1
|
Rodrigues J, Amin A, Chandra S, Mulla NJ, Nayak GS, Rai S, Ray S, Mahato KK. Machine Learning Enabled Photoacoustic Spectroscopy for Noninvasive Assessment of Breast Tumor Progression In Vivo: A Preclinical Study. ACS Sens 2024; 9:589-601. [PMID: 38288735 PMCID: PMC10897932 DOI: 10.1021/acssensors.3c01085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/25/2023] [Accepted: 01/17/2024] [Indexed: 02/24/2024]
Abstract
Breast cancer is a dreaded disease affecting women the most in cancer-related deaths over other cancers. However, early diagnosis of the disease can help increase survival rates. The existing breast cancer diagnosis tools do not support the early diagnosis of the disease. Therefore, there is a great need to develop early diagnostic tools for this cancer. Photoacoustic spectroscopy (PAS), being very sensitive to biochemical changes, can be relied upon for its application in detecting breast tumors in vivo. With this motivation, in the current study, an aseptic chamber integrated photoacoustic (PA) probe was designed and developed to monitor breast tumor progression in vivo, established in nude mice. The device served the dual purpose of transporting tumor-bearing animals to the laboratory from the animal house and performing PA experiments in the same chamber, maintaining sterility. In the current study, breast tumor was induced in the nude mice by MCF-7 cells injection and the corresponding PA spectra at different time points (day 0, 5, 10, 15, and 20) of tumor progression in vivo in the same animals. The recorded photoacoustic spectra were subsequently preprocessed, wavelet-transformed, and subjected to filter-based feature selection algorithm. The selected top 20 features, by minimum redundancy maximum relevance (mRMR) algorithm, were then used to build an input feature matrix for machine learning (ML)-based classification of the data. The performance of classification models demonstrated 100% specificity, whereas the sensitivity of 95, 100, 92.5, and 85% for the time points, day 5, 10, 15, and 20, respectively. These results suggest the potential of PA signal-based classification of breast tumor progression in a preclinical model. The PA signal contains information on the biochemical changes associated with disease progression, emphasizing its translational strength toward early disease diagnosis.
Collapse
Affiliation(s)
- Jackson Rodrigues
- Department
of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Ashwini Amin
- Department
of Computer Science and Engineering, Manipal
Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, India
| | - Subhash Chandra
- Department
of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Nitufa J. Mulla
- Department
of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - G. Subramanya Nayak
- Department
of Electronics and Communication, Manipal
Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sharada Rai
- Department
of Pathology, Kasturba Medical College Mangalore,
Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Satadru Ray
- Department
of Surgery, Kasturba Medical College, Manipal
Academy of Higher Education, Karnataka,Manipal 576104, India
| | - Krishna Kishore Mahato
- Department
of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| |
Collapse
|
2
|
Zhang Q, Zhao Y, Nie J, Long Q, Wang X, Wang X, Gong G, Liao L, Yi X, Chen BT. Pretreatment synthetic MRI features for triple-negative breast cancer. Clin Radiol 2024; 79:e219-e226. [PMID: 37935611 DOI: 10.1016/j.crad.2023.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023]
Abstract
AIM To evaluate the quantitative parameters derived from synthetic magnetic resonance imaging (SyMRI) for predicting triple-negative breast cancer (TNBC). MATERIALS AND METHODS This prospective study enrolled participants with invasive ductal breast carcinoma (IDBC) and separated them into a TNBC group and a Non-TNBC group. Preoperative breast MRI included both the SyMRI and conventional MRI sequences. The quantitative parameters derived from the SyMRI included T1 and T2 relaxation times, proton density (PD), and their standard deviations (SD). Clinicopathological characteristics, conventional MRI findings, and quantitative synthetic parameters were assessed for all participants. Multivariable logistic regression analysis was performed to determine the potential independent imaging predictors for TNBC preoperatively. Receiver operating characteristic (ROC) curve analysis was used to evaluate the performance of these parameters. RESULTS A total of 231 participants with histopathological proven IDBC were included in this study (n=46 in the TNBC group and n=185 in the Non-TNBC group). The TNBC group had significantly larger tumour size (p=0.011) and more frequent intratumoural cystic or necrotic lesions (p<0.001) as compared to the Non-TNBC group. The univariate analysis showed that the TNBC tumours had significantly higher T1 (p=0.006) and T2 (p<0.001) values than Non-TNBC tumours. Subsequent multivariable analysis indicated that T2 values and the presence of cystic or necrotic lesions were the independent predictors for TNBC. CONCLUSION The T2 from synthetic imaging and the presence of cystic degeneration or necrosis within the breast cancer may serve as potential imaging biomarkers for preoperative differentiation of TNBC from Non-TNBC.
