CI: 36.64 to 90.28) and 0.95 (95% CI: 0.93 to 0.97), respectively. The sensitivity, specificity, PLR, NLR, DOR and AUC for Doppler US were 0.93 (95% CI: 0.91 to 0.95) and 0.85 (95% CI: 0.80 to 0.89), 6.10 (95% CI: 4.59 to 8.11) and 0.08 (95% CI: 0.06 to 0.11), 61.76 (95% CI: 39.99 to 95.37) and 0.96 (95% CI: 0.94 to 0.97), respectively. The pooled sensitivity, specificity, PLR, NLR, DOR and AUC for CEUS were 0.97 (95% CI: 0.92 to 0.99) and 0.92 (95% CI: 0.85 to 0.95), 11.47 (95% CI: 6.52 to 20.17) and 0.03 (95% CI: 0.01 to 0.09), 152.11 (95% CI: 77.77 to 297.51) and 0.99 (95% CI: 0.97 to 0.99), respectively. Moreover, the AUC values for conventional US (p=0.002) and Doppler US (p=0.005) were inferior to those of CEUS. CONCLUSIONS Conventional US, Doppler US and CEUS have a relatively high differential diagnostic value for differentiating between benign and malignant ovarian masses. The diagnostic performance of CEUS was superior to that of conventional US and Doppler US.
Collapse
Affiliation(s)
- Lizhang Xun
- Medical Examination Center, Huaian City Second People's Hospital, Huaian, Jiangsu, China
| | - Lamei Zhai
- Department of Radiology, Huaian City Second People's Hospital, Huaian, Jiangsu, China
| | - Hui Xu
- Medical Examination Center, Huaian City Second People's Hospital, Huaian, Jiangsu, China
| |
Collapse
|
8
|
Zhang G, Zheng D, Chen X, Li L, Yu J. miR-152-mediated MKK7 downregulation is attenuated by MYCNOS in ovarian adenocarcinoma. Oncol Lett 2021; 22:841. [PMID: 34733360 PMCID: PMC8561216 DOI: 10.3892/ol.2021.13102] [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: 12/30/2020] [Accepted: 09/07/2021] [Indexed: 12/09/2022] Open
Abstract
MYCN opposite strand (MYCNOS) acts as an oncogenic long non-coding RNA in liver cancer. However, its role in other cancer types is unknown. The aim of the present study was to investigate the function of MYCNOS in ovarian adenocarcinoma (OA). MYCNOS expression in OA was determined using reverse transcription-quantitative PCR (RT-qPCR), and its prognostic value for OA was evaluated in a 5-year follow-up study. The predicted interaction between MYCNOS and microRNA (miR)-152 was confirmed using a dual luciferase reporter assay. The association between MYCNOS and miR-152 was also analyzed in overexpression experiments. The effects of MYCNOS and miR-152 on mitogen-activated protein kinase kinase 7 (MKK7) expression were explored using RT-qPCR and western blotting. Cell proliferation was analyzed using a Cell Counting Kit-8 assay. MYCNOS expression was found to be upregulated in OA and predicted poor survival. In addition, MYCNOS was predicted to interact with miR-152, and a dual luciferase assay confirmed this interaction. However, MYCNOS and miR-152 overexpression did not affect their mutual expression levels. MYCNOS overexpression upregulated MKK7, a target of miR-152. Cell proliferation increased following simultaneous MYCNOS and MKK7 overexpression, but was reduced following miR-152 overexpression. Moreover, MYCNOS overexpression attenuated the effects of miR-152 overexpression. In conclusion, MYCNOS may act by sponging miR-152 to upregulate MKK7 expression in OA, thereby promoting cell proliferation.
Collapse
Affiliation(s)
- Guifang Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Dan Zheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Xiaoqing Chen
- Day Operation Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330003, P.R. China
| | - Li Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Jingrong Yu
- Department of Oncology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330003, P.R. China
| |
Collapse
|
9
|
Massollo M, Fiz F, Bottoni G, Ugolini M, Paparo F, Puppo C, Provinciali N, Iacozzi M, Altrinetti V, Cistaro A, Cabria M, DeCensi A, Treglia G, Piccardo A. To Enhance or Not to Enhance? The Role of Contrast Medium 18F-FDG PET/CT in Recurrent Ovarian Carcinomas. ACTA ACUST UNITED AC 2021; 57:medicina57060561. [PMID: 34206116 PMCID: PMC8229308 DOI: 10.3390/medicina57060561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/11/2022]
Abstract
Background and Objectives: 18F-fluorodeoxyglucose (FDG) positron emission tomography/X-ray computed tomography (PET/CT) represents the mainstay diagnostic procedure for suspected ovarian cancer (OC) recurrence. PET/CT can be integrated with contrast medium and in various diagnostic settings; however, the effective benefit of this procedure is still debated. We aimed to compare the diagnostic capabilities of low-dose and contrast-enhanced PET/CT (PET/ldCT and PET/ceCT) in patients with suspected ovarian cancer relapse. Materials and Methods: 122 OC patients underwent both PET/ldCT and PET/ceCT. Two groups of nuclear medicine physicians and radiologists scored the findings as positive or negative. Clinical/radiological follow-up was used as ground truth. Sensitivity, specificity, negative/positive predictive value, and accuracy were calculated at the patient and the lesion level. Results: A total of 455 and 474 lesions were identified at PET/ldCT and PET/ceCT, respectively. At the lesion level, sensitivity, specificity, positive predictive value, negative predictive value, and accuracy were not significantly different between PET/ldCT and PET/ceCT (98%, 93.3%, 97.4%, 94.9%, and 96.9% for PET/ldCT; 99%, 95.5%, 98.3%, 97%, and 98% for PET/ceCT, p = ns). At the patient level, no significant differences in these parameters were identified (e.g., p = 0.22 and p = 0.35 for accuracy, in the peritoneum and lymph nodes, respectively). Smaller peritoneal/lymph node lesions close to physiological FDG uptake sources were found in the cases of misidentification by PET/ldCT. PET/ceCT prompted a change in clinical management in four cases (3.2%) compared to PET/ldCT. Conclusions: PET/ceCT does not perform better than PET/ldCT but can occasionally clarify doubtful peritoneal findings on PET/ldCT. To avoid unnecessary dose to the patient, PET/ceCT should be excluded in selected cases.
Collapse
Affiliation(s)
- Michela Massollo
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| | - Francesco Fiz
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Milan, Italy
- Correspondence:
| | - Gianluca Bottoni
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| | - Martina Ugolini
- Department of Medical Physics, E.O. “Ospedali Galliera”, 16128 Genoa, Italy;
- Department of Oncology, E.O. “Ospedali Galliera”, 16128 Genoa, Italy; (N.P.); (A.D.)
| | - Francesco Paparo
- Department of Radiology, E.O. “Ospedali Galliera, 16128 Genoa, Italy; (F.P.); (C.P.)
| | - Cristina Puppo
- Department of Radiology, E.O. “Ospedali Galliera, 16128 Genoa, Italy; (F.P.); (C.P.)
| | - Nicoletta Provinciali
- Department of Oncology, E.O. “Ospedali Galliera”, 16128 Genoa, Italy; (N.P.); (A.D.)
| | - Massimiliano Iacozzi
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| | - Vania Altrinetti
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| | - Angelina Cistaro
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| | - Manlio Cabria
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| | - Andrea DeCensi
- Department of Oncology, E.O. “Ospedali Galliera”, 16128 Genoa, Italy; (N.P.); (A.D.)
| | - Giorgio Treglia
- Faculty of Biology and Medicine, University of Lausanne, 1100 Lausanne, Switzerland;
- Academic Education, Research and Innovation Area, General Directorate, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland
| | - Arnoldo Piccardo
- Department of Nuclear Medicine, E.O. “Ospedali Galliera”, Mura delle Cappuccine 14, 16128 Genoa, Italy; (M.M.); (G.B.); (M.I.); (V.A.); (A.C.); (M.C.); (A.P.)
| |
Collapse
|
10
|
LncRNA HOXC-AS3 Suppresses the Formation of Mature miR-96 in Ovarian Cancer Cells to Promote Cell Proliferation. Reprod Sci 2021; 28:2342-2349. [PMID: 33651311 DOI: 10.1007/s43032-021-00500-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 02/14/2021] [Indexed: 12/09/2022]
Abstract
HOXC-AS3 has been reported to be an oncogenic lncRNA in several types of cancer, while its role in ovarian cancer (OC) is unknown. This study aimed to explore the involvement of HOXC-AS3 in OC. The expression levels of HOXC-AS3, mature miR-96, and miR-96 precursor (premature miR-96) in OC and paired non-tumor tissues from 62 OC patients were determined by RT-qPCR. Correlations were analyzed by linear regression. The expression levels of mature miR-96 and miR-96 precursor in OC cells with the overexpression of HOXC-AS3 were determined by RT-qPCR. The roles of HOXC-AS3 and mature miR-96 in regulating the proliferation of OC cells were explored by CCK-8 assay. HOXC-AS3 was upregulated in OC, while mature miR-96 and miR-96 precursor were downregulated in OC. In OC tissues, HOXC-AS3 was inversely correlated with mature miR-96, but not miR-96 precursor. In OC cells, overexpression of HOXC-AS3 reduced the expression levels of mature miR-96, but not miR-96 precursor. Cell proliferation analysis showed that overexpression of HOXC-AS3 resulted in increased cell proliferation, while overexpression of miR-96 suppressed cell proliferation. In addition, overexpression of HOXC-AS3 attenuated the effects of overexpression of miR-96. HOXC-AS3 suppresses the formation of mature miR-96 in OC cells to promote cell proliferation.
