51
|
Zhang XF, Pawlik TM. ASO Author Reflections: Hepatocellular Carcinoma-Related Tumor Necrosis is Associated with Worse Long-Term Outcomes. Ann Surg Oncol 2020; 28:806-807. [PMID: 33237450 DOI: 10.1245/s10434-020-09396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Xu-Feng Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Timothy M Pawlik
- Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
52
|
Mohanty SS. Correlation of expression of hormone and HER2 receptors with various clinico-pathological prognostic parameters and with each other in malignant breast lesion. Ann Diagn Pathol 2020; 50:151659. [PMID: 33249360 DOI: 10.1016/j.anndiagpath.2020.151659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/25/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Estrogen receptors (ER), progesterone receptors (PR) and the human epidermal growth factor receptor-2 (HER2) are basic breast cancer molecular markers that are also best recognized prognostic factors and predictors of type of targeted therapy to be given. The objectives are to study the correlation of expression of hormone and HER2receptors with various clinico-pathological prognostic parameters like patient's age at diagnosis, menopausal status, tumor size, histological grade and lymph node status of tumor and with each other in malignant breast lesion. METHODS For this study histopathology (HP) and immunohistochemistry (IHC) slides of excised specimens of 330 female patients with a palpable breast lump deposited to the pathology department of a hospital as a part of routine diagnostic procedure, were evaluated under the guidance of trained doctors who have minimum 5 years of experience in oncopathology. The author has no direct involvement with patients, informed consent was not necessary and data were collected after getting permission from concerned authority. RESULTS This study finds significant relationship between hormone receptors and all clinico-pathological prognostic parameters taken for comparison except age at diagnosis. HER2 status has significant relationship with all clinico-pathological prognostic parameters; hormone and HER2 status suggests an inverse relationship. CONCLUSIONS Mien of hormone receptors expression in breast cancer is related with better prognostic factors such as older age, postmenopausal status, smaller tumor size, low histological grade and negative lymph node status, however the opposite is correct for HER2. Hormone receptors and HER2 have an inversely proportionate relationship with each other.
Collapse
Affiliation(s)
- Swati Sucharita Mohanty
- Cytogenetics Laboratory, P.G. Department of Zoology, Utkal University, Bhubaneswar 751004, Odisha, India.
| |
Collapse
|
53
|
Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. Biochim Biophys Acta Rev Cancer 2020; 1874:188427. [PMID: 32961257 DOI: 10.1016/j.bbcan.2020.188427] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023]
Abstract
Macrophages are innate phagocytic leukocytes that are highly present in solid tumors, where they are referred to as tumor-associated macrophages (TAMs). In solid tumors, the microenvironment is often immunosuppressive and hypoxic regions are prevalent. These hypoxic conditions impose tumor cells to reprogram their metabolism, shifting from oxidative phosphorylation to anaerobic glycolysis. This so-called glycolytic switch enables hypoxic tumor cells to survive, proliferate, and eventually to outcompete untransformed cells. The hypoxia-induced change in tumor cell metabolism leads to the production of oncometabolites, among which are the glycolytic end-metabolite lactate and the tricarboxylic acid cycle intermediate succinate. TAMs can react to these oncometabolites, resulting in an altered maturation and the adoption of pro-angiogenic features. These angiogenesis-promoting TAMs have been reported to cooperate with tumor cells in the formation of new vessels, and even have been considered an important cause of resistance against anti-angiogenic therapies. For a long time, the mechanisms by which lactate and succinate activated pro-angiogenic TAMs were not understood. Researchers now start to unravel and understand some of the underlying mechanisms. Here, the importance of microenvironmental cues in inducing different macrophage activation states is discussed, as well as the role of hypoxia in the recruitment and activation of pro-angiogenic macrophages. In addition, the latest findings on the oncometabolites lactate and succinate in the activation of angiogenesis supporting macrophages are reviewed. Finally, various oncometabolite-targeting therapeutic strategies are proposed that could improve the response to anti-angiogenic therapies. SIGNIFICANCE STATEMENT: Tumor-associated macrophages (TAMs) are known promotors of tumor neovascularization, and significantly contribute to the emergence of resistance to anti-angiogenic therapies. Recent evidence suggests that the angiogenesis promoting phenotype of TAMs can be activated by hypoxic tumor cell-derived oncometabolites, including lactate and succinate. Here, the latest findings into the lactate- and succinate-mediated mechanistic activation of pro-angiogenic TAMs are reviewed, and therapeutic strategies that interfere with this mechanism and may delay or even prevent acquired resistance to anti-angiogenic agents are discussed.
Collapse
|
54
|
Zhao R, Ma WJ, Tang J, Chen YZ, Zhang LN, Lu H, Liu PF. Heterogeneity of enhancement kinetics in dynamic contrast-enhanced MRI and implication of distant metastasis in invasive breast cancer. Clin Radiol 2020; 75:961.e25-961.e32. [PMID: 32859381 DOI: 10.1016/j.crad.2020.07.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
Abstract
AIM To investigate the heterogeneity of enhancement kinetics for breast tumour in order to demonstrate the predictive power of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) features for distant metastasis (DM) in invasive breast cancer. MATERIALS AND METHODS Time-signal intensity curve (TIC) patterns from 128 patients with invasive breast cancer were analysed by a pixel-based DCE-MRI analysis. This MRI technique enabled pixels with varying TIC patterns (persistent, plateau, washout and non-enhancement) to be categorised semi-automatically and the percentage of different TIC patterns in each breast tumour to be calculated. The percentage of TIC patterns was compared between the DM and non-DM groups. DM-free survival was estimated using Kaplan-Meier survival analysis. RESULTS This study demonstrated a larger percentage of persistent TIC and non-enhancement TIC was associated with DM in invasive breast cancer. The cut-off values of persistent TIC and non-enhancement TIC were 22.5% and 2.5%. Combining TIC patterns and traditional predictors (tumour size and axillary lymph node status) can improve the prediction efficiency. The multivariable model yielded an area under the receiver operating characteristic curve (AUC) of 0.87 with 0.70 sensitivity and 0.87 specificity in leave-one-out cross-validation (LOOCV). These predictors showed significant differences in DM-free survival by Kaplan-Meier analysis. CONCLUSION This study shows that breast tumours with higher heterogeneity are more likely to metastasise, and pixel-based TIC analysis has utility in predicting distant metastasis of invasive breast cancer.
Collapse
Affiliation(s)
- R Zhao
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - W J Ma
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - J Tang
- Department of Radiology, TEDA International Cardiovascular Hospital, Tianjin, PR China
| | - Y Z Chen
- Department of Tumour Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - L N Zhang
- The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - H Lu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| | - P F Liu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| |
Collapse
|
55
|
Yoon GY, Choi WJ, Cha JH, Shin HJ, Chae EY, Kim HH. The role of MRI and clinicopathologic features in predicting the invasive component of biopsy-confirmed ductal carcinoma in situ. BMC Med Imaging 2020; 20:95. [PMID: 32787871 PMCID: PMC7424652 DOI: 10.1186/s12880-020-00494-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/03/2020] [Indexed: 12/21/2022] Open
Abstract
Background The upgrade rate of biopsy-confirmed ductal carcinoma in situ (DCIS) to invasive carcinoma is up to 50% on final pathology. We investigated MRI and clinicopathologic predictors of the invasive components of DCIS diagnosed by preoperative biopsy and then compared MRI features between patients with DCIS, microinvasive ductal carcinoma (mIDC), and invasive ductal carcinoma (IDC) diagnosed on final pathology. Methods Two hundred and one patients with 206 biopsy-confirmed DCIS lesions were enrolled. MRI and clinicopathologic features were used to predict either mIDC or IDC via a cumulative logistic regression analysis. For the lesions detected on MRI, morphologic and kinetic analyses were performed using the Chi-square, Fisher’s exact, and Kruskal-Wallis tests. Results Of all the lesions, 112 (54.4%) were diagnosed as DCIS, 50 (24.3%) were upgraded to mIDC, and 44 (21.4%) to IDC. The detection on MRI as mass (Odds ratio (OR) = 8.84, 95% confidence interval (CI) = 1.05–74.04, P = 0.045) or non-mass enhancement (NME; OR = 11.17, 95% CI = 1.35–92.36, P = 0.025), negative progesterone receptor (PR; OR = 2.40, 95% CI = 1.29–4.44, P = 0.006), and high Ki-67 level (OR = 2.42, 95% CI = 1.30–4.50, P = 0.005) were significant independent predictors of histologic upgrade. On MRI, 87 (42.2%) lesions appeared as mass and 107 (51.9%) as NME. Irregularly shaped, not-circumscribed, heterogeneous, or rim-enhancing masses with intratumoral high signal intensity or peritumoral edema, clumped or clustered ring-enhancing NMEs, and high peak enhancement were significantly associated with histologic upgrade (P < 0.001). Conclusion MRI detection, negative PR, and high Ki-67 levels are associated with a histologic upgrade in patients with biopsy-confirmed DCIS. Suspicious MRI features are more frequent in such patients.
Collapse
Affiliation(s)
- Ga Young Yoon
- Department of Radiology, Gangneung Asan Hospital, University of Ulsan College of Medicine, 38 Bangdong-gil, Sacheon-myeon, Gangneung-si, Gangwon-do, 25440, Korea.,Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Woo Jung Choi
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| | - Joo Hee Cha
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Hee Jung Shin
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Eun Young Chae
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Hak Hee Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| |
Collapse
|
56
|
Ling YH, Chen JW, Wen SH, Huang CY, Li P, Lu LH, Mei J, Li SH, Wei W, Cai MY, Guo RP. Tumor necrosis as a poor prognostic predictor on postoperative survival of patients with solitary small hepatocellular carcinoma. BMC Cancer 2020; 20:607. [PMID: 32600297 PMCID: PMC7325084 DOI: 10.1186/s12885-020-07097-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 06/22/2020] [Indexed: 02/08/2023] Open
Abstract
Background Small hepatocellular carcinoma (sHCC) is a special subtype of HCC with the maximum tumor diameter ≤ 3 cm and excellent long-term outcomes. Surgical resection or radiofrequency ablation provides the greatest chance for cure; however, many patients still undergo tumor recurrence after primary treatment. To date, there is no clinical applicable method to assess biological aggressiveness in solitary sHCC. Methods In the current study, we retrospectively evaluated tumor necrosis of 335 patients with solitary sHCC treated with hepatectomy between December 1998 and 2010 from Sun Yat-sen University Cancer Center. Results The presence of tumor necrosis was observed in 157 of 335 (46.9%) sHCC patients. Further correlation analysis showed that tumor necrosis was significantly correlated with tumor size and vascular invasion (P = 0.026, 0.003, respectively). The presence of tumor necrosis was associated closely with poorer cancer-specific overall survival (OS) and recurrence-free survival (RFS) as evidenced by univariate (P < 0.001; hazard ratio, 2.821; 95% CI, 1.643–4.842) and multivariate analysis (P = 0.005; hazard ratio, 2.208; 95% CI, 1.272–3.833). Notably, the combined model by tumor necrosis, vascular invasion and tumor size can significantly stratify the risk for RFS and OS and improve the ability to discriminate sHCC patients’ outcomes (P < 0.0001 for both). Conclusions Our results provide evidence that tumor necrosis has the potential to be a parameter for cancer aggressiveness in solitary sHCC. The combined prognostic model may be a useful tool to identify solitary sHCC patients with worse outcomes.