Collapse
Affiliation(s)
- Q Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Clinical Research Center for Breast Cancer, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China
| | - Y Zhao
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China; Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - J Nie
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China; Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Q Long
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China; Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - X Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Clinical Research Center for Breast Cancer, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China
| | - X Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Clinical Research Center for Breast Cancer, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China
| | - G Gong
- Department of Pathology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, PR China
| | - L Liao
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Clinical Research Center for Breast Cancer, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha 410008, Hunan, PR China.
| | - X Yi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Changsha 410008, Hunan, PR China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - B T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
3
|
Tan SY, Tan PH, Tan VKM, Leong LCH. Extensively infarcted breast cancer. BMJ Case Rep 2023; 16:e253823. [PMID: 37678940 PMCID: PMC10496683 DOI: 10.1136/bcr-2022-253823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023] Open
Abstract
This is a case of a tumour that appeared largely unviable after near complete infarction. The lesion presented as a regular shaped mass with cystic appearance lacking definitive malignant radiological signs. Together with the initial non-diagnostic histological result, this could have easily led to a missed diagnosis of cancer.
Collapse
Affiliation(s)
- Si Ying Tan
- Department of Breast Surgery, National Cancer Centre Singapore, Singapore
- Department of Breast Surgery, Singapore General Hospital, Singapore
- SingHealth Duke-NUS Breast Centre, Singapore
| | | | - Veronique Kiak Mien Tan
- Department of Breast Surgery, National Cancer Centre Singapore, Singapore
- Department of Breast Surgery, Singapore General Hospital, Singapore
- SingHealth Duke-NUS Breast Centre, Singapore
| | | |
Collapse
|
4
|
Chen J, Li Z, Han Z, Kang D, Ma J, Yi Y, Fu F, Guo W, Zheng L, Xi G, He J, Qiu L, Li L, Zhang Q, Wang C, Chen J. Prognostic value of tumor necrosis based on the evaluation of frequency in invasive breast cancer. BMC Cancer 2023; 23:530. [PMID: 37296414 DOI: 10.1186/s12885-023-10943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/10/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Tumor necrosis (TN) was associated with poor prognosis. However, the traditional classification of TN ignored spatial intratumor heterogeneity, which may be associated with important prognosis. The purpose of this study was to propose a new method to reveal the hidden prognostic value of spatial heterogeneity of TN in invasive breast cancer (IBC). METHODS Multiphoton microscopy (MPM) was used to obtain multiphoton images from 471 patients. According to the relative spatial positions of TN, tumor cells, collagen fibers and myoepithelium, four spatial heterogeneities of TN (TN1-4) were defined. Based on the frequency of individual TN, TN-score was obtained to investigate the prognostic value of TN. RESULTS Patients with high-risk TN had worse 5-year disease-free survival (DFS) than patients with no necrosis (32.5% vs. 64.7%; P < 0.0001 in training set; 45.8% vs. 70.8%; P = 0.017 in validation set), while patients with low-risk TN had a 5-year DFS comparable to patients with no necrosis (60.0% vs. 64.7%; P = 0.497 in training set; 59.8% vs. 70.8%; P = 0.121 in validation set). Furthermore, high-risk TN "up-staged" the patients with IBC. Patients with high-risk TN and stage I tumors had a 5-year DFS comparable to patients with stage II tumors (55.6% vs. 62.0%; P = 0.565 in training set; 62.5% vs. 66.3%; P = 0.856 in validation set), as well as patients with high-risk TN and stage II tumors had a 5-year DFS comparable to patients with stage III tumors (33.3% vs. 24.6%; P = 0.271 in training set; 44.4% vs. 39.3%; P = 0.519 in validation set). CONCLUSIONS TN-score was an independent prognostic factor for 5-year DFS. Only high-risk TN was associated with poor prognosis. High-risk TN "up-staged" the patients with IBC. Incorporating TN-score into staging category could improve its performance to stratify patients.
Collapse
Affiliation(s)
- Jianhua Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
- College of Life Science, Fujian Normal University, Fuzhou, 350117, China
| | - Zhijun Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
| | - Zhonghua Han
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jianli Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yu Yi
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Wenhui Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
| | - Gangqin Xi
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
| | - Jiajia He
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
| | - Lida Qiu
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, 350108, China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350117, China.
| |
Collapse
|
5
|
Medler TR, Blair TC, Alice AF, Dowdell AK, Piening BD, Crittenden MR, Gough MJ. Myeloid MyD88 restricts CD8 + T cell response to radiation therapy in pancreatic cancer. Sci Rep 2023; 13:8634. [PMID: 37244938 PMCID: PMC10224952 DOI: 10.1038/s41598-023-35834-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Radiation therapy induces immunogenic cell death in cancer cells, whereby released endogenous adjuvants are sensed by immune cells to direct adaptive immune responses. TLRs expressed on several immune subtypes recognize innate adjuvants to direct downstream inflammatory responses in part via the adapter protein MyD88. We generated Myd88 conditional knockout mice to interrogate its contribution to the immune response to radiation therapy in distinct immune populations in pancreatic cancer. Surprisingly, Myd88 deletion in Itgax (CD11c)-expressing dendritic cells had little discernable effects on response to RT in pancreatic cancer and elicited normal T cell responses using a prime/boost vaccination strategy. Myd88 deletion in Lck-expressing T cells resulted in similar or worsened responses to radiation therapy compared to wild-type mice and lacked antigen-specific CD8+ T cell responses from vaccination, similar to observations in Myd88-/- mice. Lyz2-specific loss of Myd88 in myeloid populations rendered tumors more susceptible to radiation therapy and elicited normal CD8+ T cell responses to vaccination. scRNAseq in Lyz2-Cre/Myd88fl/fl mice revealed gene signatures in macrophages and monocytes indicative of enhanced type I and II interferon responses, and improved responses to RT were dependent on CD8+ T cells and IFNAR1. Together, these data implicate MyD88 signaling in myeloid cells as a critical source of immunosuppression that hinders adaptive immune tumor control following radiation therapy.