Collapse
|
11
|
Diagnostic Efficiency of Gynecologic Imaging Reporting and Data System Combined With 3-dimensional Contrast-enhanced Ultrasound Scoring System in Evaluating Ovarian Tumor. Ultrasound Q 2020; 36:375-381. [DOI: 10.1097/ruq.0000000000000524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Cui Y, Shen G, Zhou D, Wu F. CASC11 Overexpression Predicts Poor Prognosis and Regulates Cell Proliferation and Apoptosis in Ovarian Carcinoma. Cancer Manag Res 2020; 12:523-529. [PMID: 32158258 PMCID: PMC6985985 DOI: 10.2147/cmar.s226801] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 11/28/2019] [Indexed: 01/01/2023] Open
Abstract
Introduction It is known that CASC11 can promote colorectal cancer. However, the function of CASC11 in ovarian carcinoma (OC) remains elusive. Methods In this study, we measured the expression levels of CASC11 and miR-182 in both OC and healthy control samples by performing qPCR. The interaction between CASC11 and miR-182 was analyzed by the overexpression experiment and qPCR. Cell apoptosis was analyzed by cell apoptosis assay, and the prognostic value of CASC11 for OC was analyzed by survival curve analysis. Results We found that CASC11 and microRNA-182 (miRNA-182) were upregulated in OC. Plasma CASC11 was upregulated in OC patients and predicted early-stage OC. Follow-up study revealed that high plasma levels of CASC11 were closely correlated with poor survival conditions of OC patients. CASC11 and miRNA-182 were positively correlated in OC. Overexpression of CASC11 mediated the upregulation of miRNA-182 in cells of OC cell lines, while miRNA-182 overexpression did not significantly affect CASC11 expression. Overexpression of CASC11 and miRNA-182 promoted cancer cell proliferation and inhibited cancer cell apoptosis. Conclusion Therefore, CASC11 overexpression predicts poor prognosis and CASC11 regulates cell proliferation and apoptosis as well as microRNA-182 expression in ovarian carcinoma.
Collapse
Affiliation(s)
- Ying Cui
- Department of Gynecology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing City 100730, People's Republic of China
| | - Guihua Shen
- Department of Gynecology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing City 100730, People's Republic of China
| | - Dan Zhou
- Department of Gynecology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing City 100730, People's Republic of China
| | - Fengli Wu
- Department of Gynecology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing City 100730, People's Republic of China
| |
Collapse
|
13
|
Záveský L, Jandáková E, Weinberger V, Minář L, Hanzíková V, Dušková D, Drábková LZ, Hořínek A. Ovarian Cancer: Differentially Expressed microRNAs in Tumor Tissue and Cell-Free Ascitic Fluid as Potential Novel Biomarkers. Cancer Invest 2019; 37:440-452. [PMID: 31530033 DOI: 10.1080/07357907.2019.1663208] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is the deadliest gynecologic cancer. The large-scale microRNA (miRNA) expression profiling and individual miRNA validation was performed to find potential novel biomarkers for ovarian cancer. The most consistent overexpression of miRs-200b-3p, 135 b-5p and 182-5p was found in both ascitic fluid and tumors and suggests their potential as oncogenes. miR-451a was consistently underexpressed so may be a tumor suppressor. Results were inconsistent for miR-204-5p, which was overexpressed in ascitic fluid but underexpressed in tumor tissue. miR-203a-3p was generally overexpressed but this failed to be proved in independent sample set in tissue validation.
Collapse
Affiliation(s)
- Luděk Záveský
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic
| | - Eva Jandáková
- Department of Pathology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Vít Weinberger
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Luboš Minář
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno , Brno , Czech Republic
| | - Veronika Hanzíková
- Faculty Transfusion Center, General University Hospital in Prague , Prague 2 , Czech Republic
| | - Daniela Dušková
- Faculty Transfusion Center, General University Hospital in Prague , Prague 2 , Czech Republic
| | | | - Aleš Hořínek
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic.,3rd Department of Medicine, Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University and General University Hospital in Prague , Prague 2 , Czech Republic
| |
Collapse
|
14
|
Wang X, Yang S, Lv G, Liao J, Wu S, Zhang W. Combination of GI-RADS and 3D-CEUS for differential diagnosis of ovarian masses. ACTA ACUST UNITED AC 2019; 65:959-964. [PMID: 31389505 DOI: 10.1590/1806-9282.65.7.959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/05/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The purpose of this study is to evaluate the efficacy of the combination of gynecologic imaging reporting and data system (GI-RADS) ultrasonographic stratification and three-dimensional contrast-enhanced ultrasonography (3D-CEUS) in order to distinguish malignant from benign ovarian masses. METHODS In this study, 102 patients with ovarian masses were examined by both two-dimensional ultrasound(2D-US) and 3D-CEUS. Sonographic features of ovarian masses obtained from 3D-CEUS were analyzed and compared with 2D-US. All patients with ovarian masses were confirmed by operational pathology or long-term follow-up results. RESULTS (1)The Chi-square test and multiple Logistic regression analysis confirmed that there were only eight independent predictors of malignant masses, including thick septa (≥3mm), thick papillary projections(≥7mm), solid areas, presence of ascites, central vascularization, contrast enhancement, distribution of contrast agent, and vascular characteristics of the solid part and their odds ratios which were 5.52, 5.39, 4.94, 4.34, 5.92, 7.44, 6.09, and 7.67, respectively (P<0.05). (2)These eight signs were used to combine the GI-RADS with 3D-CEUS scoring system in which the corresponding value of the area under the curve (AUC) was 0.969, which was superior to using GI-RADS lonely (Z-value=1.64, P<0.025). Using 4 points as the cut-off, the scoring system showed the performance was clearly better than using GI-RADS alone (P<0.05). (3) The Kappa value was 0.872 for two different clinicians with equal experience. CONCLUSIONS The combination of GI-RADS and 3D-CEUS scoring system would be a more effective method to distinguish malignant from benign ovarian masses.
Collapse
Affiliation(s)
- Xiali Wang
- Department of Clinical Medicine, Quanzhou Medical College, Quanzhou 362000, China.,Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Shuping Yang
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Guorong Lv
- Department of Clinical Medicine, Quanzhou Medical College, Quanzhou 362000, China.,Department of Ultrasound, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China.,Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Jianmei Liao
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Shufen Wu
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Weina Zhang
- Department of Ultrasound, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| |
Collapse
|
15
|
Gambhir SS, Shankar LK, Rosenthal E, Warram JM, Ghesani M, Hope TA, Jacobs PM, Jacobson GB, Wilson T, Siegel BA. Proceedings: Pathways for Successful Translation of New Imaging Agents and Modalities-Phase III Studies. J Nucl Med 2019; 60:736-744. [PMID: 30850482 DOI: 10.2967/jnumed.118.219824] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022] Open
Affiliation(s)
- Sanjiv S Gambhir
- Departments of Radiology, Bioengineering, and Materials Science and Engineering, Molecular Imaging Program, Stanford University, Stanford, California
| | - Lalitha K Shankar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Eben Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, California
| | - Jason M Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Munir Ghesani
- Department of Radiology, New York University, New York, New York
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Paula M Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | - Gunilla B Jacobson
- Department of Radiology, Molecular Imaging Program, Stanford University, Stanford, California
| | - Terri Wilson
- Blue Earth Diagnostics, Inc., Burlington, Massachusetts; and
| | - Barry A Siegel
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
16
|
Kang SK, Reinhold C, Atri M, Benson CB, Bhosale PR, Jhingran A, Lakhman Y, Maturen KE, Nicola R, Pandharipande PV, Salazar GM, Shipp TD, Simpson L, Small W, Sussman BL, Uyeda JW, Wall DJ, Whitcomb BP, Zelop CM, Glanc P. ACR Appropriateness Criteria ® Staging and Follow-Up of Ovarian Cancer. J Am Coll Radiol 2019; 15:S198-S207. [PMID: 29724422 DOI: 10.1016/j.jacr.2018.03.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/04/2018] [Indexed: 12/12/2022]
Abstract
In the management of epithelial ovarian cancers, imaging is used for cancer detection and staging, both before and after initial treatment. The decision of whether to pursue initial cytoreductive surgery for ovarian cancer depends in part on accurate staging. Contrast-enhanced CT of the abdomen and pelvis (and chest where indicated) is the current imaging modality of choice for the initial staging evaluation of ovarian cancer. Fluorine-18-2-fluoro-2-deoxy-d-glucose PET/CT and MRI may be appropriate for problem-solving purposes, particularly when lesions are present on CT but considered indeterminate. In patients who achieve remission, clinical suspicion for relapse after treatment prompts imaging evaluation for recurrence. Contrast-enhanced CT is the modality of choice to assess the extent of recurrent disease, and fluorine-18-2-fluoro-2-deoxy-d-glucose PET/CT is also usually appropriate, as small metastatic foci may be identified. If imaging or clinical examination confirms a recurrence, the extent of disease and timing of disease recurrence then determines the choice of treatments, including surgery, chemotherapy, and radiation therapy. The American College of Radiology Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision include an extensive analysis of current medical literature from peer reviewed journals and the application of well-established methodologies (RAND/UCLA Appropriateness Method and Grading of Recommendations Assessment, Development, and Evaluation or GRADE) to rate the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where evidence is lacking or equivocal, expert opinion may supplement the available evidence to recommend imaging or treatment.