Collapse
Affiliation(s)
- Yi-Hong Ling
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Jie-Wei Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Shi-Hong Wen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chao-Yun Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Peng Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Liang-He Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Jie Mei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Shao-Hua Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Wei Wei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China
| | - Mu-Yan Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China. .,Department of Pathology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China.
| | - Rong-Ping Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China. .,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
57
|
Samaratunga H, Delahunt B, Srigley JR, Berney DM, Cheng L, Evans A, Furusato B, Leite KRM, MacLennan GT, Martignoni G, Moch H, Pan CC, Paner G, Ro J, Thunders M, Tsuzuki T, Wheeler T, van der Kwast T, Varma M, Williamson SR, Yaxley JW, Egevad L. Granular necrosis: a distinctive form of cell death in malignant tumours. Pathology 2020; 52:507-514. [PMID: 32561208 DOI: 10.1016/j.pathol.2020.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
Foci of necrosis are frequently seen in malignant tumours and may be due to a variety of causes. Different types of necrosis are given various names based upon their morphological features and presumed pathogenesis, such as coagulative, liquefactive and fibrinoid necrosis. Here, we propose the term 'granular necrosis' (GN) for a specific form of tumour necrosis characterised by the presence of well-defined necrotic foci being sharply demarcated from adjacent viable tumour. A constant feature is loss of architecture resulting in an amorphous necrotic mass containing granular nuclear and cytoplasmic debris, without an associated neutrophilic infiltrate. There is usually extensive karyorrhexis, which in larger tumours is more prominent at the periphery. These foci are often microscopic but may range up to several millimetres or larger in size. This distinctive form of necrosis has been erroneously given a variety of names in the literature including coagulative necrosis and microscopic necrosis, which on the basis of the aforementioned gross and microscopic findings is inappropriate. It is apparent that this is a specific form of necrosis, hence the descriptive term 'granular necrosis' that differentiates this form of necrosis from other types. The presence of GN is recognised as occurring in a variety of tumour types, being commonly seen in renal cell carcinoma, where it has been shown to have independent prognostic significance. In some epithelial and stromal tumours of the uterus, the presence of GN also has prognostic significance and is a defining feature for the differentiation of uterine leiomyoma and leiomyosarcoma. The pathogenesis of GN is unresolved. It does not show the features of apoptosis and in recent studies has been shown to have some of the molecular changes associated with necroptosis.
Collapse
Affiliation(s)
| | - Brett Delahunt
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand.
| | - John R Srigley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Daniel M Berney
- Department of Molecular Oncology, Queen Mary University Hospital, London, United Kingdom
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew Evans
- Department of Laboratory Information Support Systems, University Health Network, Toronto, ON, Canada
| | - Bungo Furusato
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences and Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Katia R M Leite
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Gregory T MacLennan
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Guido Martignoni
- Department of Pathology and Diagnostics, School of Medicine and Surgery, University of Verona, Verona, Italy
| | - Holger Moch
- University and University Hospital Zurich, Department of Pathology and Molecular Pathology, Zurich, Switzerland
| | - Chin-Chen Pan
- Department of Pathology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Gladell Paner
- Departments of Pathology and Surgery (Section of Urology) University of Chicago, Chicago, IL, USA
| | - Jae Ro
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Medical College of Cornell University, Houston, TX, USA
| | - Michelle Thunders
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University, School of Medicine, Nagakute, Japan
| | - Thomas Wheeler
- Department of Pathology and Laboratory Medicine, Baylor St Luke's Medical Center and Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Theodorus van der Kwast
- Department of Pathology, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Murali Varma
- Department of Cellular Pathology, University Hospital of Wales, Cardiff, UK
| | | | - John W Yaxley
- Department of Medicine, University of Queensland, Wesley Urology Clinic, Royal Brisbane and Womens Hospital, Brisbane, Qld, Australia
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
58
|
Kim H, Lee J, Kang BJ, Kim SH. What shear wave elastography parameter best differentiates breast cancer and predicts its histologic aggressiveness? Ultrasonography 2020; 40:265-273. [PMID: 32660207 PMCID: PMC7994732 DOI: 10.14366/usg.20007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/15/2020] [Indexed: 01/09/2023] Open
Abstract
Purpose This study aimed to identify useful shear wave elastography (SWE) parameters for differentiating breast cancer and predicting associated immunohistochemical factors and subtypes. Methods From November 2018 to February 2019, a total of 211 breast lesions from 190 patients who underwent conventional breast ultrasonography and SWE were included. The Breast Imaging Reporting and Data System categories and qualitative and quantitative SWE parameters for each lesion were obtained. Pathologic results including immunohistochemical factors were evaluated. The diagnostic performance of each parameter and its correlation with histological characteristics, immunohistochemical factors, and subtypes of breast cancer were analyzed using analysis of variance, the independent t test, the Fisher exact test, logistic regression analysis, and the DeLong method. Results Among 211 breast lesions, 82 were malignant, and 129 were benign. Of the SWE parameters, Emax showed the highest area under the curve (AUC) for differentiating malignant from benign lesions (AUC, 0.891; cut-off>50.85). Poor tumor differentiation and progesterone receptor-negativity were correlated with higher SDmean and Emax (P<0.05). Ki-67-positive breast cancer showed higher SDmean and a heterogeneous color distribution (P<0.05). Ki-67 and cytokeratin 5/6-positive breast cancers showed higher Emax/Efat ratios (P<0.05). Luminal B, human epidermal growth factor receptor 2-enriched, and triple-negative (non-basal) subtypes showed somewhat higher SDmean values than the luminal A and triple-negative (basal) subtypes (P=0.028). Conclusion Emax is a reliable parameter for differentiating malignancies from benign breast lesions. In addition, high stiffness and SDmean values in tumors measured on SWE could be used to predict poorly differentiated, progesterone receptor-negative, or Ki-67-positive breast cancer.
Collapse
Affiliation(s)
- Hyunjin Kim
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeongmin Lee
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bong Joo Kang
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Hun Kim
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
59
|
Li J, Huang S, Zeng L, Li K, Yang L, Gao S, Guan C, Zhang S, Lao X, Liao G, Liang Y. Necroptosis in head and neck squamous cell carcinoma: characterization of clinicopathological relevance and in vitro cell model. Cell Death Dis 2020; 11:391. [PMID: 32444644 PMCID: PMC7244585 DOI: 10.1038/s41419-020-2538-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
Abstract
Necroptosis is a recently discovered form of programmed cell death (PCD) having necrotic-like morphology. However, its presence and potential impact with respect to head and neck squamous cell carcinoma (HNSCC) are still unknown. The aim of this study was to reveal the necroptosis status and its clinicopathological relevance in HNSCC and to establish an in vitro model. We first analyzed the level of p-MLKL, MLKL, and tumor necrosis in HNSCC patient tissues as well as their correlation with clinicopathological features. Results showed that approximately half of the tumor necrosis can be attributed to necroptosis, and the extent of necroptosis is an independent prognostic marker for patient's overall survival and progression-free survival. Then we established and thoroughly verified an in vitro model of necroptosis in two HNSCC cell lines using combined treatment of TNF-α, Smac mimetic and zVAD-fmk (TSZ). At last, we adopted this model and demonstrated that necroptosis can promote migration and invasion of HNSCC cells by releasing damage-associated molecular patterns. In conclusion, our study unveiled the necroptotic status in HNSCC for the first time and provided a novel in vitro model of necroptosis in two HNSCC cell lines. In addition, our results indicated that necroptosis may be a potential cancer promoter in HNSCC. This study may serve as the foundation for future researches of necroptosis in HNSCC.
Collapse
Affiliation(s)
- Jingyuan Li
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Sihui Huang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Lijuan Zeng
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Kan Li
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Le Yang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Siyong Gao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chenyu Guan
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Sien Zhang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaomei Lao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Guiqing Liao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| | - Yujie Liang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
60
|
Xiong Q, Liu B, Ding M, Zhou J, Yang C, Chen Y. Hypoxia and cancer related pathology. Cancer Lett 2020; 486:1-7. [PMID: 32439418 DOI: 10.1016/j.canlet.2020.05.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/18/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022]
Abstract
Hypoxic environments occur normally at high altitude, or in underground burrows and in deep sea habitats. They also occur pathologically in human ischemia and in hypoxic solid tumors. Hypoxia in various cancer types and its related molecular mechanisms are associated with a poor clinical outcome. This review will discuss how hypoxia can influence two aspects of tumorigenesis, namely the direct, cell-intrinsic oncogenic effects, as well as the indirect effects on tumor progression mediated by an altered tumor microenvironment. We will also discuss recent progress in identifying the functional roles of hypoxia-related factors (HIFs), along with their regulators and downstream target genes, in cancer stem cells and therapy. Importantly, we propose, using convergent evolution schemes to identify novel biomarkers for both hypoxia adaptation and hypoxic solid tumors as an important strategy in the future.
Collapse
Affiliation(s)
- Qiuxia Xiong
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Baiyang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingxia Ding
- Deparment of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, China
| | - Jumin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Cuiping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China.
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| |
Collapse
|
61
|
Lee S, Hu Y, Loo SK, Tan Y, Bhargava R, Lewis MT, Wang XS. Landscape analysis of adjacent gene rearrangements reveals BCL2L14-ETV6 gene fusions in more aggressive triple-negative breast cancer. Proc Natl Acad Sci U S A 2020; 117:9912-9921. [PMID: 32321829 PMCID: PMC7211963 DOI: 10.1073/pnas.1921333117] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for 10 to 20% of breast cancer, with chemotherapy as its mainstay of treatment due to lack of well-defined targets, and recent genomic sequencing studies have revealed a paucity of TNBC-specific mutations. Recurrent gene fusions comprise a class of viable genetic targets in solid tumors; however, their role in breast cancer remains underappreciated due to the complexity of genomic rearrangements in this cancer. Our interrogation of the whole-genome sequencing data for 215 breast tumors catalogued 99 recurrent gene fusions, 57% of which are cryptic adjacent gene rearrangements (AGRs). The most frequent AGRs, BCL2L14-ETV6, TTC6-MIPOL1, ESR1-CCDC170, and AKAP8-BRD4, were preferentially found in the more aggressive forms of breast cancers that lack well-defined genetic targets. Among these, BCL2L14-ETV6 was exclusively detected in TNBC, and interrogation of four independent patient cohorts detected BCL2L14-ETV6 in 4.4 to 12.2% of TNBC tumors. Interestingly, these fusion-positive tumors exhibit more aggressive histopathological features, such as gross necrosis and high tumor grade. Amid TNBC subtypes, BCL2L14-ETV6 is most frequently detected in the mesenchymal entity, accounting for ∼19% of these tumors. Ectopic expression of BCL2L14-ETV6 fusions induce distinct expression changes from wild-type ETV6 and enhance cell motility and invasiveness of TNBC and benign breast epithelial cells. Furthermore, BCL2L14-ETV6 fusions prime partial epithelial-mesenchymal transition and endow resistance to paclitaxel treatment. Together, these data reveal AGRs as a class of underexplored genetic aberrations that could be pathological in breast cancer, and identify BCL2L14-ETV6 as a recurrent gene fusion in more aggressive form of TNBC tumors.
Collapse
Affiliation(s)
- Sanghoon Lee
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232
| | - Yiheng Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Suet Kee Loo
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232
| | - Ying Tan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Rohit Bhargava
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232
| | - Michael T Lewis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Radiology, Baylor College of Medicine, Houston, TX 77030
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232;
- Women's Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15232
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
62
|
Sarode P, Schaefer MB, Grimminger F, Seeger W, Savai R. Macrophage and Tumor Cell Cross-Talk Is Fundamental for Lung Tumor Progression: We Need to Talk. Front Oncol 2020; 10:324. [PMID: 32219066 PMCID: PMC7078651 DOI: 10.3389/fonc.2020.00324] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Regardless of the promising results of certain immune checkpoint blockers, current immunotherapeutics have met a bottleneck concerning response rate, toxicity, and resistance in lung cancer patients. Accumulating evidence forecasts that the crosstalk between tumor and immune cells takes center stage in cancer development by modulating tumor malignancy, immune cell infiltration, and immune evasion in the tumor microenvironment (TME). Cytokines and chemokines secreted by this crosstalk play a major role in cancer development, progression, and therapeutic management. An increased infiltration of Tumor-associated macrophages (TAMs) was observed in most of the human cancers, including lung cancer. In this review, we emphasize the role of cytokines and chemokines in TAM-tumor cell crosstalk in the lung TME. Given the role of cytokines and chemokines in immunomodulation, we propose that TAM-derived cytokines and chemokines govern the cancer-promoting immune responses in the TME and offer a new immunotherapeutic option for lung cancer treatment.