Collapse
Affiliation(s)
- Terry R Medler
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Tiffany C Blair
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Alejandro F Alice
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Brian D Piening
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA
- The Oregon Clinic, Portland, OR, USA
| | - Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan Street, Suite 2N100, Portland, OR, 97213, USA.
| |
Collapse
|
6
|
Hiratsuka M, Hasebe T, Ichinose Y, Sakakibara A, Fujimoto A, Wakui N, Shibasaki S, Hirasaki M, Yasuda M, Nukui A, Shimada H, Yokogawa H, Matsuura K, Hojo T, Osaki A, Saeki T. Tumor budding and fibrotic focus-proposed grading system for tumor budding in invasive carcinoma no special type of the breast. Virchows Arch 2022; 481:161-190. [PMID: 35695928 PMCID: PMC9343319 DOI: 10.1007/s00428-022-03337-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 01/04/2023]
Abstract
Tumor budding grade is a very useful histological prognostic indicator for colorectal cancer patients. Recently, it has been also reported as a significant prognostic indicator in invasive breast carcinoma patients. Our group and others have previously reported that the presence of a fibrotic focus in the tumor is a very useful histological finding for accurately predicting the prognosis in patients with invasive carcinoma of no special type (ICNST) of the breast. The purpose of the present study was to investigate whether a grading system incorporating tumor budding in a fibrotic focus is superior to the conventional grading system for tumor budding to accurately predict outcomes in patients with ICNST. According to our new grading system, we classified the tumors into grade I (164 cases), grade II (581 cases), and grade III (110 cases), and the results clearly demonstrated the significant superiority of the new grading system over that of conventional tumor budding alone for accurately predicting outcomes in patients with ICNST. Our findings strongly suggest that tumor cells and tumor-stromal cells interaction play very important roles in tumor progression rather than tumor cells alone.
Collapse
Affiliation(s)
- Miyuki Hiratsuka
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Takahiro Hasebe
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Yuki Ichinose
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Ayaka Sakakibara
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Akihiro Fujimoto
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Noriko Wakui
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Satomi Shibasaki
- Community Health Science Center, Saitama Medical University, 29, Morohongou, Moroyama Town, Iruma district, Saitama 350-0495 Japan
| | - Masataka Hirasaki
- Department of Clinical Cancer Genomics, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Akemi Nukui
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Hiroko Shimada
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Hideki Yokogawa
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Kazuo Matsuura
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Takashi Hojo
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Akihiko Osaki
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| | - Toshiaki Saeki
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, Saitama 350-1298 Japan
| |
Collapse
|
7
|
Lamminaho M, Kujala J, Peltonen H, Tengström M, Kosma VM, Mannermaa A. High Cell-Free DNA Integrity Is Associated with Poor Breast Cancer Survival. Cancers (Basel) 2021; 13:cancers13184679. [PMID: 34572906 PMCID: PMC8467852 DOI: 10.3390/cancers13184679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary A recent point of focus in breast cancer (BC) research has been the utilization of cell-free DNA and its concentration (cfDConc) and integrity (cfDI) as potential biomarkers. Though the association of cfDConc and BC survival is already recognized, studies on the prognostic value of cfDI have had contradictory results. The aim of this study was to investigate the prognostic potential of cfDConc and cfDI in Eastern Finnish BC cases with a non-metastatic disease. While the prognostic value of cfDConc remained non-significant in our analyses, high cfDI was an independent prognostic factor for poor overall survival (OS) and breast cancer-specific survival (BCSS). Inclusion of cfDI in the multivariate logistic regression model improved the predictive performance of the model, thus suggesting that the combined use of traditional tumor features and liquid biopsy could help to discriminate BC patients with poor OS and BCSS more accurately at the time of diagnosis. Abstract Background: A recent point of focus in breast cancer (BC) research has been the utilization of cell-free DNA (cfDNA) and its concentration (cfDConc) and integrity (cfDI) as potential biomarkers. Though the association of cfDConc and poor survival is already recognized, studies on the prognostic value of cfDI have had contradictory results. Here, we provide further evidence to support the use of cfDI as a potential biomarker. Methods: We selected 204 Eastern Finnish BC cases with non-metastatic disease and isolated cfDNA from the serum collected at the time of diagnosis before any treatment was given. The cfDConc and cfDI were measured with a fluorometer and electrophoresis and analyzed with 25 years of survival data. Results: High cfDConc was not an independent prognostic factor in our analyses while high cfDI was found to be an independent prognostic factor for poor OS (p = 0.020, hazard ratio (HR) = 1.57, 95% confidence interval (CI) 1.07–2.29, Cox) and BCSS (p = 0.006, HR = 1.93, 95% CI 1.21–3.08)). Inclusion of cfDI in the multivariate logistic regression model improved the predictive performance. Conclusions: Our results show high cfDI is an independent prognostic factor for poor OS and BCSS and improves the predictive performance of logistic regression models, thus supporting its prognostic potential.