Collapse
Affiliation(s)
| | - Stella K Kang
- Principal Author, New York University Medical Center, New York, New York.
| | | | - Mostafa Atri
- Toronto General Hospital, Toronto, Ontario, Canada
| | | | | | - Anuja Jhingran
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yulia Lakhman
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Refky Nicola
- State University of New York Upstate Medical University, Syracuse, New York
| | | | | | - Thomas D Shipp
- Brigham & Women's Hospital, Boston, Massachusetts; American Congress of Obstetricians and Gynecologists
| | - Lynn Simpson
- Columbia University, New York, New York; American Congress of Obstetricians and Gynecologists
| | - William Small
- Stritch School of Medicine Loyola University Chicago, Maywood, Illinois
| | - Betsy L Sussman
- The University of Vermont Medical Center, Burlington, Vermont
| | | | | | - Bradford P Whitcomb
- Tripler Army Medical Center, Honolulu, Hawaii; Society of Gynecologic Oncology
| | - Carolyn M Zelop
- Valley Hospital, Ridgewood, New Jersey, and NYU School of Medicine, New York, New York; American Congress of Obstetricians and Gynecologists
| | - Phyllis Glanc
- Specialty Chair, Sunnybrook Health Sciences Centre Bayview Campus, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Co-expression and prognostic significance of the HER family members, EGFRvIII, c-MET, CD44 in patients with ovarian cancer. Oncotarget 2018; 9:19662-19674. [PMID: 29731973 PMCID: PMC5929416 DOI: 10.18632/oncotarget.24791] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/02/2018] [Indexed: 12/23/2022] Open
Abstract
EGFR and HER-2 are important targets but none of the monoclonal antibodies or small molecule tyrosine kinase inhibitors specific for the HER members has been approved for the treatment of patients with ovarian cancers. In some studies, co-expression of other growth factor receptors has been associated with resistance to therapy with the HER inhibitors. The aim of the present study was to determine the relative expression, cellular location, and prognostic significance of HER-family members, the EGFR mutant (EGFRvIII) c-MET, IGF-1R and the cancer stem cell biomarker CD44 in 60 patients with FIGO stage III and IV ovarian cancer. At cut off >5% of tumour cells with positive staining, 62%, 59%, 65% and 45% of the cases were EGFR, HER-2, HER-3 and HER-4 positive, and 3%, 22% and 48.3% of the cases were positive for EGFRvIII, c-MET, and CD44 respectively. Interestingly, 23% co-expressed all four members of the HER family. On univariate analysis, only EGFR staining at >50% of tumour cells (HR = 3.57, p = 0.038) and CD44 staining at 3+ intensity (HR = 7.99, p = 0.004) were associated with a poorer overall survival. EGFR expression (HR = 2.83, p = 0.019) and its co-expression with HER-2, HER-3, HER-2/HER-3, and c-MET were all associated with poorer disease-free survival. Our results suggest co-expression of the HER-family members is common in Stage III and IV ovarian cancer patients. Further studies on the prognostic significance and predictive value of all HER family member proteins for the response to treatment with various forms of the HER inhibitors are warranted.
Collapse
|
18
|
Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, Barker AD, Bastani R, Bhatia S, Bluestone JA, Brawley O, Butte AJ, Coit DG, Davidson NE, Davis M, DePinho RA, Diasio RB, Draetta G, Frazier AL, Futreal A, Gambhir SS, Ganz PA, Garraway L, Gerson S, Gupta S, Heath J, Hoffman RI, Hudis C, Hughes-Halbert C, Ibrahim R, Jadvar H, Kavanagh B, Kittles R, Le QT, Lippman SM, Mankoff D, Mardis ER, Mayer DK, McMasters K, Meropol NJ, Mitchell B, Naredi P, Ornish D, Pawlik TM, Peppercorn J, Pomper MG, Raghavan D, Ritchie C, Schwarz SW, Sullivan R, Wahl R, Wolchok JD, Wong SL, Yung A. Future cancer research priorities in the USA: a Lancet Oncology Commission. Lancet Oncol 2017; 18:e653-e706. [PMID: 29208398 PMCID: PMC6178838 DOI: 10.1016/s1470-2045(17)30698-8] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022]
Abstract
We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US$2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.
Collapse
Affiliation(s)
| | - Chi Van Dang
- Ludwig Institute for Cancer Research New York, NY; Wistar Institute, Philadelphia, PA, USA.
| | - David B Agus
- University of Southern California, Beverly Hills, CA, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Alan Ashworth
- University of California San Francisco, San Francisco, CA, USA
| | | | - Roshan Bastani
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Sangeeta Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey A Bluestone
- University of California San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Atul J Butte
- University of California San Francisco, San Francisco, CA, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Mark Davis
- California Institute for Technology, Pasadena, CA, USA
| | | | | | - Giulio Draetta
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Lindsay Frazier
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Patricia A Ganz
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Levi Garraway
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA; Eli Lilly and Company, Boston, MA, USA
| | | | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Faculty of Medicine and IHPME, University of Toronto, Toronto, Canada
| | - James Heath
- California Institute for Technology, Pasadena, CA, USA
| | - Ruth I Hoffman
- American Childhood Cancer Organization, Beltsville, MD, USA
| | - Cliff Hudis
- Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Chanita Hughes-Halbert
- Medical University of South Carolina and the Hollings Cancer Center, Charleston, SC, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Hossein Jadvar
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Kavanagh
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| | - Rick Kittles
- College of Medicine, University of Arizona, Tucson, AZ, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | - Scott M Lippman
- University of California San Diego Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - David Mankoff
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Mardis
- The Institute for Genomic Medicine at Nationwide Children's Hospital Columbus, OH, USA; College of Medicine, Ohio State University, Columbus, OH, USA
| | - Deborah K Mayer
- University of North Carolina Lineberger Cancer Center, Chapel Hill, NC, USA
| | - Kelly McMasters
- The Hiram C Polk Jr MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dean Ornish
- University of California San Francisco, San Francisco, CA, USA
| | - Timothy M Pawlik
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | | | - Martin G Pomper
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Raghavan
- Levine Cancer Institute, Carolinas HealthCare, Charlotte, NC, USA
| | | | - Sally W Schwarz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Richard Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Sandra L Wong
- Department of Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alfred Yung
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
Pandharipande PV, Lowry KP, Reinhold C, Atri M, Benson CB, Bhosale PR, Green ED, Kang SK, Lakhman Y, Maturen KE, Nicola R, Salazar GM, Shipp TD, Simpson L, Sussman BL, Uyeda J, Wall DJ, Whitcomb B, Zelop CM, Glanc P. ACR Appropriateness Criteria ® Ovarian Cancer Screening. J Am Coll Radiol 2017; 14:S490-S499. [DOI: 10.1016/j.jacr.2017.08.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 11/27/2022]
|
20
|
|
21
|
Willmann JK, Bonomo L, Testa AC, Rinaldi P, Rindi G, Valluru KS, Petrone G, Martini M, Lutz AM, Gambhir SS. Ultrasound Molecular Imaging With BR55 in Patients With Breast and Ovarian Lesions: First-in-Human Results. J Clin Oncol 2017; 35:2133-2140. [PMID: 28291391 DOI: 10.1200/jco.2016.70.8594] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose We performed a first-in-human clinical trial on ultrasound molecular imaging (USMI) in patients with breast and ovarian lesions using a clinical-grade contrast agent (kinase insert domain receptor [KDR] -targeted contrast microbubble [MBKDR]) that is targeted at the KDR, one of the key regulators of neoangiogenesis in cancer. The aim of this study was to assess whether USMI using MBKDR is safe and allows assessment of KDR expression using immunohistochemistry (IHC) as the gold standard. Methods Twenty-four women (age 48 to 79 years) with focal ovarian lesions and 21 women (age 34 to 66 years) with focal breast lesions were injected intravenously with MBKDR (0.03 to 0.08 mL/kg of body weight), and USMI of the lesions was performed starting 5 minutes after injection up to 29 minutes. Blood pressure, ECG, oxygen levels, heart rate, CBC, and metabolic panel were obtained before and after MBKDR administration. Persistent focal MBKDR binding on USMI was assessed. Patients underwent surgical resection of the target lesions, and tissues were stained for CD31 and KDR by IHC. Results USMI with MBKDR was well tolerated by all patients without safety concerns. Among the 40 patients included in the analysis, KDR expression on IHC matched well with imaging signal on USMI in 93% of breast and 85% of ovarian malignant lesions. Strong KDR-targeted USMI signal was present in 77% of malignant ovarian lesions, with no targeted signal seen in 78% of benign ovarian lesions. Similarly, strong targeted signal was seen in 93% of malignant breast lesions with no targeted signal present in 67% of benign breast lesions. Conclusion USMI with MBKDR is clinically feasible and safe, and KDR-targeted USMI signal matches well with KDR expression on IHC. This study lays the foundation for a new field of clinical USMI in cancer.
Collapse
Affiliation(s)
- Jürgen K Willmann
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Lorenzo Bonomo
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Antonia Carla Testa
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Pierluigi Rinaldi
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Guido Rindi
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Keerthi S Valluru
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Gianluigi Petrone
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Maurizio Martini
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Amelie M Lutz
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| | - Sanjiv S Gambhir
- Jürgen K. Willmann, Keerthi S. Valluru, Amelie M. Lutz, and Sanjiv S. Gambhir, Stanford University, Stanford, CA; and Lorenzo Bonomo, Antonia Carla Testa, Pierluigi Rinaldi, Guido Rindi, Gianluigi Petrone, and Maurizio Martini, Universitary Policlinic A. Gemelli-Foundation, Catholic University, Rome, Italy
| |
Collapse
|
22
|
Imaging diagnostics in ovarian cancer: magnetic resonance imaging and a scoring system guiding choice of primary treatment. Eur J Obstet Gynecol Reprod Biol 2017; 210:83-89. [DOI: 10.1016/j.ejogrb.2016.10.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 09/27/2016] [Accepted: 10/21/2016] [Indexed: 11/21/2022]
|
23
|
Sundaram KM, Zhang Y, Mitra AK, Kouadio JLK, Gwin K, Kossiakoff AA, Roman BB, Lengyel E, Piccirilli JA. Prolactin Receptor-Mediated Internalization of Imaging Agents Detects Epithelial Ovarian Cancer with Enhanced Sensitivity and Specificity. Cancer Res 2017; 77:1684-1696. [PMID: 28202518 DOI: 10.1158/0008-5472.can-16-1454] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 12/02/2016] [Accepted: 12/21/2016] [Indexed: 01/24/2023]
Abstract
Poor prognosis of ovarian cancer, the deadliest of the gynecologic malignancies, reflects major limitations associated with detection and diagnosis. Current methods lack high sensitivity to detect small tumors and high specificity to distinguish malignant from benign tissue, both impeding diagnosis of early and metastatic cancer stages and leading to costly and invasive surgeries. Tissue microarray analysis revealed that >98% of ovarian cancers express the prolactin receptor (PRLR), forming the basis of a new molecular imaging strategy. We fused human placental lactogen (hPL), a specific and tight binding PRLR ligand, to magnetic resonance imaging (gadolinium) and near-infrared fluorescence imaging agents. Both in tissue culture and in mouse models, these imaging bioconjugates underwent selective internalization into ovarian cancer cells via PRLR-mediated endocytosis. Compared with current clinical MRI techniques, this targeted approach yielded both enhanced signal-to-noise ratio from accumulation of signal via selective internalization and improved specificity conferred by PRLR upregulation in malignant ovarian cancer. These features endow PRLR-targeted imaging with the potential to transform ovarian cancer detection. Cancer Res; 77(7); 1684-96. ©2017 AACR.