Collapse
Affiliation(s)
- Poonam Sarode
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Martina Barbara Schaefer
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
63
|
Upadhyay Baskota S, Monaco SE, Xing J, Pantanowitz L. Peritheliomatous pattern: A diagnostic clue for diagnosing metastatic melanoma in cytology. Cancer Cytopathol 2020; 128:260-268. [PMID: 31985897 DOI: 10.1002/cncy.22241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/18/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND A peritheliomatous pattern (PP) in tumors is characterized by a sheath of viable tumor cells closely surrounding a central blood vessel. In the authors' cytology practice, such a PP has been recognized in several metastatic melanoma specimens. The aim of this study was to evaluate the frequency of a PP in cytology samples of melanoma in comparison with other tumors. METHODS Eighty archival fine-needle aspiration (FNA) cases of metastatic melanoma were compared with 65 control cases (35 poorly differentiated/metastatic carcinomas, 15 lymphomas, and 15 recurrent/metastatic/high-grade sarcomas). Cytologic findings were correlated with corresponding histologic specimens, which were available for 44 cases (55%) in the melanoma group and for 23 cases (35.38%) in the control group. All slides were examined for a PP and were semiquantitatively graded for comparison. RESULTS A PP was present in 51.3% of the cytologic preparations (n = 41) among the melanoma group cases, whereas in the control group, a PP was present in only 3.1% of the cases (n = 2). A PP was present in 65.9% of melanomas with available histologic sections (n = 29) and in 8.7% of tissue samples from the control group (n = 2). A PP was seen more often in cell blocks than direct smear preparations (51.3% vs 40.0%) from patients with melanoma. CONCLUSIONS The presence of a characteristic PP can be helpful in diagnosing melanoma in FNA samples because it was present in almost half of the metastatic melanoma cases in this study and was rarely present in other tumor types. A PP is easier to recognize and more often presents in cell blocks than aspirate smears. Ancillary studies such as immunohistochemistry are helpful for excluding other entities that may exhibit a PP.
Collapse
Affiliation(s)
| | - Sara E Monaco
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Juan Xing
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Liron Pantanowitz
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
64
|
Burgess B, Levine B, Taylor RN, Kelly MG. Preoperative Circulating Lymphocyte and Monocyte Counts Correlate with Patient Outcomes in Type I and Type II Endometrial Cancer. Reprod Sci 2020; 27:194-203. [PMID: 32046381 PMCID: PMC11758426 DOI: 10.1007/s43032-019-00009-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/25/2019] [Indexed: 12/23/2022]
Abstract
Tumor-associated macrophages and tumor-infiltrating lymphocytes are associated with survival in solid malignancies. Given the physiological link to peripheral immune cell counts, we evaluated if peripheral immune cell counts were predictors of outcomes in endometrial cancer. A retrospective study was completed for endometrial cancer cases between 2000 and 2010. Kaplan-Meier, bivariate, and multivariable Cox proportion hazard analyses were performed examining the relations between survival and peripheral immune cell counts. Three hundred ten patients were identified. In bivariate analyses, high monocyte counts (> 0.7 × 109 cells/L) trended with decreased progression free survival (PFS) (p = 0.10) and poorer overall survival (OS) (p = 0.16). By contrast, high lymphocyte level (> 1.5 × 109 cells/L) was associated with improved PFS (p = 0.008) and OS (p = 0.006). These findings were consistent for type I and type II endometrial cancers. In a multivariable Cox model, high monocyte level was associated with a greater risk of disease recurrence (hazard ratio (HR) = 1.63, p < 0.035). Other significant predictors of recurrence were age, non-endometrioid histology, and the presence of lymph vascular space invasion (LVSI). In a multivariable Cox model, high lymphocyte count trended with a lower risk of death (HR = 0.66, p = 0.07). Age, surgical stage, non-endometrioid histology, and LVSI were also associated with death in this model. In this sample of endometrial cancer patients, we found that high preoperative lymphocyte counts were associated with improved overall improved survival. High monocyte counts were associated with poorer disease-free survival outcomes. Further studies that focused on understanding tumor-antagonizing and pro-tumoral effects of lymphocytes and monocytes, respectively, in endometrial cancer are recommended.
Collapse
Affiliation(s)
- Brian Burgess
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Beverly Levine
- Department of Social Sciences and Health Policy, Wake Forest University Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Michael G Kelly
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
65
|
Yun SG, An YY, Kim SH, Kang BJ. Early Recurrence of Breast Cancer after the Primary Treatment: Analysis of Clinicopathological and Radiological Predictive Factors. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2020; 81:395-408. [PMID: 36237380 PMCID: PMC9431821 DOI: 10.3348/jksr.2020.81.2.395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 12/09/2022]
Abstract
목적 유방암 일차치료 후 조기 재발 예측에 도움이 되는 임상병리학적 인자 및 원발암의 영상 의학적 특징을 알아보고자 하였다. 대상과 방법 본원에서 2010년 1월부터 2014년 12월까지 유방 보존 수술 및 보조요법 치료 후 추적관찰을 받은 480명을 대상으로 하였다. 일차치료 완결 후 3년 이내 재발한 경우를 조기 재발으로 정의하고, 이와 관련된 임상병리학적 및 영상의학적 예측인자를 알아보기 위해 단변량 및 다변량 로지스틱 회귀분석을 시행하였다. 결과 단변량 분석에서 임상병리학적 인자 중 높은 종양 병기(p=0.021), 고등급 조직학적 분화도(p < 0.001), estrogen receptor 수용체 음성(p=0.002), Ki-67 과발현(p=0.017), 삼중음성유방 아형(p=0.019)과, 영상 소견 중 유방암의 다초점성(p < 0.001), 도플러 검사에서 테두리 혈류(p=0.012), 자기공명영상에서 테두리 조영증강(p < 0.001)이 조기 재발과 유의한 연관성을 보였다. 다변량 분석에서 원발암의 높은 종양 병기[odds ratio (이하 OR) = 3.47, 95% confidence interval (이하 CI) 1.12~10.73, p=0.031]와 도플러 검사에서 테두리 혈류 (OR = 3.32, 95% CI 1.38~8.02,p=0.008)가 조기 재발과 관련된 독립적인 예측인자였다. 결론 유방암 치료 전 도플러 검사에서 원발암의 테두리 혈류 소견은 유방암의 일차치료 후 조기 재발의 독립적인 영상의학적 예측인자이다.
Collapse
Affiliation(s)
- Sun Geun Yun
- Department of Radiology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Yeong Yi An
- Department of Radiology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Sung Hun Kim
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bong Joo Kang
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
66
|
Atanasov G, Dino K, Schierle K, Dietel C, Aust G, Pratschke J, Seehofer D, Schmelzle M, Hau HM. Angiogenic inflammation and formation of necrosis in the tumor microenvironment influence patient survival after radical surgery for de novo hepatocellular carcinoma in non-cirrhosis. World J Surg Oncol 2019; 17:217. [PMID: 31830991 PMCID: PMC6909650 DOI: 10.1186/s12957-019-1756-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022] Open
Abstract
Background Tumor escape mechanisms mediated in the tumor microenvironment can significantly reduce the capacity of the anti-tumor function of the immune system. TIE2-expressing monocytes (TEMs), related angiopoietins, and tumor necrosis are considered to have a key role in this process. We aimed to investigate the abundance and clinical significance of these biomarkers in hepatocellular carcinoma (HCC). Methods In this retrospective study, 58 HCC patients received surgery with a curative intent. The abundance of TEMs, angiopoietin-1 and -2 were detected in tumor specimens of the HCC patients (n = 58), and together with the occurrence of histologic tumor necrosis, were associated with established clinicopathological characteristics and survival. Results Patients with HCC characterized by necrosis and TEMs revealed reduced both overall survival and recurrence-free survival (all p < 0.05). Angiopoietins and TEMs were associated with metastatic and recurrent HCC. Furthermore, the formation of histologic tumor necrosis was associated with advanced tumor stage and density of TEMs (all p < 0.05). Conclusions Histologic tumor necrosis, TEMs, and related angiopoietins were associated with multiple HCC parameters and patient survival. The tumor necrosis–TEM–angiopoietin axis may offer a novel diagnostic modality to predict patient outcome after surgery for HCC.
Collapse
Affiliation(s)
- Georgi Atanasov
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany. .,Department of Surgery, Campus Charité Mitte und Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Berlin Institute of Health, Berlin, Germany.
| | - Karoline Dino
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Katrin Schierle
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Corinna Dietel
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Gabriela Aust
- Department of Surgery, Research Laboratories, University of Leipzig, Leipzig, Germany
| | - Johann Pratschke
- Department of Surgery, Campus Charité Mitte und Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Daniel Seehofer
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Moritz Schmelzle
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany.,Department of Surgery, Campus Charité Mitte und Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Hans-Michael Hau
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
67
|
Reduced RhoA expression enhances breast cancer metastasis with a concomitant increase in CCR5 and CXCR4 chemokines signaling. Sci Rep 2019; 9:16351. [PMID: 31705019 PMCID: PMC6841971 DOI: 10.1038/s41598-019-52746-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/22/2019] [Indexed: 12/31/2022] Open
Abstract
The role of RhoA GTPases in breast cancer tumorigenesis and metastasis is unclear. Early studies within which mutations in RhoA were designed based on cancer-associated mutations in Ras supported an oncogene role for RhoA. However, recent whole-genome sequencing studies of cancers raised the possibility that RhoA may have a tumor suppression function. Here, using a syngeneic triple negative breast cancer murine model we investigated the physiological effects of reduced RhoA expression on breast cancer tumorigenesis and metastasis. RhoA knockdown had no effect on primary tumor formation and tumor proliferation, concurring with our in vitro findings where reduced RhoA had no effect on breast cancer cell proliferation and clonogenic growth. In contrast, primary tumors with RhoA knockdown efficiently invaded sentinel lymph nodes and significantly metastasized to lungs compared to control tumors. Mechanistically, the current study demonstrated that this is achieved by promoting a pro-tumor microenvironment, with increased cancer-associated fibroblasts and macrophage infiltration, and by modulating the CCL5-CCR5 and CXCL12-CXCR4 chemokine axes in the primary tumor. To our knowledge, this is the first such mechanistic study in breast cancer showing the ability of RhoA to suppress chemokine receptor expression in breast tumor cells. Our work suggests a physiological lung and lymph node metastasis suppressor role for RhoA GTPase in breast cancer.
Collapse
|
68
|
Proctor DT, Huang J, Lama S, Albakr A, Van Marle G, Sutherland GR. Tumor-associated macrophage infiltration in meningioma. Neurooncol Adv 2019; 1:vdz018. [PMID: 32642654 PMCID: PMC7212927 DOI: 10.1093/noajnl/vdz018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Meningioma, a most common brain tumor, has a high rate of recurrence. Tumor-associated macrophages (TAMs) are the most abundant immune cell type in meningioma. TAMs display functional phenotypic diversity and may establish either an inflammatory and anti-tumoral or an immunosuppressive and pro-tumoral microenvironment. TAM subtypes present in meningioma and potential contribution to growth and recurrence is unknown. Methods Immunofluorescence staining was used to quantify M1 and M2 TAM populations in tissues obtained from 30 meningioma patients. Associations between M1 and M2 cells, M1:M2 cell ratio to tumor characteristics, WHO grade, recurrence, size, location, peri-tumoral edema, and patient demographics such as age and sex were examined. Results TAM cells accounted for ~18% of all cells in meningioma tissues. More than 80% of infiltrating TAMs were found to be of pro-tumoral M2 phenotype and correlated to tumor size (P = .0409). M1:M2 cell ratio was significantly decreased in WHO grade II, compared to grade I tumors (P = .009). Furthermore, a 2.3-fold difference in M1:M2 ratio between primary (0.14) and recurrent (0.06) tumors was observed (n = 18 and 12 respectively, P = .044). Conclusion This study is the first to confirm existence of pro-tumoral M2 TAMs in the meningioma microenvironment, emphasizing its potential role in tumor growth and recurrence.