Collapse
Affiliation(s)
- Maria Lamminaho
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.L.); (J.K.); (H.P.); (V.-M.K.)
| | - Jouni Kujala
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.L.); (J.K.); (H.P.); (V.-M.K.)
| | - Hanna Peltonen
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.L.); (J.K.); (H.P.); (V.-M.K.)
| | - Maria Tengström
- Cancer Center, Kuopio University Hospital, FI-70029 Kuopio, Finland;
| | - Veli-Matti Kosma
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.L.); (J.K.); (H.P.); (V.-M.K.)
- Department of Clinical Pathology, Kuopio University Hospital, FI-70029 Kuopio, Finland
- Multidisciplinary Cancer Research Community (RC Cancer), University of Eastern Finland, FI-70211 Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, FI-70029 Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (M.L.); (J.K.); (H.P.); (V.-M.K.)
- Multidisciplinary Cancer Research Community (RC Cancer), University of Eastern Finland, FI-70211 Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, FI-70029 Kuopio, Finland
- Correspondence:
| |
Collapse
|
8
|
Li JJ, Tsang JY, Tse GM. Tumor Microenvironment in Breast Cancer-Updates on Therapeutic Implications and Pathologic Assessment. Cancers (Basel) 2021; 13:cancers13164233. [PMID: 34439387 PMCID: PMC8394502 DOI: 10.3390/cancers13164233] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment (TME) in breast cancer comprises local factors, cancer cells, immune cells and stromal cells of the local and distant tissues. The interaction between cancer cells and their microenvironment plays important roles in tumor proliferation, propagation and response to therapies. There is increasing research in exploring and manipulating the non-cancerous components of the TME for breast cancer treatment. As the TME is now increasingly recognized as a treatment target, its pathologic assessment has become a critical component of breast cancer management. The latest WHO classification of tumors of the breast listed stromal response pattern/fibrotic focus as a prognostic factor and includes recommendations on the assessment of tumor infiltrating lymphocytes and PD-1/PD-L1 expression, with therapeutic implications. This review dissects the TME of breast cancer, describes pathologic assessment relevant for prognostication and treatment decision, and details therapeutic options that interacts with and/or exploits the TME in breast cancer.
Collapse
Affiliation(s)
| | | | - Gary M. Tse
- Correspondence: ; Tel.: 852-3505-2359; Fax: 852-2637-4858
| |
Collapse
|
9
|
The Influence of the Exclusion of Central Necrosis on [ 18F]FDG PET Radiomic Analysis. Diagnostics (Basel) 2021; 11:diagnostics11071296. [PMID: 34359379 PMCID: PMC8304274 DOI: 10.3390/diagnostics11071296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Central necrosis can be detected on [18F]FDG PET/CT as a region with little to no tracer uptake. Currently, there is no consensus regarding the inclusion of regions of central necrosis during volume of interest (VOI) delineation for radiomic analysis. The aim of this study was to assess how central necrosis affects radiomic analysis in PET. Methods: Forty-three patients, either with non-small cell lung carcinomas (NSCLC, n = 12) or with pheochromocytomas or paragangliomas (PPGL, n = 31), were included retrospectively. VOIs were delineated with and without central necrosis. From all VOIs, 105 radiomic features were extracted. Differences in radiomic features between delineation methods were assessed using a paired t-test with Benjamini–Hochberg multiple testing correction. In the PPGL cohort, performances of the radiomic models to predict the noradrenergic biochemical profile were assessed by comparing the areas under the receiver operating characteristic curve (AUC) for both delineation methods. Results: At least 65% of the features showed significant differences between VOIvital-tumour and VOIgross-tumour (65%, 79% and 82% for the NSCLC, PPGL and combined cohort, respectively). The AUCs of the radiomic models were not significantly different between delineation methods. Conclusion: In both tumour types, almost two-third of the features were affected, demonstrating that the impact of whether or not to include central necrosis in the VOI on the radiomic feature values is significant. Nevertheless, predictive performances of both delineation methods were comparable. We recommend that radiomic studies should report whether or not central necrosis was included during delineation.