Collapse
Affiliation(s)
- Karthik M Sundaram
- Department of Biochemistry & Molecular Biology, and Chemistry, The University of Chicago, Chicago, Illinois
| | - Yilin Zhang
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, The University of Chicago, Chicago, Illinois
| | - Anirban K Mitra
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, The University of Chicago, Chicago, Illinois
| | - Jean-Louis K Kouadio
- Department of Biochemistry & Molecular Biology, and Chemistry, The University of Chicago, Chicago, Illinois
| | - Katja Gwin
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Anthony A Kossiakoff
- Department of Biochemistry & Molecular Biology, and Chemistry, The University of Chicago, Chicago, Illinois
| | - Brian B Roman
- Department of Radiology, The University of Chicago, Chicago, Illinois
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, The University of Chicago, Chicago, Illinois.
| | - Joseph A Piccirilli
- Department of Biochemistry & Molecular Biology, and Chemistry, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
24
|
Nawaz M, Fatima F, Nazarenko I, Ekström K, Murtaza I, Anees M, Sultan A, Neder L, Camussi G, Valadi H, Squire JA, Kislinger T. Extracellular vesicles in ovarian cancer: applications to tumor biology, immunotherapy and biomarker discovery. Expert Rev Proteomics 2016; 13:395-409. [PMID: 26973172 DOI: 10.1586/14789450.2016.1165613] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years there has been tremendous interest in both the basic biology and applications of extracellular vesicles (EVs) in translational cancer research. This includes a better understanding of their biogenesis and mechanisms of selective cargo packaging, their precise roles in horizontal communication, and their application as non-invasive biomarkers. The rapid advances in next-generation omics technologies are the driving forces for these discoveries. In this review, the authors focus on recent results of EV research in ovarian cancer. A deeper understanding of ovarian cancer-derived EVs, the types of cargo molecules and their biological roles in cancer growth, metastases and drug resistance, could have significant impact on the discovery of novel biomarkers and innovative therapeutics. Insights into the role of EVs in immune regulation could lead to novel approaches built on EV-based immunotherapy.
Collapse
Affiliation(s)
- Muhammad Nawaz
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil.,b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Farah Fatima
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil.,b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Irina Nazarenko
- c Institute for Environmental Health Sciences and Hospital Infection Control , University Medical Centre Freiburg , Freiburg im Breisgau , Germany
| | - Karin Ekström
- d Department of Biomaterials , Sahlgrenska Academy at the University of Gothenburg , Gothenburg , Sweden.,e BIOMATCELL VINN Excellence Centre of Biomaterials and Cell Therapy , Gothenburg , Sweden
| | - Iram Murtaza
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Mariam Anees
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Aneesa Sultan
- f Department of Biochemistry, Faculty of Biological Sciences , Quaid-i-Azam University Islamabad , Islamabad , Pakistan
| | - Luciano Neder
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil
| | - Giovanni Camussi
- g Department of Medical Sciences and Molecular Biotechnology Centre , University of Torino , Torino , Italy
| | - Hadi Valadi
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy at the University of Gothenburg , Guldhedsgatan Sweden
| | - Jeremy A Squire
- a Department of Pathology and Forensic Medicine, Ribeirao Preto School of Medicine , University of Sao Paulo , Sao Paulo , Brazil
| | - Thomas Kislinger
- h Princess Margaret Cancer Centre and Department of Medical Biophysics , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
25
|
Valluru KS, Wilson KE, Willmann JK. Photoacoustic Imaging in Oncology: Translational Preclinical and Early Clinical Experience. Radiology 2016; 280:332-49. [PMID: 27429141 PMCID: PMC4976462 DOI: 10.1148/radiol.16151414] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Photoacoustic imaging has evolved into a clinically translatable platform with the potential to complement existing imaging techniques for the management of cancer, including detection, characterization, prognosis, and treatment monitoring. In photoacoustic imaging, tissue is optically excited to produce ultrasonographic images that represent a spatial map of optical absorption of endogenous constituents such as hemoglobin, fat, melanin, and water or exogenous contrast agents such as dyes and nanoparticles. It can therefore provide functional and molecular information that allows noninvasive soft-tissue characterization. Photoacoustic imaging has matured over the years and is currently being translated into the clinic with various clinical studies underway. In this review, the current state of photoacoustic imaging is presented, including techniques and instrumentation, followed by a discussion of potential clinical applications of this technique for the detection and management of cancer. (©) RSNA, 2016.
Collapse
Affiliation(s)
- Keerthi S. Valluru
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Katheryne E. Wilson
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| | - Jürgen K. Willmann
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, 300 Pasteur Dr, Room H1307, Stanford, CA 94305-5621
| |
Collapse
|
26
|
Katabathina VS, Amanullah FS, Menias CO, Chen MM, Valente PT, Chintapalli KN, Prasad SR. Extrauterine Pelvic Serous Carcinomas: Current Update on Pathology and Cross-sectional Imaging Findings. Radiographics 2016; 36:918-32. [DOI: 10.1148/rg.2016150130] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
27
|
Giusti I, Cervelli C, D'Ascenzo S, Di Francesco M, Ligas C, D'Alessandro E, Papola F, Dolo V. The human ovarian cancer cell line CABA I: A peculiar genetic evolution. Int J Mol Med 2016; 37:879-88. [PMID: 26934856 PMCID: PMC4790663 DOI: 10.3892/ijmm.2016.2501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/22/2015] [Indexed: 12/21/2022] Open
Abstract
The objective of this study was to study the human ovarian cancer cell line CABA I by means of short tandem repeats (STR) profiling and cytogenetic analysis in order to prevent future misidentification or cross-contamination and verify its stability during in vitro cultivation. To this end, cells at passages 18 and 38 were analyzed using cytogenetic techniques in order to verify possible chromosomal aberrations and the karyotypic evolution of this cell line; GTG-banding and FISH were also performed. For STR analysis, DNA was extracted using the automated extractor MagNA pure and analyzed by means of PowerPlex 16 HS. STR profiles were analyzed by GeneMapper 3.2.1 software. Whereas comparative cytogenetic analysis of CABA I cells at passage 18 and 38 has demonstrated considerable genetic instability, we found that STR profiles were essentially unaltered in both analyzed passages, suggesting that the STR profile is reliable and could be used for the regular authentication of CABA I over time. It should be emphasized, however, that of the 16 loci generally used in human STR profiles, only 3 were properly detectable in CABA I. The data highlight that the CABA I cell line demonstrates an anomalous STR profile that does not fully adjust the criteria currently used for the identification of human cells; in spite of this, it remains stable during the in vitro maintainance. Moreover, the genetic instability of the CABA I cell line overlaps with those observed in vivo in tumor cells, making it a suitable candidate to analyze, in vitro, the peculiar genetic evolution of ovarian cancer cells.
Collapse
Affiliation(s)
- Ilaria Giusti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Carla Cervelli
- Regional Center of Immunohematology and Tissue Typing, L'Aquila, Italy
| | - Sandra D'Ascenzo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marianna Di Francesco
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudio Ligas
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elvira D'Alessandro
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Franco Papola
- Regional Center of Immunohematology and Tissue Typing, L'Aquila, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
28
|
Fonville NC, Vaksman Z, McIver LJ, Garner HR. Population analysis of microsatellite genotypes reveals a signature associated with ovarian cancer. Oncotarget 2016; 6:11407-20. [PMID: 25779658 PMCID: PMC4484465 DOI: 10.18632/oncotarget.2933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/16/2014] [Indexed: 11/29/2022] Open
Abstract
Ovarian cancer (OV) ranks fifth in cancer deaths among women, yet there remain few informative biomarkers for this disease. Microsatellites are repetitive genomic regions which we hypothesize could be a source of novel biomarkers for OV and have traditionally been under-appreciated relative to Single Nucleotide Polymorphisms (SNPs). In this study, we explore microsatellite variation as a potential novel source of genomic variation associated with OV. Exomes from 305 OV patient germline samples and 54 tumors, sequenced as part of The Cancer Genome Atlas, were analyzed for microsatellite variation and compared to healthy females sequenced as part of the 1,000 Genomes Project. We identified a subset of 60 microsatellite loci with genotypes that varied significantly between the OV and healthy female populations. Using these loci as a signature set, we classified germline genomes as ‘at risk’ for OV with a sensitivity of 90.1% and a specificity of 87.6%. Cross-analysis with a similar set of breast cancer associated loci identified individuals ‘at risk’ for both diseases. This study revealed a genotype-based microsatellite signature present in the germlines of individuals diagnosed with OV, and provides the basis for a potential novel risk assessment diagnostic for OV and new personal genomics targets in tumors.