Collapse
Affiliation(s)
- Dustin T Proctor
- Project neuroArm, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jordan Huang
- Project neuroArm, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sanju Lama
- Project neuroArm, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Abdulrahman Albakr
- Project neuroArm, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neurosurgery, King Saud University, Riyadh, Saudi Arabia
| | - Guido Van Marle
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Garnette R Sutherland
- Project neuroArm, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
69
|
La X, Zhang L, Yang Y, Li H, Song G, Li Z. Tumor-secreted GRP78 facilitates the migration of macrophages into tumors by promoting cytoskeleton remodeling. Cell Signal 2019; 60:1-16. [PMID: 30959099 DOI: 10.1016/j.cellsig.2019.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 11/25/2022]
Abstract
Glucose-regulated protein 78 (GRP78), an important molecular chaperone in the endoplasmic reticulum, is often over-expressed in the central region of advanced tumor and acts as a promoter of tumor progression. As main immune cells in the tumor microenvironment, infiltration of abundant macrophages into advanced tumor further facilitates growth of tumor. Although has potential association between GRP78 and infiltration of macrophages, its underlying mechanisms are poorly understood. Here, we report that secreted GRP78 facilitates recruitment of macrophages into tumors both in vitro and in vivo. Further studies reveal that secreted GRP78 transports into macrophages and bound to intracellular Ca2+, which lead to uneven distribution of Ca2+ and subsequent polarization of macrophages. The polarization of macrophages activates expression of microRNA-200b-3p. By directly targeting RhoGDI, miR-200b-3p stimulates the activity of RhoGTPase and ultimately leads to the distribution of GTP-Rac1 and GTP-Cdc42 in front protrusion and GTP-RhoA in rear contraction, which further results in migration of macrophages in a certain direction. Our results reveal a novel function of GRP78 to promote the recruitment of macrophages to tumor and provide a potential therapeutic target for malignancies.
Collapse
Affiliation(s)
- Xiaoqin La
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Lichao Zhang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Yufei Yang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Hanqing Li
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Guisheng Song
- Department of Medicine, University of Minnesota, MN 55455, USA.
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China; Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; School of Life Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
70
|
You C, Li J, Zhi W, Chen Y, Yang W, Gu Y, Peng W. The volumetric-tumour histogram-based analysis of intravoxel incoherent motion and non-Gaussian diffusion MRI: association with prognostic factors in HER2-positive breast cancer. J Transl Med 2019; 17:182. [PMID: 31262334 PMCID: PMC6604303 DOI: 10.1186/s12967-019-1911-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022] Open
Abstract
Background To evaluate the imaging biomarkers of human epidermal growth factor receptor 2 (HER2) positive breast cancer in comparison to other molecular subtypes and to determine the feasibility of identifying hormone receptor (HR) status and lymph node metastasis status using volumetric-tumour histogram-based analysis through intravoxel incoherent motion (IVIM) and non-Gaussian diffusion. Methods This study included 145 breast cancer patients with 148 lesions between January and November in 2018. Among the 148 lesions, 74 were confirmed to be HER2-positive. The volumetric-tumour histogram-based features were extracted from the combined IVIM and non-Gaussian diffusion model. IVIM and non-Gaussian diffusion parameters obtained from images of the subjects with different molecular prognostic biomarker statuses were compared by Student’s t test or the Mann–Whitney U test. The area under the curve (AUC), sensitivity, and specificity at the best cut-off point were reported. The Spearman correlation coefficient was calculated to analyse the correlations of clinical tumor nodule metastasis (TNM) stage and Ki67 with the IVIM and non-Gaussian diffusion parameters. Results The entropy of mean kurtosis (MK) was significantly higher in the HER2-positive group than in the HER2-negative group (p = 0.015), with an AUC of 0.629 (95% CI 0.546, 0.707), a sensitivity of 62.6%, and a specificity of 66.2%. For HR status, the MD 5th percentile was higher in the HR-positive group of HER2-positive breast cancer (p = 0.041), with an AUC of 0.643 (95% CI 0.523, 0.751), while for lymph node status, the entropy of mean diffusivity (MK) was lower in the lymph node positive group (p = 0.040), with an AUC of 0.587 (95% CI 0.504, 0.668). The clinical TNM stage and Ki67 index were correlated with several histogram parameters. Conclusion Volumetric-lesion histogram analysis of IVIM and the non-Gaussian diffusion model can be used to provide prognostic information about HER2-positive breast cancers and potentially contribute to individualized anti-HER2 targeted therapy plans . Electronic supplementary material The online version of this article (10.1186/s12967-019-1911-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chao You
- Department of Radiology, Fudan University Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dongan Road, Shanghai, 200032, People's Republic of China
| | - Jianwei Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Wenxiang Zhi
- Department of Ultrasound, Fudan University Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yanqiong Chen
- Department of Radiology, Fudan University Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dongan Road, Shanghai, 200032, People's Republic of China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University Shanghai, Shanghai, People's Republic of China
| | - Yajia Gu
- Department of Radiology, Fudan University Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dongan Road, Shanghai, 200032, People's Republic of China.
| | - Weijun Peng
- Department of Radiology, Fudan University Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dongan Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
71
|
Muhammad T, Sakhawat A, Khan AA, Ma L, Gjerset RA, Huang Y. Mesenchymal stem cell-mediated delivery of therapeutic adenoviral vectors to prostate cancer. Stem Cell Res Ther 2019; 10:190. [PMID: 31238944 PMCID: PMC6593580 DOI: 10.1186/s13287-019-1268-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/29/2019] [Accepted: 05/15/2019] [Indexed: 01/04/2023] Open
Abstract
Background There is an urgent need for targeted biological therapies for prostate cancer with greater efficacy and less toxicity, particularly for metastatic disease, where current therapies are not curative. Therapeutic adenoviral vectors or oncolytic adenoviruses offer the possibility of a competent, nontoxic therapeutic alternative for prostate cancer. However, free viral particles must be delivered locally, an approach that does not address metastatic disease, and they display poor tumor penetration. To fully exploit the potential of these vectors, we must develop methods that improve intratumoral dissemination and allow for systemic delivery. This study establishes a proof-of-principle rationale for a novel human mesenchymal stem (stromal) cell-based approach to improving vector delivery to tumors. Methods/results We have generated mesenchymal stem cell-derived packaging cells for adenoviruses (E1-modified mesenchymal stem cells) by modifying human mesenchymal stem cells with the adenovirus (type C) E1A/B genes needed for viral replication. Using cell-based assays, we have demonstrated that two adenoviral vectors, replication-defective adenovirus expressing p14 and p53 or conditionally replicating oncolytic adenovirus, packaged by E1A/B-modified mesenchymal stem cells, suppress the growth of prostate cancer cells in culture. Using subcutaneous xenograft models for human prostate cancer in mice, we have shown that E1A/B-modified mesenchymal stem cells display tumor tropism in tumor-bearing nude mice, that E1A/B-modified mesenchymal stem cells disseminate well within tumors, and that replication-defective adenovirus expressing p14 and p53 or conditionally replicating oncolytic adenovirus-loaded E1-modified mesenchymal stem cells suppresses tumor growth in mice. Conclusion The results show that this approach, if optimized, could circumvent the obstacles to efficient gene delivery encountered with current gene delivery approaches and provide an effective, nontoxic therapeutic alternative for metastatic disease.
Collapse
Affiliation(s)
- Tahir Muhammad
- College of life sciences and Bio-engineering, Beijing University of Technology, Beijing, China
| | - Ali Sakhawat
- College of life sciences and Bio-engineering, Beijing University of Technology, Beijing, China
| | - Aamir Ali Khan
- College of life sciences and Bio-engineering, Beijing University of Technology, Beijing, China
| | - Ling Ma
- College of life sciences and Bio-engineering, Beijing University of Technology, Beijing, China
| | - Ruth A Gjerset
- Torrey Pines Institute for Molecular Studies, San Diego, CA, USA
| | - Yinghui Huang
- College of life sciences and Bio-engineering, Beijing University of Technology, Beijing, China.
| |
Collapse
|
72
|
Spinelli FM, Vitale DL, Icardi A, Caon I, Brandone A, Giannoni P, Saturno V, Passi A, García M, Sevic I, Alaniz L. Hyaluronan preconditioning of monocytes/macrophages affects their angiogenic behavior and regulation of TSG-6 expression in a tumor type-specific manner. FEBS J 2019; 286:3433-3449. [PMID: 31044513 DOI: 10.1111/febs.14871] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 03/18/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022]
Abstract
Hyaluronan is a glycosaminoglycan normally present in the extracellular matrix in most tissues. Hyaluronan is a crucial player in many processes associated with cancer, such as angiogenesis, invasion, and metastasis. However, little has been reported regarding the action of hyaluronan on monocytes/macrophages (Mo/MØ) in tumor angiogenesis and its consequences on tumor development. In the present study, we investigated the effects of hyaluronan of different sizes on human Mo/MØ angiogenic behavior in colorectal and breast carcinoma. In vitro, the treatment of Mo/MØ with lysates and conditioned media from a breast but not from colorectal carcinoma cell line plus high-molecular weight hyaluronan induced: (a) an increased expression of angiogenic factors VEGF, IL-8, FGF-2, and MMP-2, (b) an increased endothelial cell migration, and (c) a differential expression of hyaluronan-binding protein TSG-6. Similar results were observed in Mo/MØ derived from breast cancer patients treated with tumor lysates. Besides, macrophages primed with high-molecular weight hyaluronan and inoculated in human breast cancer xenograft tumor increased blood vessel formation and diminished TSG-6 levels. In contrast, the effects triggered by high-molecular weight hyaluronan on Mo/MØ in breast cancer context were not observed in the context of colorectal carcinoma. Taken together, these results indicate that the effect of high-molecular weight hyaluronan as an inductor of the angiogenic behavior of macrophages in breast tumor context is in part consequence of the presence of TSG-6.