Collapse
|
10
|
Noortman WA, Vriens D, Mooij CDY, Slump CH, Aarntzen EH, van Berkel A, Timmers HJLM, Bussink J, Meijer TWH, de Geus-Oei LF, van Velden FHP. The Influence of the Exclusion of Central Necrosis on [ 18F]FDG PET Radiomic Analysis. DIAGNOSTICS (BASEL, SWITZERLAND) 2021. [PMID: 34359379 DOI: 10.3390/diagnostics] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Central necrosis can be detected on [18F]FDG PET/CT as a region with little to no tracer uptake. Currently, there is no consensus regarding the inclusion of regions of central necrosis during volume of interest (VOI) delineation for radiomic analysis. The aim of this study was to assess how central necrosis affects radiomic analysis in PET. METHODS Forty-three patients, either with non-small cell lung carcinomas (NSCLC, n = 12) or with pheochromocytomas or paragangliomas (PPGL, n = 31), were included retrospectively. VOIs were delineated with and without central necrosis. From all VOIs, 105 radiomic features were extracted. Differences in radiomic features between delineation methods were assessed using a paired t-test with Benjamini-Hochberg multiple testing correction. In the PPGL cohort, performances of the radiomic models to predict the noradrenergic biochemical profile were assessed by comparing the areas under the receiver operating characteristic curve (AUC) for both delineation methods. RESULTS At least 65% of the features showed significant differences between VOIvital-tumour and VOIgross-tumour (65%, 79% and 82% for the NSCLC, PPGL and combined cohort, respectively). The AUCs of the radiomic models were not significantly different between delineation methods. CONCLUSION In both tumour types, almost two-third of the features were affected, demonstrating that the impact of whether or not to include central necrosis in the VOI on the radiomic feature values is significant. Nevertheless, predictive performances of both delineation methods were comparable. We recommend that radiomic studies should report whether or not central necrosis was included during delineation.
Collapse
Affiliation(s)
- Wyanne A Noortman
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Dennis Vriens
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Charlotte D Y Mooij
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Technical Medicine, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Cornelis H Slump
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Erik H Aarntzen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Anouk van Berkel
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Henri J L M Timmers
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Tineke W H Meijer
- Department of Radiation Oncology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- TechMed Centre, University of Twente, 7522 NB Enschede, The Netherlands
| | - Floris H P van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
11
|
Medler TR, Blair TC, Crittenden MR, Gough MJ. Defining Immunogenic and Radioimmunogenic Tumors. Front Oncol 2021; 11:667075. [PMID: 33816320 PMCID: PMC8017281 DOI: 10.3389/fonc.2021.667075] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
In the cancer literature tumors are inconsistently labeled as ‘immunogenic’, and experimental results are occasionally dismissed since they are only tested in known ‘responsive’ tumor models. The definition of immunogenicity has moved from its classical definition based on the rejection of secondary tumors to a more nebulous definition based on immune infiltrates and response to immunotherapy interventions. This review discusses the basis behind tumor immunogenicity and the variation between tumor models, then moves to discuss how these principles apply to the response to radiation therapy. In this way we can identify radioimmunogenic tumor models that are particularly responsive to immunotherapy only when combined with radiation, and identify the interventions that can convert unresponsive tumors so that they can also respond to these treatments.
Collapse
Affiliation(s)
- Terry R Medler
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States
| | - Tiffany C Blair
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,Molecular Microbiology and Immunology, OHSU, Portland, OR, United States
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,Molecular Microbiology and Immunology, OHSU, Portland, OR, United States.,The Oregon Clinic, Portland, OR, United States
| | - Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,Molecular Microbiology and Immunology, OHSU, Portland, OR, United States
| |
Collapse
|
12
|
Wei T, Zhang XF, Bagante F, Ratti F, Marques HP, Silva S, Soubrane O, Lam V, Poultsides GA, Popescu I, Grigorie R, Alexandrescu S, Martel G, Workneh A, Guglielmi A, Hugh T, Aldrighetti L, Endo I, Pawlik TM. Tumor Necrosis Impacts Prognosis of Patients Undergoing Curative-Intent Hepatocellular Carcinoma. Ann Surg Oncol 2020; 28:797-805. [PMID: 33249525 DOI: 10.1245/s10434-020-09390-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The impact of tumor necrosis relative to prognosis among patients undergoing curative-intent resection for hepatocellular carcinoma (HCC) remains ill-defined. METHODS Patients who underwent curative-intent resection for HCC without any prior treatment between 2000 and 2017 were identified from an international multi-institutional database. Tumor necrosis was graded as absent, moderate (< 50% area), or extensive (≥ 50% area) on histological examination. The relationship between tumor necrosis, clinicopathologic characteristics, and long-term survival were analyzed. RESULTS Among 919 patients who underwent curative-intent resection for HCC, the median tumor size was 5.0 cm (IQR, 3.0-8.5). Tumor necrosis was present in 367 (39.9%) patients (no necrosis: n = 552, 60.1% vs < 50% necrosis: n = 256, 27.9% vs ≥ 50% necrosis: n = 111, 12.1%). Extent of tumor necrosis was also associated with more advanced tumor characteristics. HCC necrosis was associated with OS (median OS: no necrosis, 84.0 months vs < 50% necrosis, 73.6 months vs ≥ 50% necrosis: 59.3 months; p < 0.001) and RFS (median RFS: no necrosis, 49.6 months vs < 50% necrosis, 38.3 months vs ≥ 50% necrosis: 26.5 months; p < 0.05). Patients with T1 tumors with extensive ≥ 50% necrosis had an OS comparable to patients with T2 tumors (median OS, 62.9 vs 61.8 months; p = 0.645). In addition, patients with T2 disease with necrosis had long-term outcomes comparable to patients with T3 disease (median OS, 61.8 vs 62.4 months; p = 0.713). CONCLUSION Tumor necrosis was associated with worse OS and RFS, as well as T-category upstaging of patients. A modified AJCC T classification that incorporates tumor necrosis should be considered in prognostic stratification of HCC patients.