Collapse
Affiliation(s)
- Natalie C Fonville
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zalman Vaksman
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Lauren J McIver
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Harold R Garner
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
29
|
Yildirim N, Dikmen Y, Terek MC, Akman L, Gunel NS, Aktan C, Zekioglu O, Gunduz C. Do preoperative serum vascular endothelial growth factor and migration-inhibitory factor predict the nature of the adnexal masses? A prospective-controlled trial. J OBSTET GYNAECOL 2016; 36:533-7. [PMID: 26758243 DOI: 10.3109/01443615.2015.1121978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this study was to identify the role of preoperative serum vascular endothelial growth factor (VEGF) and migration inhibitor factor (MIF) in differentiation of benign and malignant adnexal masses, as well as the relationship between prognostic factors and VEGF and MIF in ovarian cancer patients. This prospective study included 41 patients who were admitted between November 2010 and March 2012. In the malignant group, there were 21 patients, and remaining 20 had benign adnexal masses. Age, CA125 levels, grade, stage, presence of ascites and the degree of cytoreduction performed were noted. There was no significant difference between two groups in preoperative serum VEGF and MIF levels (p = 0.118 and p = 0.297, respectively). CA125 levels were significantly higher in the malignant group (p < 0.0001). There was no significant difference for VEGF and MIF between the groups evaluated for tumour grade, stage, presence of ascites and degree of cytoreduction performed in the malignant group. Preoperative serum, VEGF and MIF levels are not suitable for the differentiation of malignant and benign adnexal masses, and they do not correlate with the prognostic factors of ovarian cancer in this cohort of patients.
Collapse
Affiliation(s)
- Nuri Yildirim
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ege University , Izmir , Turkey
| | - Yilmaz Dikmen
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ege University , Izmir , Turkey
| | - Mustafa Cosan Terek
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ege University , Izmir , Turkey
| | - Levent Akman
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ege University , Izmir , Turkey
| | - Nur Selvi Gunel
- b Department of Medical Biology, Faculty of Medicine , Ege University , Izmir , Turkey , and
| | - Cagdas Aktan
- b Department of Medical Biology, Faculty of Medicine , Ege University , Izmir , Turkey , and
| | - Osman Zekioglu
- c Department of Pathology, Faculty of Medicine , Ege University , Izmir , Turkey
| | - Cumhur Gunduz
- b Department of Medical Biology, Faculty of Medicine , Ege University , Izmir , Turkey , and
| |
Collapse
|
30
|
Tian Y, Wang C, Cheng L, Zhang A, Liu W, Guo L, Ye H, Huang Y, Chen J, Wen X, Xing Y, Zheng G, Sun Z, Li H, Zhang P, Liu W, Chen Y, Zhang Z, Xu Y, Huo Y, Ou Q. Determination of reference intervals of serum levels of human epididymis protein 4 (HE4) in Chinese women. J Ovarian Res 2015; 8:72. [PMID: 26552478 PMCID: PMC4637994 DOI: 10.1186/s13048-015-0201-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
Background To determine reference intervals for serum levels of human epididymis protein 4 (HE4) in Chinese women. Methods In this multicenter (n = 9) study, 618 healthy women, 767 patients with non-malignant diseases, and 951 patients with malignant tumors were enrolled. Serum levels of HE4 were measured in all patients using electrochemiluminescence immunoassays. The influence of age, menopause, malignancy status and other characteristics on the levels of HE4 was evaluated using univariate and multivariate analyses. Confidence intervals (2.5–97.5 %) were determined in different populations. Results There were significant differences in HE4 levels among groups with different ages, menopause or malignancy status. Higher levels of HE4 were detected in elder compared to younger, post- compare to pre- menopause and malignant compared to benign subjects. Multivariate analysis showed that menopause and malignancy status, as well as smoking and pelvic masses were independent factors involved in serum HE4 levels. In pre-menopause stage, the reference ranges of HE4 level were 29.30–68.79, 28.12–1284.83 and 34.75–981.91 pmol/L in healthy, benign and malignant populations, respectively. In post-menopause stage, the reference ranges are 35.96–114.43, 39.11–2208.70 and 39.40–1678.13 pmol/L for those populations. Conclusions The present study has established the reference intervals of HE4 levels in pre- and post-menopause populations with different malignancy status. Electronic supplementary material The online version of this article (doi:10.1186/s13048-015-0201-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yaping Tian
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Chuanxin Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| | - Liming Cheng
- Laboratory Medicine Department, Tongji Hospital, Tongji Medical College of HUST, Wuhan, 430030, Hubei Province, China.
| | - Aimin Zhang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China.
| | - Wen Liu
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Huiming Ye
- Department of Clinical Laboratory, Zhongshan Hospital Xiamen University, Xiamen, 361004, Fujian Province, China.
| | - Yanchun Huang
- Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, 830011, Xinjiang Province, China.
| | - Jing Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China.
| | - Xinyu Wen
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yuelei Xing
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Guixi Zheng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| | - Ziyong Sun
- Laboratory Medicine Department, Tongji Hospital, Tongji Medical College of HUST, Wuhan, 430030, Hubei Province, China.
| | - Huijun Li
- Laboratory Medicine Department, Tongji Hospital, Tongji Medical College of HUST, Wuhan, 430030, Hubei Province, China.
| | - Peng Zhang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China.
| | - Wanli Liu
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong Province, China.
| | - Ying Chen
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Zhongying Zhang
- Department of Clinical Laboratory, Zhongshan Hospital Xiamen University, Xiamen, 361004, Fujian Province, China.
| | - Yi Xu
- Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, 830011, Xinjiang Province, China.
| | - Yishan Huo
- Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, 830011, Xinjiang Province, China.
| | - Qishui Ou
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian Province, China.
| |
Collapse
|
31
|
Cao L, Zhang Y, Fu Z, Dong L, Yang S, Meng W, Li Y, Zhang W, Zhang J, Zheng C, Zhu H, Fan L. Diagnostic value of plasma lysophosphatidic acid levels in ovarian cancer patients: A case-control study and updated meta-analysis. J Obstet Gynaecol Res 2015; 41:1951-8. [PMID: 26472266 DOI: 10.1111/jog.12806] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/01/2015] [Indexed: 01/01/2023]
Abstract
AIM We investigated whether plasma levels of lysophosphatidic acid (LPA) could serve as a diagnostic indicator for assessing disease progression in ovarian cancer (OC) patients. MATERIAL AND METHODS In this study, we enrolled 98 patients with OC, 70 patients with benign ovarian tumors and 75 healthy controls. Plasma levels of LPA and cancer antigen 125 (CA-125) were measured in all study subjects. The diagnostic values of LPA and CA-125 plasma levels were evaluated and an updated meta-analysis was performed to examine the association between LPA plasma levels and OC progression. Statistical analyses were performed with SPSS 18.0 and R 3.1.0 software. RESULTS In our case-control study, OC patients showed significantly higher plasma LPA levels compared to patients with benign tumors and healthy controls (all P < 0.05). Plasma LPA levels exhibited higher diagnostic sensitivity (P = 0.008) and specificity (P = 0.042) in detecting OC, compared to an established marker, CA-125. Notably, the sensitivity for early stage OC was significantly higher for plasma LPA levels in comparison to CA-125 (P < 0.05). Consistent with this, the area under the receiver-operator curve was greater for LPA (0.899) in comparison to that for CA-125 (0.751). Further, meta-analysis showed that plasma LPA levels were significantly higher in OC patients compared to patients with benign tumors or healthy controls (all P < 0.05). CONCLUSION Plasma LPA levels are elevated in OC patients and correlate with disease progression. Further, LPA shows higher sensitivity and specificity in OC diagnosis, compared to CA-125, especially in early stage OC.
Collapse
Affiliation(s)
- Liyan Cao
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Yunjie Zhang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Zhanzhao Fu
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Lixin Dong
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Sen Yang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Wei Meng
- Department of Oncology, The First Affiliated Hospital, Hebei North University, Zhangjiakou, China
| | - Yiyang Li
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Wenchao Zhang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Jialing Zhang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Changjun Zheng
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - He Zhu
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Limei Fan
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| |
Collapse
|
32
|
The clinical value of dynamic contrast-enhanced MRI in differential diagnosis of malignant and benign ovarian lesions. Tumour Biol 2015; 36:5515-22. [DOI: 10.1007/s13277-015-3219-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/03/2015] [Indexed: 10/23/2022] Open
|
33
|
ROMA, an algorithm for ovarian cancer. Clin Chim Acta 2015; 440:143-51. [DOI: 10.1016/j.cca.2014.11.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/03/2014] [Accepted: 11/14/2014] [Indexed: 11/23/2022]
|
34
|
Markushin Y, Sivakumar P, Connolly D, Melikechi N. Tag-femtosecond laser-induced breakdown spectroscopy for the sensitive detection of cancer antigen 125 in blood plasma. Anal Bioanal Chem 2015; 407:1849-55. [PMID: 25577361 DOI: 10.1007/s00216-014-8433-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 11/30/2022]
Abstract
Successful treatment of cancers requires detecting early signs of the disease. One promising way to approach this is to develop minimally invasive tests for the sensitive and specific detection of biomarkers in blood. Irrespective of the detection approach one uses, this remains a challenging task because biomarkers are typically present in low concentrations and there are signals that interfere strongly with prevailing compounds of human fluids. In this paper, we show that elemental encoded particle assay coupled with femtosecond laser-induced breakdown spectroscopy for simultaneous multi-elemental analysis can significantly improve biomarker detectability. An estimated near single molecule per particle efficiency of this method leads to sensitive detection of ovarian cancer biomarker CA125 in human blood plasma. This work opens new ways for earlier detection of cancers and for multiplex assay developments in various analytical applications from proteomics, genomics, and neurology fields.