Collapse
Affiliation(s)
- Fiorella M Spinelli
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional de la Pcia. de Bs. As. Centro de Investigaciones y Transferencia del Noroeste de la Pcia. de Bs. As. (CIT NOBA, UNNOBA-CONICET), Junín, Argentina
| | - Daiana L Vitale
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional de la Pcia. de Bs. As. Centro de Investigaciones y Transferencia del Noroeste de la Pcia. de Bs. As. (CIT NOBA, UNNOBA-CONICET), Junín, Argentina
| | - Antonella Icardi
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional de la Pcia. de Bs. As. Centro de Investigaciones y Transferencia del Noroeste de la Pcia. de Bs. As. (CIT NOBA, UNNOBA-CONICET), Junín, Argentina
| | - Ilaria Caon
- Dipartimento di Medicina e Chirurgia, Università degli Studi dell'Insubria, Varese, Italia
| | - Alejandra Brandone
- Hospital Interzonal General de Agudos Dr. Abraham F. Piñeyro, Junín, Argentina
| | | | - Virginia Saturno
- Hospital Interzonal General de Agudos Dr. Abraham F. Piñeyro, Junín, Argentina
| | - Alberto Passi
- Dipartimento di Medicina e Chirurgia, Università degli Studi dell'Insubria, Varese, Italia
| | - Mariana García
- Laboratorio de Terapia Génica, IIMT - CONICET, Universidad Austral, Derqui-Pilar, Argentina
| | - Ina Sevic
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional de la Pcia. de Bs. As. Centro de Investigaciones y Transferencia del Noroeste de la Pcia. de Bs. As. (CIT NOBA, UNNOBA-CONICET), Junín, Argentina
| | - Laura Alaniz
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional de la Pcia. de Bs. As. Centro de Investigaciones y Transferencia del Noroeste de la Pcia. de Bs. As. (CIT NOBA, UNNOBA-CONICET), Junín, Argentina
| |
Collapse
|
73
|
Zhang X, Ashcraft KA, Betof Warner A, Nair SK, Dewhirst MW. Can Exercise-Induced Modulation of the Tumor Physiologic Microenvironment Improve Antitumor Immunity? Cancer Res 2019; 79:2447-2456. [PMID: 31068341 DOI: 10.1158/0008-5472.can-18-2468] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/10/2019] [Accepted: 03/05/2019] [Indexed: 12/12/2022]
Abstract
The immune system plays an important role in controlling cancer growth. However, cancers evolve to evade immune detection. Immune tolerance and active immune suppression results in unchecked cancer growth and progression. A major contributor to immune tolerance is the tumor physiologic microenvironment, which includes hypoxia, hypoglucosis, lactosis, and reduced pH. Preclinical and human studies suggest that exercise elicits mobilization of leukocytes into circulation (also known as "exercise-induced leukocytosis"), especially cytotoxic T cells and natural killer cells. However, the tumor physiologic microenvironment presents a significant barrier for these cells to enter the tumor and, once there, properly function. We hypothesize that the effect of exercise on the immune system's ability to control cancer growth is linked to how exercise affects the tumor physiologic microenvironment. Normalization of the microenvironment by exercise may promote more efficient innate and adaptive immunity within the tumor. This review summarizes the current literature supporting this hypothesis.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Duke University Medical Center, Durham, North Carolina
| | | | | | - Smita K Nair
- Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
74
|
Poor outcome in hypoxic endometrial carcinoma is related to vascular density. Br J Cancer 2019; 120:1037-1044. [PMID: 31011231 PMCID: PMC6738053 DOI: 10.1038/s41416-019-0461-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 01/05/2023] Open
Abstract
Background Identification of endometrial carcinoma (EC) patients at high risk of recurrence is lacking. In this study, the prognostic role of hypoxia and angiogenesis was investigated in EC patients. Methods Tumour slides from EC patients were stained by immunofluorescence for carbonic anhydrase IX (CAIX) as hypoxic marker and CD34 for assessment of microvessel density (MVD). CAIX expression was determined in epithelial tumour cells, with a cut-off of 1%. MVD was assessed according to the Weidner method. Correlations with disease-specific survival (DSS), disease-free survival (DFS) and distant disease-free survival (DDFS) were calculated using Kaplan–Meier curves and Cox regression analysis. Results Sixty-three (16.4%) of 385 ECs showed positive CAIX expression with high vascular density. These ECs had a reduced DSS compared to tumours with either hypoxia or high vascular density (log-rank p = 0.002). Multivariable analysis showed that hypoxic tumours with high vascular density had a reduced DSS (hazard ratio [HR] 3.71, p = 0.002), DDFS (HR 2.68, p = 0.009) and a trend for reduced DFS (HR 1.87, p = 0.054). Conclusions This study has shown that adverse outcome in hypoxic ECs is seen in the presence of high vascular density, suggesting an important role of angiogenesis in the metastatic process of hypoxic EC. Differential adjuvant treatment might be indicated for these patients.
Collapse
|
75
|
Hugdahl E, Bachmann IM, Schuster C, Ladstein RG, Akslen LA. Prognostic value of uPAR expression and angiogenesis in primary and metastatic melanoma. PLoS One 2019; 14:e0210399. [PMID: 30640942 PMCID: PMC6331131 DOI: 10.1371/journal.pone.0210399] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/21/2018] [Indexed: 11/18/2022] Open
Abstract
Angiogenesis is important for the progression of cutaneous melanoma. Here, we analyzed the prognostic impact of the angiogenic factor urokinase plasminogen activator resecptor (uPAR), vascular proliferation index (VPI) and tumor necrosis as a measure of hypoxia in a patient series of nodular melanomas (n = 255) and matched loco-regional metastases (n = 78). Expression of uPAR was determined by immunohistochemistry and VPI was assessed by dual immunohistochemistry using Factor-VIII/Ki67 staining. Necrosis was recorded based on HE-slides. As novel findings, high uPAR expression and high VPI were associated with each other, and with increased tumor thickness, presence of tumor necrosis, tumor ulceration, increased mitotic count and reduced cancer specific survival in primary melanoma. In matched cases, VPI was decreased in metastases, whereas the frequency of necrosis was increased. Our findings demonstrate for the first time the impact on melanoma specific survival of uPAR expression and VPI in primary tumors, and of increased necrosis as an indicator of tumor hypoxia in loco-regional metastases. These findings support the importance of tumor angiogenesis in melanoma aggressiveness, and suggest uPAR as an indicator of vascular proliferation and a potential biomarker in melanoma.
Collapse
Affiliation(s)
- Emilia Hugdahl
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Dermatology, Haukeland University Hospital, Bergen, Norway
| | - Ingeborg M. Bachmann
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Dermatology, Haukeland University Hospital, Bergen, Norway
| | - Cornelia Schuster
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology Haukeland University Hospital, Bergen, Norway
| | - Rita G. Ladstein
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Dermatology, Haukeland University Hospital, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| |
Collapse
|
76
|
Abstract
Background Inflammation is a major player in breast cancer (BC) progression. Allograft-inflammatory factor-1 (AIF1) is a crucial mediator in the inflammatory response. AIF1 reportedly plays a role in BC, but the mechanism remains to be elucidated. We identified two AIF1 isoforms, AIF1v1 and AIF1v3, which were differentially expressed between affected and unaffected sisters from families with high risk of BC with no deleterious BRCA1/BRCA2 mutations (BRCAX). We investigated potential functions of AIFv1/v3 in BC of varying severity and breast adipose tissue by evaluating their expression, and association with metabolic and clinical parameters of BC patients. Methods AIF1v1/v3 expression was determined in BC tissues and cell lines using quantitative real-time PCR. Potential roles and mechanisms were examined in the microenvironment (fibroblasts, adipose tissue, monocytes and macrophages), inflammatory response (cell reaction in BC subgroups), and metabolism [treatment with docosahexaenoic acid (DHA)]. Association of AIF1 transcript expression with clinical factors was determined by Spearman’s rank correlation. Bioinformatics analyses were performed to characterize transcripts. Results AIF1v1/v3 were mostly expressed in the less severe BC samples, and their expression appeared to originate from the tumor microenvironment. AIF1 isoforms had different expression rates and sources in breast adipose tissue; lymphocytes mostly expressed AIF1v1 while activated macrophages mainly expressed AIF1v3. Bioinformatics analysis revealed major structural differences suggesting distinct functions in BC progression. Lymphocytes were the most infiltrating cells in breast tumors and their number correlated with AIF1v1 adipose expression. Furthermore, DHA supplementation significantly lowered the expression of AIF1 isoforms in BRCAX cell lines. Finally, the expression of AIF1 isoforms in BC and breast adipose tissue correlated with clinical parameters of BC patients. Conclusions Results strongly suggest that AIF1v1 as much as AIF1v3 play a major role in the crosstalk between BC and infiltrating immune cells mediating tumor progression, implying their high potential as target molecules for BC diagnostic, prognostication and treatment. Electronic supplementary material The online version of this article (10.1186/s12935-018-0663-3) contains supplementary material, which is available to authorized users.
Collapse
|
77
|
Choi BB, Lee JS, Kim KH. Association between MRI Features and Standardized Uptake Value of 18F-FDG PET/CT in Triple-Negative Breast Cancer. Oncol Res Treat 2018; 41:706-711. [PMID: 30321870 DOI: 10.1159/000492341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/23/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a breast cancer subgroup that lacks the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Certain magnetic resonance imaging (MRI) features of TNBC might reflect the poor prognosis of TNBC. Standardized uptake value (SUV) of [18F]fluorodeoxyglucose positron emission tomography/computed tomography is 1 of predictive prognostic factors for breast cancer. The purpose of this study was to correlate MRI features of TNBC with SUVmax to determine whether MRI findings in TNBC could be helpful for predicting prognosis. METHODS We analyzed MRI findings of TNBC according to breast imaging reporting and data system (BI-RADS) MRI lexicon. We also assessed intratumoral high signal intensity on T2-weighted MRI, time-intensity curve analysis and peritumoral edema. The relationship between MRI features of TNBC and SUVmax was then statistically analyzed. RESULTS Significant correlations of SUVmax with the internal enhancement pattern, intratumoral high signal intensity on T2-weighted images, and peritumoral edema were noted. There was no significant correlation of SUVmax with mass shape, margin, or kinetics. CONCLUSION Certain MRI features of TNBC such as heterogeneous or rim enhancement, intratumoral very high signal intensity on T2 images, and peritumoral edema might be useful in predicting the prognosis of patients with TNBC.
Collapse
|
78
|
Medler T, Patel JM, Alice A, Baird JR, Hu HM, Gough MJ. Activating the Nucleic Acid-Sensing Machinery for Anticancer Immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 344:173-214. [PMID: 30798988 PMCID: PMC6754183 DOI: 10.1016/bs.ircmb.2018.08.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nucleic acid sensing pathways have likely evolved as part of a broad pathogen sensing strategy intended to discriminate infectious agents and initiate appropriate innate and adaptive controls. However, in the absence of infectious agents, nucleic acid sensing pathways have been shown to play positive and negative roles in regulating tumorigenesis, tumor progression and metastatic spread. Understanding the normal biology behind these pathways and how they are regulated in malignant cells and in the tumor immune environment can help us devise strategies to exploit nucleic acid sensing to manipulate anti-cancer immunity.
Collapse
Affiliation(s)
- Terry Medler
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Jaina M Patel
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Jason R Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Hong-Ming Hu
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States.
| |
Collapse
|
79
|
Serganova I, Cohen IJ, Vemuri K, Shindo M, Maeda M, Mane M, Moroz E, Khanin R, Satagopan J, Koutcher JA, Blasberg R. LDH-A regulates the tumor microenvironment via HIF-signaling and modulates the immune response. PLoS One 2018; 13:e0203965. [PMID: 30248111 PMCID: PMC6153000 DOI: 10.1371/journal.pone.0203965] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/30/2018] [Indexed: 01/01/2023] Open
Abstract
Previous studies show that LDH-A knockdown reduces orthotopic 4T1 breast tumor lactate and delays tumor growth and the development of metastases in nude mice. Here, we report significant changes in the tumor microenvironment (TME) and a more robust anti-tumor response in immune competent BALB/c mice. 4T1 murine breast cancer cells were transfected with shRNA plasmids directed against LDH-A (KD) or a scrambled control plasmid (NC). Cells were also transduced with dual luciferase-based reporter systems to monitor HIF-1 activity and the development of metastases by bioluminescence imaging, using HRE-sensitive and constitutive promoters, respectively. The growth and metastatic profile of orthotopic 4T1 tumors developed from these cell lines were compared and a primary tumor resection model was studied to simulate the clinical management of breast cancer. Primary tumor growth, metastasis formation and TME phenotype were significantly different in LDH-A KD tumors compared with controls. In LDH-A KD cells, HIF-1 activity, hexokinase 1 and 2 expression and VEGF secretion were reduced. Differences in the TME included lower HIF-1α expression that correlated with lower vascularity and pimonidazole staining, higher infiltration of CD3+ and CD4+ T cells and less infiltration of TAMs. These changes resulted in a greater delay in metastases formation and 40% long-term survivors (>20 weeks) in the LDH-A KD cohort following surgical resection of the primary tumor. We show for the first time that LDH-depletion inhibits the formation of metastases and prolongs survival of mice through changes in tumor microenvironment that modulate the immune response. We attribute these effects to diminished HIF-1 activity, vascularization, necrosis formation and immune suppression in immune competent animals. Gene-expression analyses from four human breast cancer datasets are consistent with these results, and further demonstrate the link between glycolysis and immune suppression in breast cancer.