Collapse
Affiliation(s)
- Tao Wei
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Surgery, Division of Surgical Oncology, Professor of Surgery, Oncology, Health Services Management and Policy, 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
| | - Fabio Bagante
- Department of Surgery, Division of Surgical Oncology, Professor of Surgery, Oncology, Health Services Management and Policy, 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.,Department of Surgery, University of Verona, Verona, Italy
| | | | - Hugo P Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Silvia Silva
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Olivier Soubrane
- Department of Hepatobiliopancreatic Surgery, APHP, Beaujon Hospital, Clichy, France
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Sydney, Australia
| | | | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | - Razvan Grigorie
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | | | | | - Aklile Workneh
- Department of Surgery, University of Ottawa, Ottawa, Canada
| | | | - Tom Hugh
- Department of Surgery, School of Medicine, The University of Sydney, Sydney, Australia
| | | | - Itaru Endo
- Yokohama City University School of Medicine, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, Professor of Surgery, Oncology, Health Services Management and Policy, 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
|
13
|
Abdelhafez AH, Musall BC, Adrada BE, Hess K, Son JB, Hwang KP, Candelaria RP, Santiago L, Whitman GJ, Le-Petross HT, Moseley TW, Arribas E, Lane DL, Scoggins ME, Leung JWT, Mahmoud HS, White JB, Ravenberg EE, Litton JK, Valero V, Wei P, Thompson AM, Moulder SL, Pagel MD, Ma J, Yang WT, Rauch GM. Tumor necrosis by pretreatment breast MRI: association with neoadjuvant systemic therapy (NAST) response in triple-negative breast cancer (TNBC). Breast Cancer Res Treat 2020; 185:1-12. [PMID: 32920733 DOI: 10.1007/s10549-020-05917-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/01/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To determine if tumor necrosis by pretreatment breast MRI and its quantitative imaging characteristics are associated with response to NAST in TNBC. METHODS This retrospective study included 85 TNBC patients (mean age 51.8 ± 13 years) with MRI before NAST and definitive surgery during 2010-2018. Each MRI included T2-weighted, diffusion-weighted (DWI), and dynamic contrast-enhanced (DCE) imaging. For each index carcinoma, total tumor volume including necrosis (TTV), excluding necrosis (TV), and the necrosis-only volume (NV) were segmented on early-phase DCE subtractions and DWI images. NV and %NV were calculated. Percent enhancement on early and late phases of DCE and apparent diffusion coefficient were extracted from TTV, TV, and NV. Association between necrosis with pathological complete response (pCR) was assessed using odds ratio (OR). Multivariable analysis was used to evaluate the prognostic value of necrosis with T stage and nodal status at staging. Mann-Whitney U tests and area under the curve (AUC) were used to assess performance of imaging metrics for discriminating pCR vs non-pCR. RESULTS Of 39 patients (46%) with necrosis, 17 had pCR and 22 did not. Necrosis was not associated with pCR (OR, 0.995; 95% confidence interval [CI] 0.4-2.3) and was not an independent prognostic factor when combined with T stage and nodal status at staging (P = 0.46). None of the imaging metrics differed significantly between pCR and non-pCR in patients with necrosis (AUC < 0.6 and P > 0.40). CONCLUSION No significant association was found between necrosis by pretreatment MRI or the quantitative imaging characteristics of tumor necrosis and response to NAST in TNBC.
Collapse
Affiliation(s)
- Abeer H Abdelhafez
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Benjamin C Musall
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1472, Houston, TX, 77030, USA
| | - Beatriz E Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - KennethR Hess
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1411, Houston, TX, 77030, USA
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1472, Houston, TX, 77030, USA
| | - Ken-Pin Hwang
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1472, Houston, TX, 77030, USA
| | - Rosalind P Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Lumarie Santiago
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Gary J Whitman
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Huong T Le-Petross
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Tanya W Moseley
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Elsa Arribas
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Deanna L Lane
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Marion E Scoggins
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Jessica W T Leung
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Hagar S Mahmoud
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Jason B White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Elizabeth E Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1411, Houston, TX, 77030, USA
| | - Alastair M Thompson
- Department of Surgery, Baylor College of Medicine, 7200 Cambridge St., Houston, TX, 77030, USA
| | - Stacy L Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354, Houston, TX, 77030, USA
| | - Mark D Pagel
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1472, Houston, TX, 77030, USA.,Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1907, Houston, TX, 77030, USA
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1472, Houston, TX, 77030, USA
| | - Wei T Yang
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA
| | - Gaiane M Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1350, Houston, TX, 77030, USA. .,Division of Diagnostic Imaging, Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1473, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Macropinocytosis confers resistance to therapies targeting cancer anabolism. Nat Commun 2020; 11:1121. [PMID: 32111826 PMCID: PMC7048872 DOI: 10.1038/s41467-020-14928-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 02/06/2020] [Indexed: 01/07/2023] Open
Abstract
Macropinocytic cancer cells scavenge amino acids from extracellular proteins. Here, we show that consuming necrotic cell debris via macropinocytosis (necrocytosis) offers additional anabolic benefits. A click chemistry-based flux assay reveals that necrocytosis provides not only amino acids, but sugars, fatty acids and nucleotides for biosynthesis, conferring resistance to therapies targeting anabolic pathways. Indeed, necrotic cell debris allow macropinocytic breast and prostate cancer cells to proliferate, despite fatty acid synthase inhibition. Standard therapies such as gemcitabine, 5-fluorouracil (5-FU), doxorubicin and gamma-irradiation directly or indirectly target nucleotide biosynthesis, creating stress that is relieved by scavenged nucleotides. Strikingly, necrotic debris also render macropinocytic, but not non-macropinocytic, pancreas and breast cancer cells resistant to these treatments. Selective, genetic inhibition of macropinocytosis confirms that necrocytosis both supports tumor growth and limits the effectiveness of 5-FU in vivo. Therefore, this study establishes necrocytosis as a mechanism for drug resistance. Macropinocytosis allows cancer cells to cope with nutrient stress. Here, the authors use a selective, genetic approach to inhibit macropinocytosis and show that consuming necrotic cell debris via macropinocytosis—necrocytosis—affords resistance to many therapies that target biosynthesis.