Collapse
Affiliation(s)
- Yuri Markushin
- Optical Science Center for Applied Research and Applications and Department of Physics and Engineering, Delaware State University, Dover, DE, 19901, USA
| | | | | | | |
Collapse
|
35
|
Wan WN, Zhang YX, Wang XM, Liu YJ, Zhang YQ, Que YH, Zhao WJ. ATAD2 is highly expressed in ovarian carcinomas and indicates poor prognosis. Asian Pac J Cancer Prev 2015; 15:2777-83. [PMID: 24761900 DOI: 10.7314/apjcp.2014.15.6.2777] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The purpose of this study was to explore the expression of ATAD2 in ovarian tumor tissue as well as its relationship with degree of malignancy. Tumor tissue from 110 cases of ovarian cancer was collected in accordance with the Declaration of Helsinki for evaluation of ATAD2 expression immunohistochemistry, quantitative PCR (qPCR) and Western blotting. The correlation between the ATAD2 expression and and the prognosis of ovarian cancer was evaluated by Cox regression model. In addition, HO-8910 and OVCAR-3 cells were transfected with two siRNAs targeting ATAD2. Cell viability was evaluated with MTT assay, and cell migration by transwell migration assay. ATAD2 was shown to be highly expressed in 65.5% (72/110) of ovarian cancer cases, both at transcriptional and protein levels. Moreover, highly expression was positively correlated with degree of malignancy. Knock-down of ATAD2 in HO-8910 and OVCAR-3 cells was found to reduce cell migration. In addition, follow-up visits of the patients demonstrated that the 5-year survival rate was lower in patients with high expression of ATAD2. Our study suggested that ovarian tumor tissue may have highly expressed ATAD2, which is associated with tumor stage, omentum-metastasis, ascites and CA-125. Increased ATAD2 may play important roles in tumor proliferation and migration. ATAD2 could serve in particular as a prognostic marker and a therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Wei-Na Wan
- Department of Ultrasound, First Affiliated Hospital of China Medical University, Shenyang, Liaoning E-mail :
| | | | | | | | | | | | | |
Collapse
|
36
|
Patel S, Ahmed S. Emerging field of metabolomics: big promise for cancer biomarker identification and drug discovery. J Pharm Biomed Anal 2014; 107:63-74. [PMID: 25569286 DOI: 10.1016/j.jpba.2014.12.020] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/07/2014] [Accepted: 12/14/2014] [Indexed: 02/07/2023]
Abstract
Most cancers are lethal and metabolic alterations are considered a hallmark of this deadly disease. Genomics and proteomics have contributed vastly to understand cancer biology. Still there are missing links as downstream to them molecular divergence occurs. Metabolomics, the omic science that furnishes a dynamic portrait of metabolic profile is expected to bridge these gaps and boost cancer research. Metabolites being the end products are more stable than mRNAs or proteins. Previous studies have shown the efficacy of metabolomics in identifying biomarkers associated with diagnosis, prognosis and treatment of cancer. Metabolites are highly informative about the functional status of the biological system, owing to their proximity to organismal phenotypes. Scores of publications have reported about high-throughput data generation by cutting-edge analytic platforms (mass spectrometry and nuclear magnetic resonance). Further sophisticated statistical softwares (chemometrics) have enabled meaningful information extraction from the metabolomic data. Metabolomics studies have demonstrated the perturbation in glycolysis, tricarboxylic acid cycle, choline and fatty acid metabolism as traits of cancer cells. This review discusses the latest progress in this field, the future trends and the deficiencies to be surmounted for optimally implementation in oncology. The authors scoured through the most recent, high-impact papers archived in Pubmed, ScienceDirect, Wiley and Springer databases to compile this review to pique the interest of researchers towards cancer metabolomics.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego 92182, USA.
| | - Shadab Ahmed
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune 411007, India
| |
Collapse
|
37
|
Sehouli J, Reinthaller A, Marth C, Reimer D, Reimer T, Stummvoll W, Angleitner-Boubenizek L, Brandt B, Chekerov R. Intra- and postoperative catumaxomab in patients with epithelial ovarian cancer: safety and two-year efficacy results from a multicentre, single-arm, phase II study. Br J Cancer 2014; 111:1519-25. [PMID: 25225907 PMCID: PMC4200087 DOI: 10.1038/bjc.2014.443] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 06/25/2014] [Accepted: 07/13/2014] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This is the first study investigating the safety and efficacy of the trifunctional antibody catumaxomab administered i.p. at the end of cytoreductive surgery and postoperatively prior to standard chemotherapy in patients with primary epithelial ovarian cancer (EOC). METHODS Patients received i.p. catumaxomab 10 μg intraoperatively and 10, 20, 50 and 150 μg on days 7, 10, 13 and 16, respectively, postoperatively. After the study, patients received standard chemotherapy and were followed for 23 months. The primary endpoint was the rate of postoperative complications. RESULTS Forty-one patients entered the study and were evaluable for safety and 34 were alive at 24 months. Complete tumour resection rate was 68%. Postoperative complications were observed in 51%, the most common anastomotic leakage (7%) and wound infections (5%). The most common catumaxomab-related adverse events were abdominal pain, nausea, vomiting and pyrexia. Thirty-nine percent discontinued catumaxomab therapy, and 98% received chemotherapy post study. Kaplan-Meier estimates of disease-free and overall survival after 24 months were 56% and 85%, respectively. CONCLUSIONS Intra- and close postoperative catumaxomab seems feasible, but efficacy and safety were limited by postsurgical complications. In the future prospective trials are needed to investigate the best schedule of integration of catumaxomab into current treatment strategies for EOC.
Collapse
Affiliation(s)
- J Sehouli
- 1] NOGGO, Ovarian cancer study group of the North-Eastern German Society of Gynaecological Oncology (NOGGO), Berlin, Germany [2] Department of Gynaecology, Charité University Hospital, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
| | - A Reinthaller
- Department of Gynaecology and Gynecologic Oncology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - C Marth
- Department of Obstetrics and Gynaecology, Innsbruck Medical University, Anichstraße 35, A-6020 Innsbruck, Austria
| | - D Reimer
- Department of Gynaecology, Charité University Hospital, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
| | - T Reimer
- Department of Obstetrics and Gynaecology, Klinikum Suedstadt, University of Rostock, Südring 81, D-18059 Rostock, Germany
| | - W Stummvoll
- Department of Obstetrics and Gynaecology, Landesfrauenklinik Linz, Krankenhausstraße 26-30, A-4020 Linz, Austria
| | - L Angleitner-Boubenizek
- Department of Obstetrics and Gynaecology, Landesfrauenklinik Linz, Krankenhausstraße 26-30, A-4020 Linz, Austria
| | - B Brandt
- Neovii (formerly Fresenius) Biotech GmbH, Frankfurter Ring 193a, 80807 Munich, Germany
| | - R Chekerov
- Department of Gynaecology, Charité University Hospital, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
38
|
Bodelon C, Pfeiffer RM, Buys SS, Black A, Sherman ME. Analysis of serial ovarian volume measurements and incidence of ovarian cancer: implications for pathogenesis. J Natl Cancer Inst 2014; 106:dju262. [PMID: 25217774 DOI: 10.1093/jnci/dju262] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Accumulating evidence suggests that many ovarian cancers represent metastases from occult fallopian tube carcinomas or tumors arising within ovarian endometriosis. We hypothesized that small ovarian volumes, as reflected in nonvisualization by transvaginal ultrasound (TVU), would be a marker of lower ovarian cancer risk, whereas enlargement on serial examinations would indicate higher risk. METHODS To address these hypotheses, we analyzed serial ovarian volume measurements determined by TVU for 29321 women (102787 scans) performed in the ovarian cancer screening arm of the Prostate, Lung, Colorectal, and Ovarian (PLCO) trial. Cox models were used to compute hazard ratios (HRs) and 95% confidence intervals (CIs) as measures of association between TVU results and ovarian cancer. We assessed whether increasing ovarian volume preceded diagnosis of ovarian cancer in a nested case-control analysis comparing case patients and control patients matched on age, center, and screening year (1:4 ratio). All statistical tests were two-sided. RESULTS Visualization of normal-appearing ovaries at the last TVU scan was associated with marginally higher ovarian cancer risk compared with nonvisualization of the ovaries (HR = 1.42, 95% CI = 1.00 to 2.01). Ovarian volume increased statistically significantly in ovarian cancer case patients one to two years before diagnosis (P < .001), but not in matched control patients. CONCLUSION Our analysis of TVU data suggests that increasing ovarian volume is associated with greater ovarian cancer risk, but it is only detectable one to two years before diagnosis.
Collapse
Affiliation(s)
- Clara Bodelon
- Division of Cancer Epidemiology and Genetics (CB, RMP, AB, MES) and Division of Cancer Prevention (MES), National Cancer Institute, Rockville, MD; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT (SSB).