Collapse
Affiliation(s)
- Inna Serganova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Ivan J. Cohen
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Kiranmayi Vemuri
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Masahiro Shindo
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Masatomo Maeda
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Mayuresh Mane
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Ekaterina Moroz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Raya Khanin
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jaya Satagopan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jason A. Koutcher
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Ronald Blasberg
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
80
|
Association among T2 signal intensity, necrosis, ADC and Ki-67 in estrogen receptor-positive and HER2-negative invasive ductal carcinoma. Magn Reson Imaging 2018; 54:176-182. [PMID: 30172938 DOI: 10.1016/j.mri.2018.08.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/13/2018] [Accepted: 08/27/2018] [Indexed: 12/23/2022]
Abstract
PURPOSE To determine whether T2 signal intensity, necrosis, and ADC values are associated with Ki-67 in patients with Estrogen Receptor (ER)-positive and Human epidermal growth factor receptor type 2 (HER2)-negative invasive ductal carcinoma (IDC). MATERIALS AND METHODS Between March 2012 and February 2013, one hundred eighty seven women with ER-positive and HER2-negative IDC who underwent breast MRI and subsequent surgery were included. Intratumoral signal intensity was evaluated based on a combination of T2-weighted (low or equal, high, or very high) and contrast-enhanced MR images (enhancement or not). Necrosis was defined as very high T2 and no enhancement. Using the analysis of variance and pairwise t-test, a model based on intratumoral signal intensity was developed to assess Ki-67 of the surgical specimen. Inter-observer agreement for the developed model was analyzed. Conventional mean and minimum apparent diffusion coefficient (ADC) measurements were performed and correlated with Ki-67. RESULTS As the grade of the developed model increased (Grade I: low or equal T2, Grade II: high T2, or necrosis < 50%, Grade III: necrosis ≥ 50%), mean Ki-67 significantly increased (Grade I to III: 12.5%, 17.6%, 45.0%, respectively; P < 0.001). Good inter-observer agreement was found for the model (κ = 0.846, P < 0.001). ADC did not show significant correlations with Ki-67 (Pearson's correlation coefficient, 0.140 [P = 0.057] for mean ADC; -0.079 [P = 0.284] for minimum ADC). CONCLUSION Intratumoral signal intensity but not ADC was associated with Ki-67 in patients with ER-positive and HER2-negative IDC.
Collapse
|
81
|
Mehrabi M, Amini F, Mehrabi S. Active Role of the Necrotic Zone in Desensitization of Hypoxic Macrophages and Regulation of CSC-Fate: A hypothesis. Front Oncol 2018; 8:235. [PMID: 29988496 PMCID: PMC6026632 DOI: 10.3389/fonc.2018.00235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/11/2018] [Indexed: 01/30/2023] Open
Abstract
Fast-proliferating cancer cells in the hypoxic region face a shortage of oxygen and nutrients, undergo necrotic cell death, and release numerous signaling components. Hypoxia-induced chemo-attractants signal for macrophages/monocytes to clear debris and return the system to steady state. Accordingly, macrophages arrange into pre-necrotic positions, where they are continuously exposed to stress signals. It can thus be hypothesized that gradual alteration of gene expression in macrophages eventually turns off their phagocytic machinery. Uncleared cell corpses within the hypoxic region potentially provide a rich source of building blocks for anaerobic metabolism of cancer stem cells via macropinocytosis, and are conceivably implicated in tumor progression and invasion.
Collapse
Affiliation(s)
| | - Fatemeh Amini
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| | - Shima Mehrabi
- Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
82
|
Bcl6/p53 expression, macrophages/mast cells infiltration and microvascular density in invasive breast carcinoma. Oncotarget 2018; 9:22727-22740. [PMID: 29854311 PMCID: PMC5978261 DOI: 10.18632/oncotarget.25220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/24/2018] [Indexed: 12/17/2022] Open
Abstract
To better understand the breast cancer progression and therapeutic resistance is crucial deepen the molecular mechanisms related to regulation of cells behavior in the tumor microenvironment. Inappropriate expression or activation of transcription factors in tumor breast microenvironment can lead to the malignant behavior of breast cancer cells. Bcl6 is a transcriptional factor that may play a role in the pathogenesis of breast cancer. Moreover, cells surrounding tumor cells, including macrophages and mast cells play an important role during tumor progression enhancing angiogenesis. We have demonstrated: 1) An increase of the BCL6 translocation and Bcl6 positive cells in G3 degree of disease; 2) A reduction of the expression of p53 in G3 breast cancer samples as compared to G1/G2 specimens; 3) Macrophages CD68+ and CD163+ in interstitial and periglandular position, increase in G3 specimens as compared to G1/G2 and control samples; 4) Tryptase-positive mast cells in periglandular position are more numerous in G3 tumor specimens as compared to G1/G2 and control samples. Overall, these data confirm the important role played by epigenetic events, including BCL6 translocation, p53 expression, and microenvironment components, including macrophage and mast cell infiltration and microvascular density involved in the regulation of breast cancer progression.
Collapse
|
83
|
Mantovani A, Schioppa T, Biswas SK, Marchesi F, Allavena P, Sica A. Tumor-Associated Macrophages and Dendritic Cells as Prototypic Type II Polarized Myeloid Populations. TUMORI JOURNAL 2018; 89:459-68. [PMID: 14870765 DOI: 10.1177/030089160308900501] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Environmental signals polarize mononuclear phagocytes which can express different functional programmes. Fully polarized type I and type II (or alternatively activated) macrophages are the extremes of a continuum of functional states. Tumor-derived and T cell-derived cytokines stimulate tumor associated macrophages (TAM) to acquire a polarized type II phenotype. These functionally polarized cells, and similarly oriented or immature dendritic cells present in tumors, play a key role in subversion of adaptive immunity and in inflammatory circuits which promote tumor growth and progression.
Collapse
|
84
|
Shrivastava R, Asif M, Singh V, Dubey P, Ahmad Malik S, Lone MUD, Tewari BN, Baghel KS, Pal S, Nagar GK, Chattopadhyay N, Bhadauria S. M2 polarization of macrophages by Oncostatin M in hypoxic tumor microenvironment is mediated by mTORC2 and promotes tumor growth and metastasis. Cytokine 2018; 118:130-143. [PMID: 29625858 DOI: 10.1016/j.cyto.2018.03.032] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/16/2022]
Abstract
Oncostatin M (OSM), an inflammatory cytokine belonging to the interleukin-6 (IL-6) superfamily, plays a vital role in multitude of physiological and pathological processes. Its role in breast tumor progression and metastasis to distant organs is well documented. Recent reports implicate OSM in macrophage M2 polarization, a key pro-tumoral phenomenon. M2 polarization of macrophages is believed to promote tumor progression by potentiating metastasis and angiogenesis. In the current study, we delineated the mechanism underlying OSM induced macrophage M2 polarization. The findings revealed that OSM skews macrophages towards an M2 polarized phenotype via mTOR signaling complex 2 (mTORC2). mTORC2 relays signals through two effector kinases i.e. PKC-α and Akt. Our results indicated that mTORC2 mediated M2 polarization of macrophages is not dependent on PKC-α and is primarily affected via Akt, particularly Akt1. In vivo studies conducted on 4T1/BALB/c mouse orthotropic model of breast cancer further corroborated these observations wherein i.v. reintroduction of mTORC2 abrogated monocytes into orthotropic mouse model resulted in diminished acquisition of M2 specific attributes by tumor associated macrophages. Metastasis to distant organs like lung, liver and bone was reduced as evident by decrease in formation of focal metastatic lesions in mTORC2 abrogated monocytes mice. Our study pinpoints key role of mTORC2-Akt1 axis in OSM induced macrophage polarization and suggests for possible usage of Oncostatin-M blockade and/or selective mTORC2 inhibition as a potential anti-cancer strategy particularly with reference to metastasis of breast cancer to distant organs such as lung, liver and bone.
Collapse
Affiliation(s)
- Richa Shrivastava
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India
| | - Mohammad Asif
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Varsha Singh
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Parul Dubey
- Department of Surgical Oncology, King George Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Showkat Ahmad Malik
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Mehraj-U-Din Lone
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Brij Nath Tewari
- Department of Surgical Oncology, King George Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Khemraj Singh Baghel
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Subhashis Pal
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Geet Kumar Nagar
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India
| | - Smrati Bhadauria
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110025, India.
| |
Collapse
|
85
|
Lee JH, Kim K, Jo YH, Hwang JE, Chung HJ, Yang C. Reoxygenation speed and its implication for cellular injury responses in hypoxic RAW 264.7 cells. J Surg Res 2018; 227:88-94. [PMID: 29804868 DOI: 10.1016/j.jss.2017.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 09/12/2017] [Accepted: 11/03/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Ischemia/reperfusion injury is characterized by excess generation of reactive oxygen species (ROS). The purpose of this study is to test the effect of reoxygenation speed on ROS production and the cellular injury responses in hypoxic macrophages RAW 264.7 cells and its potential mechanisms for the generation of ROS. MATERIALS AND METHODS After hypoxic exposure of RAW 264.7 cells for 20 h, reoxygenation was performed for 6 h by stepwise increase in oxygen concentration (0.8% increase of oxygen every 15 min) in the slow reoxygenation (SRox) group or by moving the culture flasks quickly to a normoxic incubator in the rapid reoxygenation (RRox) group. To identify the potential effect of reoxygenation speed on the generation of ROS, the cells were pretreated with apocynin, VAS2870, and MitoTEMPO before the induction of hypoxia. RESULTS SRox significantly decreased cell death and cytotoxicity compared with RRox (P < 0.05). RRox resulted in significantly more generation of ROS, interleukin-1β, interleukin-6, and nitric oxide than SRox (P < 0.05). SRox also increased the expression of prosurvival proteins and decreased apoptosis. In cells pretreated with VAS2870 or MitoTEMPO, the reduced ROS generation by SRox was maintained. However, pretreatment with apocynin abolished the effect of reoxygenation speed on ROS generation. CONCLUSIONS SRox compared with RRox decreased cellular injury in hypoxic RAW 264.7 cells by decreasing ROS and inflammatory cytokine production and decreasing apoptosis.
Collapse
Affiliation(s)
- Jae Hyuk Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Kyuseok Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea.
| | - You Hwan Jo
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Ji Eun Hwang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Hea Jin Chung
- Department of Emergency Medicine, Emergency Care Center, Soonchunhyang University Hospital, Seoul, Republic of Korea
| | - Chungmi Yang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| |
Collapse
|
86
|
Kim SY, Shin J, Kim DH, Kim EK, Moon HJ, Yoon JH, You JK, Kim MJ. Correlation between electrical conductivity and apparent diffusion coefficient in breast cancer: effect of necrosis on magnetic resonance imaging. Eur Radiol 2018; 28:3204-3214. [DOI: 10.1007/s00330-017-5291-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/10/2017] [Accepted: 12/27/2017] [Indexed: 11/28/2022]
|
87
|
Crittenden MR, Baird J, Friedman D, Savage T, Uhde L, Alice A, Cottam B, Young K, Newell P, Nguyen C, Bambina S, Kramer G, Akporiaye E, Malecka A, Jackson A, Gough MJ. Mertk on tumor macrophages is a therapeutic target to prevent tumor recurrence following radiation therapy. Oncotarget 2018; 7:78653-78666. [PMID: 27602953 PMCID: PMC5346667 DOI: 10.18632/oncotarget.11823] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 08/25/2016] [Indexed: 12/19/2022] Open
Abstract
Radiation therapy provides a means to kill large numbers of cancer cells in a controlled location resulting in the release of tumor-specific antigens and endogenous adjuvants. However, by activating pathways involved in apoptotic cell recognition and phagocytosis, irradiated cancer cells engender suppressive phenotypes in macrophages. We demonstrate that the macrophage-specific phagocytic receptor, Mertk is upregulated in macrophages in the tumor following radiation therapy. Ligation of Mertk on macrophages results in anti-inflammatory cytokine responses via NF-kB p50 upregulation, which in turn limits tumor control following radiation therapy. We demonstrate that in immunogenic tumors, loss of Mertk is sufficient to permit tumor cure following radiation therapy. However, in poorly immunogenic tumors, TGFβ inhibition is also required to result in tumor cure following radiation therapy. These data demonstrate that Mertk is a highly specific target whose absence permits tumor control in combination with radiation therapy.