Collapse
|
15
|
Is the presence of edema and necrosis on T2WI pretreatment breast MRI the key to predict pCR of triple negative breast cancer? Eur Radiol 2020; 30:3363-3370. [DOI: 10.1007/s00330-020-06662-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/10/2019] [Accepted: 01/17/2020] [Indexed: 11/26/2022]
|
16
|
Li Y, Wei Y, Tang W, Luo J, Wang M, Lin H, Guo H, Ma Y, Zhang J, Li Q. Association between the degree of fibrosis in fibrotic focus and the unfavorable clinicopathological prognostic features of breast cancer. PeerJ 2019; 7:e8067. [PMID: 31741805 PMCID: PMC6859872 DOI: 10.7717/peerj.8067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/20/2019] [Indexed: 12/22/2022] Open
Abstract
Objective To explore the association between the degree of fibrosis in fibrotic focus (FF) and the unfavorable clinicopathological prognostic features of breast cancer. Methods A total of 169 cases of breast invasive ductal carcinoma (IDC) were included in the study. Hematoxylin and eosin (H&E) staining was performed in the primary lesion of breast IDC and the degree of fibrosis in tumor-stromal FF was assessed. The association between the degree of fibrosis in FF and the well-known clinicopathologic features of breast cancer was investigated and the influence of the degree of fibrosis in FF on the survival was analyzed. Results Tumor size >2 cm (P = 0.023), vascular invasion (P = 0.011), lymphatic vessel invasion (P < 0.001) and HER-2+ (P = 0.032) were positively correlated with the degree of fibrosis in FF in breast IDC. The result of multivariate analysis showed that lymphatic vessel invasion was the only independent correlation factor of high fibrosis in FF in breast IDC (OR = 3.82, 95% CI[1.13 ∼ 12.82], P = 0.031). The Nottingham prognostic index (NPI) of high fibrosis in FF was significantly higher than that of mild and moderate fibrosis in FF in the no vascular infiltration subgroup, the no nerve infiltration subgroup, and the Luminal A subgroup (P = 0.014, 0.039, and 0.018; respectively). Conclusions The high fibrosis in FF is closely associated with the strong invasiveness and the high malignancy of breast IDC. The degree of fibrosis in FF might be considered as a very practical and meaningful pathological feature of breast cancer.
Collapse
Affiliation(s)
- Yongfu Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Department of Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Yuhan Wei
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenjun Tang
- Department of Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Jingru Luo
- Department of Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Minghua Wang
- Department of Pathology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Haifeng Lin
- Department of Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Hong Guo
- Department of Surgery, Beijing Changping District Hospital of Traditional Medicine, Beijing, China
| | - Yuling Ma
- Department of Gynecology, Beijing Changping District Hospital of Traditional Medicine, Beijing, China
| | - Jun Zhang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Yang DM, Arai TJ, Campbell JW, Gerberich JL, Zhou H, Mason RP. Oxygen-sensitive MRI assessment of tumor response to hypoxic gas breathing challenge. NMR IN BIOMEDICINE 2019; 32:e4101. [PMID: 31062902 PMCID: PMC6581571 DOI: 10.1002/nbm.4101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/16/2019] [Accepted: 03/08/2019] [Indexed: 06/09/2023]
Abstract
Oxygen-sensitive MRI has been extensively used to investigate tumor oxygenation based on the response (R2 * and/or R1 ) to a gas breathing challenge. Most studies have reported response to hyperoxic gas indicating potential biomarkers of hypoxia. Few studies have examined hypoxic gas breathing and we have now evaluated acute dynamic changes in rat breast tumors. Rats bearing syngeneic subcutaneous (n = 15) or orthotopic (n = 7) 13762NF breast tumors were exposed to a 16% O2 gas breathing challenge and monitored using blood oxygen level dependent (BOLD) R2 * and tissue oxygen level dependent (TOLD) T1 -weighted measurements at 4.7 T. As a control, we used a traditional hyperoxic gas breathing challenge with 100% O2 on a subset of the subcutaneous tumor bearing rats (n = 6). Tumor subregions identified as responsive on the basis of R2 * dynamics coincided with the viable tumor area as judged by subsequent H&E staining. As expected, R2 * decreased and T1 -weighted signal increased in response to 100% O2 breathing challenge. Meanwhile, 16% O2 breathing elicited an increase in R2 *, but divergent response (increase or decrease) in T1 -weighted signal. The T1 -weighted signal increase may signify a dominating BOLD effect triggered by 16% O2 in the relatively more hypoxic tumors, whereby the influence of increased paramagnetic deoxyhemoglobin outweighs decreased pO2 . The results emphasize the importance of combined BOLD and TOLD measurements for the correct interpretation of tumor oxygenation properties.