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics (CB, RMP, AB, MES) and Division of Cancer Prevention (MES), National Cancer Institute, Rockville, MD; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT (SSB)
| | - Saundra S Buys
- Division of Cancer Epidemiology and Genetics (CB, RMP, AB, MES) and Division of Cancer Prevention (MES), National Cancer Institute, Rockville, MD; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT (SSB)
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics (CB, RMP, AB, MES) and Division of Cancer Prevention (MES), National Cancer Institute, Rockville, MD; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT (SSB)
| | - Mark E Sherman
- Division of Cancer Epidemiology and Genetics (CB, RMP, AB, MES) and Division of Cancer Prevention (MES), National Cancer Institute, Rockville, MD; Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT (SSB)
| |
Collapse
|
39
|
Jokerst JV, Van de Sompel D, Bohndiek SE, Gambhir SS. Cellulose Nanoparticles are a Biodegradable Photoacoustic Contrast Agent for Use in Living Mice. PHOTOACOUSTICS 2014; 2:119-127. [PMID: 25225633 PMCID: PMC4161983 DOI: 10.1016/j.pacs.2014.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/04/2014] [Accepted: 07/16/2014] [Indexed: 05/29/2023]
Abstract
Molecular imaging with photoacoustic ultrasound is an emerging field that combines the spatial and temporal resolution of ultrasound with the contrast of optical imaging. However, there are few imaging agents that offer both high signal intensity and biodegradation into small molecules. Here we describe a cellulose-based nanoparticle with peak photoacoustic signal at 700 nm and an in vitro limit of detection of 6 pM (0.02 mg/mL). Doses down to 0.35 nM (1.2 mg/mL) were used to image mouse models of ovarian cancer. Most importantly, the nanoparticles were shown to biodegrade in the presence of cellulase both through a glucose assay and electron microscopy.
Collapse
Affiliation(s)
- Jesse V. Jokerst
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, 318 Campus Drive, Stanford, CA 94305-5427, United States1
| | - Dominique Van de Sompel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, 318 Campus Drive, Stanford, CA 94305-5427, United States1
| | - Sarah E. Bohndiek
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, 318 Campus Drive, Stanford, CA 94305-5427, United States1
- Bioengineering, Materials Science & Engineering, Bio-X Stanford University, Stanford, CA 94305, United States
| | - Sanjiv S. Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, 318 Campus Drive, Stanford, CA 94305-5427, United States1
- Bioengineering, Materials Science & Engineering, Bio-X Stanford University, Stanford, CA 94305, United States
| |
Collapse
|
40
|
Singh RK, Gaikwad SM, Jinager A, Chaudhury S, Maheshwari A, Ray P. IGF-1R inhibition potentiates cytotoxic effects of chemotherapeutic agents in early stages of chemoresistant ovarian cancer cells. Cancer Lett 2014; 354:254-62. [PMID: 25157649 DOI: 10.1016/j.canlet.2014.08.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 08/13/2014] [Accepted: 08/16/2014] [Indexed: 12/30/2022]
Abstract
The kinetics and effect of hyper activated IGF-1R signaling is not well investigated during acquirement of platinum and taxol resistance in ovarian cancer cells. Herein we reported an upregulated IGF-1R expression in early stages of cisplatin paclitaxel and cisplatin-taxol resistance. Picropodophyllin, an IGF-1R inhibitor, alone and in combination with cisplatin, paclitaxel or both at lowest possible doses could reverse the resistance at early stages. Upregulated IGF-1R was also found in primary tumors of ovarian cancer patients after three to four cycles of platinum-taxol treatment. These findings indicate that a combination of cytotoxic agents and IGF-1R inhibitor is more effective at early stages of chemoresistant ovarian cancer.
Collapse
Affiliation(s)
- Ram K Singh
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Snehal M Gaikwad
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Ankit Jinager
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Smrita Chaudhury
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Amita Maheshwari
- Gynecologic Oncology, Tata Memorial Hospital, Dr. E Borges Road, Parel, Mumbai, Maharashtra, India
| | - Pritha Ray
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India.
| |
Collapse
|
41
|
Karlan BY, Thorpe J, Watabayashi K, Drescher CW, Palomares M, Daly MB, Paley P, Hillard P, Andersen MR, Anderson G, Drapkin R, Urban N. Use of CA125 and HE4 serum markers to predict ovarian cancer in elevated-risk women. Cancer Epidemiol Biomarkers Prev 2014; 23:1383-93. [PMID: 24789859 DOI: 10.1158/1055-9965.epi-13-1361] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Serum markers are used before pelvic imaging to improve specificity and positive predictive value (PPV) of ovarian cancer multimodal screening strategies. METHODS We conducted a randomized controlled pilot trial to estimate surgical PPV of a "2 of 3 tests positive" screening rule, and to compare use of HE4 as a first-line (Arm 1) versus a second-line (Arm 2) screen, in women at high and elevated risk for epithelial ovarian cancer (EOC) at five study sites. Semiannual screening was offered to 208 women ages 25 to 80 years with deleterious BRCA germline mutations and to 834 women ages 35 to 80 years with pedigrees suggesting inherited susceptibility. Annual screening was offered to 130 women ages 45 to 80 years (Risk Group 3) with epidemiologic and serum marker risk factors. Rising marker levels were identified using the parametric empirical Bayes algorithm. RESULTS Both strategies yielded surgical PPV above 25%. Protocol-indicated surgery was performed in 6 women, identifying two ovarian malignancies and yielding a surgical PPV in both arms combined of 33% (95% confidence interval: 4%-78%), 25% in Arm 1 and 50% in Arm 2. Surgical consultation was recommended for 37 women (26 in Arm 1 and 11 in Arm 2). On the basis of 12 women with at least 2 of 3 tests positive (CA125, HE4, or imaging), an intent-to-treat analysis yielded PPV of 14% in Arm 1 and 20% in Arm 2. CONCLUSIONS Positive screens were more frequent when HE4 was included in the primary screen. IMPACT HE4 may be useful as a confirmatory screen when rising CA125 is used alone as a primary screen.
Collapse
Affiliation(s)
- Beth Y Karlan
- Authors' Affiliations: Cedars-Sinai Medical Center, Los Angeles;
| | | | | | - Charles W Drescher
- Fred Hutchinson Cancer Research Center; Pacific Gynecology Specialists, Seattle, Washington
| | | | - Mary B Daly
- Fox Chase Cancer Center, Philadelphia, Pennsylvania; and
| | - Pam Paley
- Pacific Gynecology Specialists, Seattle, Washington
| | | | | | | | | | | |
Collapse
|
42
|
Lutz AM, Bachawal SV, Drescher CW, Pysz MA, Willmann JK, Gambhir SS. Ultrasound molecular imaging in a human CD276 expression-modulated murine ovarian cancer model. Clin Cancer Res 2014; 20:1313-22. [PMID: 24389327 DOI: 10.1158/1078-0432.ccr-13-1642] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE To develop a mouse ovarian cancer model that allows modulating the expression levels of human vascular targets in mouse xenograft tumors and to test whether expression of CD276 during tumor angiogenesis can be visualized by molecularly targeted ultrasound in vivo. EXPERIMENTAL DESIGN CD276-expressing MILE SVEN 1 (MS1) mouse endothelial cells were engineered and used for coinjection with 2008 human ovarian cancer cells for subcutaneous xenograft tumor induction in 15 nude mice. Fourteen control mice were injected with 2008 cells only. After confirming their binding specificity in flow chamber cell attachment studies, anti-CD276 antibody-functionalized contrast microbubbles were used for in vivo CD276-targeted contrast-enhanced ultrasound imaging. RESULTS CD276-targeted ultrasound imaging signal was significantly higher (P = 0.006) in mixed MS1/2008 tumors than in control tumors. Compared with control microbubbles, the ultrasound signal using CD276-targeted microbubbles was significantly higher (P = 0.002), and blocking with purified anti-CD276 antibody significantly decreased (P = 0.0096) the signal in mixed MS1/2008 tumors. Immunofluorescence analysis of the tumor tissue confirmed higher quantitative immunofluorescence signal in mixed MS1/2008 tumors than in control 2008 only tumors, but showed not significantly different (P = 0.54) microvessel density. CONCLUSIONS Our novel small animal model allows for modulating the expression of human tumor-associated vascular endothelial imaging targets in a mouse host and these expression differences can be visualized noninvasively by ultrasound molecular imaging. The animal model can be applied to other human vascular targets and may facilitate the preclinical development of new imaging probes such as microbubbles targeted at human vascular markers not expressed in mice.
Collapse
Affiliation(s)
- Amelie M Lutz
- Authors' Affiliations: Departments of Radiology and Bioengineering and Materials Science and Engineering; Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California; and Division of Public Health Sciences, Fred Hutchinson, Cancer Research Center, Seattle, Washington
| | | | | | | | | | | |
Collapse
|
43
|
Xu G, Wang J, Liu T, Wang M, Zhou S, Wu B, Jiang M. Synthesis and crystal structure of a novel copper(ii) complex of curcumin-type and its application in in vitro and in vivo imaging. J Mater Chem B 2014; 2:3659-3666. [DOI: 10.1039/c4tb00133h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The results of TPEF imaging in a tumor-bearing mouse model demonstrated the potential of the obtained complex for in vivo tumor diagnosis.