Collapse
Affiliation(s)
- Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA.,The Oregon Clinic, Portland OR, USA
| | - Jason Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - David Friedman
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Talicia Savage
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Lauren Uhde
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Benjamin Cottam
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Kristina Young
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA.,The Oregon Clinic, Portland OR, USA
| | - Pippa Newell
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA.,Providence Hepatobiliary and Pancreatic Cancer Program, Providence Portland Medical Center, Portland OR, USA
| | - Cynthia Nguyen
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Emmanuel Akporiaye
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| | - Anna Malecka
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, University of Nottingham, UK
| | - Andrew Jackson
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, University of Nottingham, UK
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland OR, USA
| |
Collapse
|
88
|
Shin SH, Park SH, Kim SW, Kim M, Kim D. Fluorine MR Imaging Monitoring of Tumor Inflammation after High-Intensity Focused Ultrasound Ablation. Radiology 2018; 287:476-484. [PMID: 29369752 DOI: 10.1148/radiol.2017171603] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose To investigate whether high-intensity focused ultrasound (HIFU)-induced macrophage infiltration could be longitudinally monitored with fluorine 19 (19F) magnetic resonance (MR) imaging in a quantitative manner. Materials and Methods BALB/c mice were subcutaneously inoculated with 4T1 cells and were separated into three groups: untreated mice (control, n = 9), HIFU-treated mice (HIFU, n = 9), and HIFU- and clodronate-treated mice (HIFU+Clod, n = 9). Immediately after HIFU treatment, all mice were intravenously given perfluorocarbon (PFC) emulsion. MR imaging examinations were performed 2, 4, 7, 10, and 14 days after HIFU treatment. Two-way repeated measures analysis of variance was used to analyze the changes in 19F signal over time and differences between groups. Histologic examinations were performed to confirm in vivo data. Results Fluorine 19 signals were detected at the rims of tumors and the peripheries of ablated lesions. Mean 19F signal in tumors was significantly higher in HIFU-treated mice than in control mice up to day 4 (0.82 ± 0.26 vs 0.42 ± 0.17, P < .001). Fluorine 19 signals were higher in the HIFU+Clod group than in the control group from day 4 (0.82 ± 0.23, P < .001) to day 14 (0.55 ± 0.16 vs 0.28 ± 0.06, P < .05). Histologic examination revealed macrophage infiltration around ablated lesions. Immunofluorescence staining confirmed PFC labeling of macrophages. Conclusion Fluorine 19 MR imaging can longitudinally capture and quantify HIFU-induced macrophage infiltration in preclinical tumor models. © RSNA, 2018 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Soo Hyun Shin
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Sang Hyun Park
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Seung Won Kim
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Minsun Kim
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| | - Daehong Kim
- From the Molecular Imaging Branch, Division of Convergence Technology, National Cancer Center, Research Building, Ilsanro-323, Ilsandong-gu, Goyang 10408, Korea
| |
Collapse
|
89
|
Alipanah H, Bigdeli MR, Esmaeili MA. Inhibitory Effect of Viola odorata Extract on Tumor Growth and Metastasis in 4T1 Breast Cancer Model. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:276-291. [PMID: 29755559 PMCID: PMC5937098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Viola odorata as a medical herb is used in liver disorders and relieving cancer pain. In the present study, the cytotoxic, antioxidant, and anti-metastatic properties of Viola odorata hydro-alcoholic extract (VOE) were investigated in 4T1 breast cancer model. After treatment of 4T1 breast cancer cells with VOE, cell viability was measured by MTT assay. The implanted mice were treated with different concentration of VOE (50, 150 and 250 mg/kg) for 21 days. Levels of lactate dehydrogenase (LDH), γ -glutamyl transferase (GGT), alkaline phosphatase (ALP), carcinoembryonic antigen (CEA) and cancer antigen 15-3(CA15-3) in serum, and also catalase (CAT) and superoxide dismutase (SOD) activities in tumor tissue were measured. Metastatic rate was investigated in liver, spleen and lung tissues. VOE decreased cell viability of 4T1 cells, significantly. VOE significantly inhibited the cell proliferation, but not vasculature in the tumors that revealed by immunohistochemical analysis for Ki-67 and CD31 expression, respectively. VOE increased the Bax/Bcl-2 ratio in VOE250-treated group compared to control group. Serum analysis showed that treatment with 250 mg/kg of VOE significantly reduced LDH (not ALP and GGT) levels compared to controls. No linear correlation was found between the values of CEA and CA15-3 with tumor size. The rate of CAT activity was increased in VOE250-treated rats whereas, CAT and SOD activities were reduced in VOE50 group. VOE250 significantly decreased the metastatic rate in liver and lung compared to the other doses of VOE. Consequently, Viola odorata has cytotoxic effects on 4T1 cells and affects antioxidant activity and metastasis in breast cancer.
Collapse
Affiliation(s)
- Hiva Alipanah
- Faculty of life sciences and biotechnology, Shahid Beheshti University, G.C., Tehran, Iran.
| | - Mohammad Reza Bigdeli
- Faculty of life sciences and biotechnology, Shahid Beheshti University, G.C., Tehran, Iran. ,Corresponding author: E-mail:
| | - Mohammad Ali Esmaeili
- Department of Biology, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
90
|
Rohne P, Wolf S, Dörr C, Ringen J, Holtz A, Gollan R, Renner B, Prochnow H, Baiersdörfer M, Koch-Brandt C. Exposure of vital cells to necrotic cell lysates induce the IRE1α branch of the unfolded protein response and cell proliferation. Cell Stress Chaperones 2018; 23:77-88. [PMID: 28687980 PMCID: PMC5741583 DOI: 10.1007/s12192-017-0825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/15/2017] [Accepted: 06/16/2017] [Indexed: 10/19/2022] Open
Abstract
Necrosis is a form of cell death that is detrimental to the affected tissue because the cell ruptures and releases its content (reactive oxygen species among others) into the extracellular space. Clusterin (CLU), a cytoprotective extracellular chaperone has been shown to be upregulated in the face of necrosis. We here show that in addition to CLU upregulation, necrotic cell lysates induce JNK/SAPK signaling, the IRE1α branch of the unfolded protein response (UPR), the MAPK/ERK1/2, and the mTOR signaling pathways and results in an enhanced proliferation of the vital surrounding cells. We name this novel response mechanism: Necrosis-induced Proliferation (NiP).
Collapse
Affiliation(s)
- Philipp Rohne
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Steven Wolf
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
- Department of Pathology, The University of Chicago, Chicago, IL USA
| | - Carolin Dörr
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Julia Ringen
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Andrew Holtz
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - René Gollan
- Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Benjamin Renner
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Hans Prochnow
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH, Braunschweig, Germany
| | - Markus Baiersdörfer
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| | - Claudia Koch-Brandt
- Institute of Pharmacy and Biochemistry - Therapeutical Life Sciences, Johannes Gutenberg University of Mainz, Johann-Joachim Becherweg 30, 55128 Mainz, Germany
| |
Collapse
|
91
|
Greish K, Jasim A, Parayath N, Abdelghany S, Alkhateeb A, Taurin S, Nehoff H. Micellar formulations of Crizotinib and Dasatinib in the management of glioblastoma multiforme. J Drug Target 2017; 26:692-708. [PMID: 29251531 DOI: 10.1080/1061186x.2017.1419357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) defies the currently practiced management of radiotherapy, chemotherapy and surgery and hence, it is associated with a high fatality rate with a median survival of 14.6 months. In our previous work investigating different tyrosine kinase inhibitors (TKIs), we established that a combination of Crizotinib and Dasatinib exerted the most potent effect on different GBM cell lines. In this work, to improve targeted therapy at the site of the tumour and avoid systemic toxicity, we exploited the enhanced permeability and retention effect by designing micellar formulations of these two TKIs. Crizotinib and Dasatinib were successfully encapsulated in poly(styrene-co-maleic acid) (SMA) micelles which were then evaluated for their physicochemical characteristics, anti-proliferative effect, mode of cell death, efficacy in spheroid models, effect on cell signalling, antiangiogenic potential and in vivo anticancer activity. Our results showed that this combination had induced a potent anti-proliferative effect in four GBM cell lines grown as a monolayer and as a spheroid. The combination was also efficacious in in vitro models of angiogenesis and vascular mimicry. In vivo data showed the enhanced activity of the micellar TKIs compared to free drugs. In conclusion, we proved that micellar formulations of Crizotinib and Dasatinib carry promising in vitro and in vivo efficacy that warrant further investigation.
Collapse
Affiliation(s)
- Khaled Greish
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Anfal Jasim
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Neha Parayath
- b Department of Pharmaceutical Sciences , Northeastern University , Boston , MA , USA
| | - Sara Abdelghany
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Ali Alkhateeb
- a College of Medicine and Medical Sciences, Department of Molecular Medicine, and Nanomedicine Unit , Princess Al-Jawhara Center for Molecular Medicine, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Sebastien Taurin
- c Department of Obstetrics and Gynecology , University of Utah , Salt Lake City , UT , USA
| | - Hayley Nehoff
- d Department of Pharmacology and Toxicology , University of Otago , Dunedin , New Zealand
| |
Collapse
|
92
|
Fluorine-19 Magnetic Resonance Imaging and Positron Emission Tomography of Tumor-Associated Macrophages and Tumor Metabolism. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:4896310. [PMID: 29362559 PMCID: PMC5736905 DOI: 10.1155/2017/4896310] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 10/31/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
Abstract
The presence of tumor-associated macrophages (TAMs) is significantly associated with poor prognosis of tumors. Currently, magnetic resonance imaging- (MRI-) based TAM imaging methods that use nanoparticles such as superparamagnetic iron oxide and perfluorocarbon nanoemulsions are available for quantitative monitoring of TAM burden in tumors. However, whether MRI-based measurements of TAMs can be used as prognostic markers has not been evaluated yet. In this study, we used positron emission tomography (PET) with 18F-2-fluoro-2-deoxy-D-glucose (18F-FDG) as a radioactive tracer and fluorine-19- (19F-) MRI for imaging mouse breast cancer models to determine any association between TAM infiltration and tumor metabolism. Perfluorocarbon nanoemulsions were intravenously administered to track and quantify TAM infiltration using a 7T MR scanner. To analyze glucose uptake in tumors, 18F-FDG-PET images were acquired immediately after 19F-MRI. Coregistered 18F-FDG-PET and 19F-MR images enabled comparison of spatial patterns of glucose uptake and TAM distribution in tumors. 19F-MR signal intensities from tumors exhibited a strong inverse correlation with 18F-FDG uptake while having a significant positive correlation with tumor growth from days 2 to 7. These results show that combination of 19F-MRI and 18F-FDG-PET can improve our understanding of the relationship between TAM and tumor microenvironment.
Collapse
|
93
|
Abstract
Radiation therapy is primarily a modality to kill cancer cells in the treatment field. It is becoming increasingly clear that radiation therapy can also be used to direct immune responses that have the potential to clear residual local or distant disease outside the treatment field. We believe that cancer cell death is the critical link between these processes. Understanding the handling of dying cancer cells by immune cells in the tumor environment is crucial to facilitate immune responses following radiation therapy. We review the role of the TAM (Tyro3 Axl Mertk) group of receptor tyrosine kinases and their role following radiation-induced cancer cell death in the tumor environment.