Collapse
Affiliation(s)
- Guoyong Xu
- Center of Modern Experimental Technology
- Anhui University
- Hefei 230039, P.R. China
| | - Jiafeng Wang
- Department of Pharmacy
- Anhui University of Chinese Medicine
- Hefei 230038, P.R. China
| | - Tao Liu
- Department of Pharmacy
- Anhui University of Chinese Medicine
- Hefei 230038, P.R. China
| | - Mahong Wang
- Center of Modern Experimental Technology
- Anhui University
- Hefei 230039, P.R. China
| | - Shuangsheng Zhou
- Center of Modern Experimental Technology
- Anhui University
- Hefei 230039, P.R. China
- Department of Pharmacy
- Anhui University of Chinese Medicine
| | - Baoxing Wu
- Department of Pharmacy
- Anhui University of Chinese Medicine
- Hefei 230038, P.R. China
- State Key Laboratory of Crystal Materials
- Shandong University
| | - Minghua Jiang
- State Key Laboratory of Crystal Materials
- Shandong University
- Jinan 502100, P.R. China
| |
Collapse
|
44
|
Rubini G, Altini C, Notaristefano A, Merenda N, Rubini D, Ianora AS, Asabella AN. Role of 18F-FDG PET/CT in diagnosing peritoneal carcinomatosis in the restaging of patient with ovarian cancer as compared to contrast enhanced CT and tumor marker Ca-125. Rev Esp Med Nucl Imagen Mol 2014. [DOI: 10.1016/j.remnie.2013.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Abstract
Ascites tumor cells (ATCs) represent a potentially valuable source of cells for monitoring treatment of ovarian cancer as it would obviate the need for more invasive surgical biopsies. The ability to perform longitudinal testing of ascites in a point-of-care setting could significantly impact clinical trials, drug development, and clinical care. Here, we developed a microfluidic chip platform to enrich ATCs from highly heterogeneous peritoneal fluid and then perform molecular analyses on these cells. We evaluated 85 putative ovarian cancer protein markers and found that nearly two-thirds were either nonspecific for malignant disease or had low abundance. Using four of the most promising markers, we prospectively studied 47 patients (33 ovarian cancer and 14 control). We show that a marker set (ATCdx) can sensitively and specifically map ATC numbers and, through its reliable enrichment, facilitate additional treatment-response measurements related to proliferation, protein translation, or pathway inhibition.
Collapse
|
46
|
Mitchell DG, Javitt MC, Glanc P, Bennett GL, Brown DL, Dubinsky T, Harisinghani MG, Harris RD, Horowitz NS, Pandharipande PV, Pannu HK, Podrasky AE, Royal HD, Shipp TD, Siegel CL, Simpson L, Wong-You-Cheong JJ, Zelop CM. ACR Appropriateness Criteria Staging and Follow-up of Ovarian Cancer. J Am Coll Radiol 2013; 10:822-7. [DOI: 10.1016/j.jacr.2013.07.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 01/22/2023]
|
47
|
Foygel K, Wang H, Machtaler S, Lutz AM, Chen R, Pysz M, Lowe AW, Tian L, Carrigan T, Brentnall TA, Willmann JK. Detection of pancreatic ductal adenocarcinoma in mice by ultrasound imaging of thymocyte differentiation antigen 1. Gastroenterology 2013; 145:885-894.e3. [PMID: 23791701 PMCID: PMC3783557 DOI: 10.1053/j.gastro.2013.06.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 05/14/2013] [Accepted: 06/14/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Early detection of pancreatic ductal adenocarcinoma (PDAC) allows for surgical resection and increases patient survival times. Imaging agents that bind and amplify the signal of neovascular proteins in neoplasms can be detected by ultrasound, enabling accurate detection of small lesions. We searched for new markers of neovasculature in PDAC and assessed their potential for tumor detection by ultrasound molecular imaging. METHODS Thymocyte differentiation antigen 1 (Thy1) was identified as a specific biomarker of PDAC neovasculature by proteomic analysis. Up-regulation in PDAC was validated by immunohistochemical analysis of pancreatic tissue samples from 28 healthy individuals, 15 with primary chronic pancreatitis tissues, and 196 with PDAC. Binding of Thy1-targeted contrast microbubbles was assessed in cultured cells, in mice with orthotopic PDAC xenograft tumors expressing human Thy1 on the neovasculature, and on the neovasculature of a genetic mouse model of PDAC. RESULTS Based on immunohistochemical analyses, levels of Thy1 were significantly higher in the vascular of human PDAC than chronic pancreatitis (P = .007) or normal tissue samples (P < .0001). In mice, ultrasound imaging accurately detected human Thy1-positive PDAC xenografts, as well as PDACs that express endogenous Thy1 in genetic mouse models of PDAC. CONCLUSIONS We have identified and validated Thy1 as a marker of PDAC that can be detected by ultrasound molecular imaging in mice. The development of a specific imaging agent and identification of Thy1 as a new biomarker could aid in the diagnosis of this cancer and management of patients.
Collapse
Affiliation(s)
- Kira Foygel
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS); Stanford University, Stanford, California, USA
| | - Huaijun Wang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS); Stanford University, Stanford, California, USA
| | - Steven Machtaler
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS); Stanford University, Stanford, California, USA
| | - Amelie M. Lutz
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS); Stanford University, Stanford, California, USA
| | - Ru Chen
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Marybeth Pysz
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS); Stanford University, Stanford, California, USA
| | - Anson W. Lowe
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Lu Tian
- Department of Health, Research & Policy, Stanford University, Stanford, California, USA
| | - Tricia Carrigan
- Translational Diagnostics, Ventana Medical Systems, INC, Tucson, Arizona, USA
| | | | - Jürgen K. Willmann
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS); Stanford University, Stanford, California, USA
| |
Collapse
|
48
|
Role of 18F-FDG PET/CT in diagnosing peritoneal carcinomatosis in the restaging of patient with ovarian cancer as compared to contrast enhanced CT and tumor marker Ca-125. Rev Esp Med Nucl Imagen Mol 2013; 33:22-7. [PMID: 23948509 DOI: 10.1016/j.remn.2013.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/26/2013] [Accepted: 06/30/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVES To investigate the role of whole-body fluorine-18-2-deoxy-2-fluoro-d-glucose positron emission tomography/computed tomography ((18)F-FDG PET/CT) in the identification of peritoneal carcinomatosis in patients with ovarian cancer (OC). MATERIAL AND METHODS Seventy-nine patients with histologically proven stages III-IV OC who underwent (18)F-FDG PET/CT were studied retrospectively. We considered group A as 51 patients who also underwent computed-tomography with contrast-enhancement (CECT), and group B as 35 patients who had also been tested for biomarker Ca-125. Sensitivity, specificity, accuracy, positive predictive values (PPV) and negative predictive values (NPV) of (18)F-FDG PET/CT as compared to CECT and to Ca-125 were evaluated. RESULTS (18)F-FDG PET/CT' sensitivity, specificity, accuracy, PPV and NPV for all 79 patients were: 85%, 92.31%, 88.61%, 91.89% and 85.71%, respectively. (18)F-FDG PET/CT sensitivity in group A was 78.6%, while it was 53.6% for CECT. (18)F-FDG PET/CT specificity, calculated in the same group, was 91.3%, while that of CECT was 60.9% (statistically significant difference, McNemar 4, P=0.039). Accuracy was 84.3% and 56.9%, respectively. (18)F-FDG PET/CT' sensitivity in group B was 86.4%, while that of Ca-125 was 81.8% (no statistical difference, McNemar 0, P=1). (18)F-FDG PET/CT specificity in group B was 84.6% while that of Ca-125 was 38.5% (clear but not statistically significant difference, McNemar 3.12, P=0.070). Accuracy calculated in the same group was 85.7% for (18)F-FDG PET/CT and 65.7% for Ca-125. CONCLUSION (18)F-FDG PET/CT is a useful diagnostic tool when peritoneal biopsy cannot be performed and it can better select those who are candidates for adjuvant chemotherapy.
Collapse
|
49
|
Vargas HA, Barrett T, Sala E. MRI of ovarian masses. J Magn Reson Imaging 2013; 37:265-81. [PMID: 23355430 DOI: 10.1002/jmri.23721] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 05/02/2012] [Indexed: 11/10/2022] Open
Abstract
MRI provides exquisite views of the pelvic anatomy through its high spatial resolution and tissue contrast, and as such plays a key role in the work up of ovarian lesions, identifying features that distinguish benign and malignant lesions. In the case of primary tumors it enables local staging and detection of metastatic disease to help guide management options such as complex surgery or the consideration of neoadjuvant chemotherapy. Functional MRI techniques such as diffusion-weighted MRI (DW-MRI), dynamic contrast-enhanced MRI (DCE-MRI) and tumor-selective molecular imaging are currently being evaluated as possible predictive and prognostic biomarkers in the context of ovarian malignancy, and may play a larger role in routine clinical practice in the future. Herein we provide an overview of the conventional and advanced MRI techniques used to characterize ovarian masses and of the role that MR plays in the staging, treatment selection and follow up of patients with ovarian cancer.
Collapse
Affiliation(s)
- Hebert Alberto Vargas
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| | | | | |
Collapse
|
50
|
Targeted paclitaxel nanoparticles modified with follicle-stimulating hormone β 81-95 peptide show effective antitumor activity against ovarian carcinoma. Int J Pharm 2013; 453:498-505. [PMID: 23811008 DOI: 10.1016/j.ijpharm.2013.06.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 04/28/2013] [Accepted: 06/12/2013] [Indexed: 02/07/2023]
Abstract
The majority of patients with advanced ovarian cancer will experience a relapse and ultimately die from refractory diseases. Targeted therapy shows promise for these patients. Novel therapeutic strategies should be developed on the basis of the molecular mechanisms involved in ovarian cancer and the steroid hormone environment of ovaries. The ovary is the main target organ of follicle-stimulating hormone (FSH), which bind to its receptor with high affinity. In this study a FSH receptor-targeting ligand, FSH β 81-95 peptide, was used as a targeting moiety to synthesize an FSH receptor-mediated drug delivery system. FSH β 81-95 peptide-conjugated nanoparticles (FSH81-NPs) and paclitaxel-loaded FSH81-NPs (FSH81-NP-PTXs) were synthesized. In vitro studies showed that FSH β 81-95 peptide enabled the specific uptake of cytotoxic drugs and increased the intracellular paclitaxel concentration in FSH receptor-expressing cancer cells, resulting in enhanced cytotoxic effects. In vivo studies showed that FSH81-NP-PTXs possessed higher antitumor efficacy against FSH receptor-expressing tumors without any clinical signs of adverse side effects or body weight loss due to modification with FSH β 81-95 peptide. Therefore, FSH binding peptide-targeted drug delivery system exhibited high potential in the treatment of ovarian cancer, and tumor targeting via reproductive hormone receptors might improve the outcome of diseases.
Collapse
|