Collapse
|
94
|
尚 柳, 杨 家, 卢 晶, 王 婷, 周 颖, 邢 新, 王 鑫, 杨 淑, 胡 明. [Correlations between apparent diffusion coefficient in diffusion?weighted magnetic resonance imaging and molecular subtypes of invasive breast cancer masses]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1410-1414. [PMID: 29070476 PMCID: PMC6743964 DOI: 10.3969/j.issn.1673-4254.2017.10.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To study the correlation of apparent diffusion coefficient (ADC) measured by diffusion-weighted magnetic resonance imaging (MRI) with the molecular subtypes and biological prognostic factors of invasive breast cancer masses. METHODS Breast MRI data (including dynamic enhanced and diffusion-weighted imaging) were collected from 64 patients with pathologically confirmed invasive breast cancer masses (a total of 69 lesions). The mean ADC values of the lesions were calculated and their correlations were analyzed with the 5 molecular subtypes of invasive breast cancer and the biological prognostic factors including estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor 2 (HER2), and Ki-67 index. RESULTS The ADC values did not differ significantly among the 5 molecular subtypes of invasive breast cancer masses (P>0.05) or among lesions with different ER, PR, or HER2 status (P>0.05). The mean ADC values were significantly higher in Ki-67-positive lesions than in the negative lesions (P=0.023 and negatively correlated with the expressions of Ki-67 (r=-0.249). CONCLUSION ADC value can not be used to identify the molecular subtypes of invasive breast cancer masses or to evaluate the biological prognosis of the lesions, but its correlation with Ki-67 expression may help in prognostic evaluation and guiding clinical therapy of the tumors.
Collapse
Affiliation(s)
- 柳彤 尚
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 家斐 杨
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 晶 卢
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 婷婷 王
- 新疆医科大学公共卫生学院儿少卫生与妇幼保健学教研室, 新疆 乌鲁木齐 830000Department of Maternal, Child and Adolescent Health, School of Public Health, Xinjiang Medical University, Urumqi 830000, China
| | - 颖 周
- 解放军总医院第一附属医院 病理科, 北京 100047Department of Pathology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 新博 邢
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 鑫坤 王
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 淑辉 杨
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| | - 明艳 胡
- 解放军总医院第一附属医院 放射科, 北京 100047Department of Radiology, First Affiliated Hospital of General Hospital of PLA, Beijing 100047, China
| |
Collapse
|
95
|
Hong YM, Cho M, Yoon KT, Chu CW, Yang KH, Park YM, Rhu JH. Risk factors of early recurrence after curative hepatectomy in hepatocellular carcinoma. Tumour Biol 2017; 39:1010428317720863. [PMID: 29034775 DOI: 10.1177/1010428317720863] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Early recurrence is common after curative hepatectomy for hepatocellular carcinoma and is associated with poor prognosis. This study aimed to identify risk factors of early recurrence after curative hepatectomy in hepatocellular carcinoma. Overall, 63 patients who underwent curative hepatectomy for hepatocellular carcinoma were enrolled. Patients were divided into the early recurrence group, who developed recurrence within 12 months after hepatectomy (n = 10), and the non-early recurrence group (n = 53). Clinicopathological factors of early recurrence were retrospectively analyzed. Among the 63 patients, 10 (15.9%) patients experienced early recurrence. Univariate analysis showed tumor necrosis (p = 0.012), level of PIVKA-II (prothrombin induced by vitamin K absence or antagonist-II; p = 0.002), and microvascular invasion (p = 0.029) to be associated with early recurrence. By multivariate analysis, there were significant differences in high PIVKA-II (p < 0.001) and tumor necrosis (p = 0.012) in patients with early recurrence. The optimal cutoff values of PIVKA-II and tumor necrosis were 46 mAU/mL and 3% of total tumor volume, respectively. Patients with a high preoperative PIVKA-II level and extent of tumor necrosis, which are independent risk factors for early recurrence, should be actively treated and monitored closely after hepatectomy.
Collapse
Affiliation(s)
- Young Mi Hong
- 1 Department of Internal Medicine, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Mong Cho
- 1 Department of Internal Medicine, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Ki Tae Yoon
- 1 Department of Internal Medicine, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Chong Woo Chu
- 2 Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Kwang Ho Yang
- 2 Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Yong Mok Park
- 2 Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Je Ho Rhu
- 2 Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, College of Medicine Pusan National University, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
96
|
Dielectric imaging for differentiation between cancer and inflammation in vivo. Sci Rep 2017; 7:13137. [PMID: 29030581 PMCID: PMC5640678 DOI: 10.1038/s41598-017-13545-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
In this study, we develop an in vivo dielectric imaging technique that measures capacitance using pin-type electrode arrays. Compared to normal tissues, cancer tissues exhibit higher capacitance values, allowing us to image the cancer region and monitor the chemotherapeutic effects of cancer in real-time. A comparison with the histopathological results shows that the in vivo dielectric imaging technique is able to detect small tumors (<3 mm) and tumor-associated changes. In addition, we demonstrate that cancer and inflammation may be distinguished by measuring the capacitance images at different frequencies. In contrast, the positron emission tomography using 2-[18F]-fluoro-2-deoxy-D-glucose was not capable of discriminating between cancer and inflammation.
Collapse
|
97
|
Correlation of X-ray diffraction signatures of breast tissue and their histopathological classification. Sci Rep 2017; 7:12998. [PMID: 29021531 PMCID: PMC5636903 DOI: 10.1038/s41598-017-13399-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/25/2017] [Indexed: 12/05/2022] Open
Abstract
This pilot study examines the correlation of X-ray diffraction (XRD) measurements with the histopathological analysis of breast tissue. Eight breast cancer samples were investigated. Each sample contained a mixture of normal and cancerous tissues. In total, 522 separate XRD measurements were made at different locations across the samples (8 in total). The resulting XRD spectra were subjected to principal component analysis (PCA) in order to determine if there were any distinguishing features that could be used to identify different tissue components. 99.0% of the variation between the spectra were described by the first two principal components (PC). Comparing the location of points in PC space with the classification determined by histopathology indicated correlation between the shape/magnitude of the XRD spectra and the tissue type. These results are encouraging and suggest that XRD could be used for the intraoperative or postoperative classification of bulk tissue samples.
Collapse
|
98
|
Chawla H, Urs AB, Augustine J. Association of Macrophages With Angiogenesis in Oral Epithelial Dysplasia, Oral Verrucous Carcinoma, and Oral Squamous Cell Carcinoma: An Immunohistochemical Study. Appl Immunohistochem Mol Morphol 2017; 25:203-208. [PMID: 26657870 DOI: 10.1097/pai.0000000000000284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The present study aimed to elucidate the role of tumor-associated macrophages (TAMs) and angiogenesis in tumor progression by assessing their immunohistochemical expression in oral epithelial dysplasia (OED), oral verrucous carcinoma (OVC), and oral squamous cell carcinoma (OSCC). About 20 histopathologically confirmed cases of OED, OVC, and OSCC each and 10 cases of normal oral mucosa taken as controls were stained immunohistochemically using CD68 and CD31 antibodies. The average TAM count and the microvessel density (MVD) were calculated for each group and expressed as mean±SD and compared using the Mann-Whitney U Test. Pearson correlation was applied to assess the correlation between TAM and MVD in different groups. The CD68 count was found to be significantly higher in all the groups as compared with controls, with the highest counts in OSCC, followed by OED and OVC. MVD was significantly higher in all the test groups as compared with controls. The increase in MVD was highly significant in OSCC as compared with OVC, and in OVC as compared with OED. No association could be determined between TAM and MVD. There does not seem to be any direct influence of macrophages on angiogenesis in the microenvironment of OED, OVC, and OSCC.
Collapse
Affiliation(s)
- Himanshi Chawla
- Department of Oral & Maxillofacial Pathology, Maulana Azad Institute of Dental Sciences, New Delhi, India
| | | | | |
Collapse
|
99
|
Hodgson A, Xu B, Satkunasivam R, Downes MR. Tumour front inflammation and necrosis are independent prognostic predictors in high-grade urothelial carcinoma of the bladder. J Clin Pathol 2017; 71:154-160. [DOI: 10.1136/jclinpath-2017-204562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/08/2017] [Accepted: 06/13/2017] [Indexed: 11/04/2022]
Abstract
AimsInflammation and necrosis have been associated with prognosis in multiple epithelial malignancies. Our objective was to evaluate inflammation and necrosis in a cohort of patients with high-grade urothelial carcinomas of the bladder to determine their association with pathological parameters and their prognostic effect on relapse-free and disease-specific survival.MethodsA retrospective cohort that underwent radical cystectomy for urothelial carcinomas (n=235) was evaluated for invasive front and central inflammation using the Klintrup-Makinen assessment method. Necrosis was scored using a four-point scale. The relationship of inflammation and necrosis with stage, nodal status, carcinoma in situ, tumour size, margin status and vascular space invasion and the impact on relapse-free and disease-specific survival were calculated using appropriate statistical tests.ResultsOn multivariate analysis, invasive front inflammation (p=0.003) and necrosis (p=0.000) were independent predictors of relapse-free survival. Both invasive front inflammation (p=0.009) and necrosis (p=0.002) again were independent predictors of disease-specific survival. For pathological features, low invasive front inflammation was associated with lymphovascular space invasion (p=0.008), a positive soft tissue margin (p=0.028) and carcinoma in situ (p=0.042). Necrosis was statistically associated with tumours >3 cm in size (p=0.013) and carcinoma in situ (p<0.001).ConclusionsNecrosis and invasive front inflammation are additional histological variables with independent prognostic relevance in high-grade urothelial carcinoma of the bladder.
Collapse
|
100
|
Yin J, Oh YT, Kim JY, Kim SS, Choi E, Kim TH, Hong JH, Chang N, Cho HJ, Sa JK, Kim JC, Kwon HJ, Park S, Lin W, Nakano I, Gwak HS, Yoo H, Lee SH, Lee J, Kim JH, Kim SY, Nam DH, Park MJ, Park JB. Transglutaminase 2 Inhibition Reverses Mesenchymal Transdifferentiation of Glioma Stem Cells by Regulating C/EBPβ Signaling. Cancer Res 2017; 77:4973-4984. [PMID: 28754668 DOI: 10.1158/0008-5472.can-17-0388] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/30/2017] [Accepted: 07/11/2017] [Indexed: 11/16/2022]
Abstract
Necrosis is a hallmark of glioblastoma (GBM) and is responsible for poor prognosis and resistance to conventional therapies. However, the molecular mechanisms underlying necrotic microenvironment-induced malignancy of GBM have not been elucidated. Here, we report that transglutaminase 2 (TGM2) is upregulated in the perinecrotic region of GBM and triggered mesenchymal (MES) transdifferentiation of glioma stem cells (GSC) by regulating master transcription factors (TF), such as C/EBPβ, TAZ, and STAT3. TGM2 expression was induced by macrophages/microglia-derived cytokines via NF-κB activation and further degraded DNA damage-inducible transcript 3 (GADD153) to induce C/EBPβ expression, resulting in expression of the MES transcriptome. Downregulation of TGM2 decreased sphere-forming ability, tumor size, and radioresistance and survival in a xenograft mouse model through a loss of the MES signature. A TGM2-specific inhibitor GK921 blocked MES transdifferentiation and showed significant therapeutic efficacy in mouse models of GSC. Moreover, TGM2 expression was significantly increased in recurrent MES patients and inversely correlated with patient prognosis. Collectively, our results indicate that TGM2 is a key molecular switch of necrosis-induced MES transdifferentiation and an important therapeutic target for MES GBM. Cancer Res; 77(18); 4973-84. ©2017 AACR.
Collapse
Affiliation(s)
- Jinlong Yin
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.,Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Young Taek Oh
- Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Jeong-Yub Kim
- Division of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.,Department of Pathology, College of Medicine, Korea University, Seoul, Korea
| | - Sung Soo Kim
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Eunji Choi
- Department of Cancer Control and Policy, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Tae Hoon Kim
- Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Jun Hee Hong
- Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Nakho Chang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Hee Jin Cho
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Jason K Sa
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Jeong Cheol Kim
- Division of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hyung Joon Kwon
- Department of Cancer Control and Policy, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Saewhan Park
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Weiwei Lin
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama.,UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ho-Shin Gwak
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.,Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Heon Yoo
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.,Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Seung-Hoon Lee
- Department of Neurosurgery, Eulji University School of Medicine, Daejeon, Korea
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jong Heon Kim
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.,Cancer Cell and Molecular Biology Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Soo-Youl Kim
- Cancer Cell and Molecular Biology Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea. .,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Jin Park
- Division of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.
| | - Jong Bae Park
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea. .,Specific Organs Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| |
Collapse
|