Collapse
Affiliation(s)
- Donghan M Yang
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Tatsuya J Arai
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - James W Campbell
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - Heling Zhou
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ralph P Mason
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
18
|
Tirada N, Aujero M, Khorjekar G, Richards S, Chopra J, Dromi S, Ioffe O. Breast Cancer Tissue Markers, Genomic Profiling, and Other Prognostic Factors: A Primer for Radiologists. Radiographics 2018; 38:1902-1920. [PMID: 30312139 DOI: 10.1148/rg.2018180047] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An understanding of prognostic factors in breast cancer is imperative for guiding patient care. Increased tumor size and more advanced nodal status are established independent prognostic factors of poor outcomes and are incorporated into the American Joint Committee on Cancer (AJCC) TNM (primary tumor, regional lymph node, distant metastasis) staging system. However, other factors including imaging findings, histologic evaluation results, and molecular findings can have a direct effect on a patient's prognosis, including risk of recurrence and relative survival. Several microarray panels for gene profiling of tumors are approved by the U.S. Food and Drug Administration and endorsed by the American Society of Clinical Oncology. This article highlights prognostic factors currently in use for individualizing and guiding breast cancer therapy and is divided into four sections. The first section addresses patient considerations, in which modifiable and nonmodifiable prognostic factors including age, race and ethnicity, and lifestyle factors are discussed. The second part is focused on imaging considerations such as multicentric and/or multifocal disease, an extensive intraductal component, and skin or chest wall involvement and their effect on treatment and prognosis. The third section is about histopathologic findings such as the grade and presence of lymphovascular invasion. Last, tumor biomarkers and tumor biology are discussed, namely hormone receptors, proliferative markers, and categorization of tumors into four recognized molecular subtypes including luminal A, luminal B, human epidermal growth factor receptor 2-enriched, and triple-negative tumors. By understanding the clinical effect of these prognostic factors, radiologists, along with a multidisciplinary team, can use these tools to achieve individualized patient care and to improve patient outcomes. ©RSNA, 2018.
Collapse
Affiliation(s)
- Nikki Tirada
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| | - Mireille Aujero
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| | - Gauri Khorjekar
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| | - Stephanie Richards
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| | - Jasleen Chopra
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| | - Sergio Dromi
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| | - Olga Ioffe
- From the Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD 21201
| |
Collapse
|
19
|
Tan W, Yang M, Yang H, Zhou F, Shen W. Predicting the response to neoadjuvant therapy for early-stage breast cancer: tumor-, blood-, and imaging-related biomarkers. Cancer Manag Res 2018; 10:4333-4347. [PMID: 30349367 PMCID: PMC6188192 DOI: 10.2147/cmar.s174435] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neoadjuvant therapy (NAT) has been used increasingly in patients with locally advanced or early-stage breast cancer. However, the accurate evaluation and prediction of response to NAT remain the great challenge. Biomarkers could prove useful to identify responders or nonresponders, or even to distinguish between early and delayed responses. These biomarkers could include markers from the tumor itself, such as versatile proteins, genes, and ribonucleic acids, various biological factors or peripheral blood cells, and clinical and pathological features. Possible predictive markers could also include multiple features from functional imaging, such as standard uptake values in positron emission tomography, apparent diffusion coefficient in magnetic resonance, or radiomics imaging biomarkers. In addition, cells that indirectly present the immune status of tumor cells and/or their host could also potentially be used as biomarkers, eg, tumor-infiltrating lymphocytes, tumor-associated macrophages, and myeloid-derived suppressor cells. Though numerous biomarkers have been widely investigated, only estrogen and/or progesterone receptors and human epidermal growth factor receptor have been proven to be reliable biomarkers to predict the response to NAT. They are the only biomarkers recommended in several international guidelines. The other aforementioned biomarkers warrant further validation studies. Some multigene profiling assays that are commercially available, eg, Oncotype DX and MammaPrint, should be used with caution when extrapolated to NAT settings. A panel of combined multilevel biomarkers might be able to predict the response to NAT more robustly than individual biomarkers. To establish such a panel and its prediction model, reliable methods and extensive clinical validation are warranted.
Collapse
Affiliation(s)
- Wenyong Tan
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, People's Republic of China, ;
- Clinical Medical Research Center, The Second Clinical Medical College (Shenzhen People Hospital), Jinan University, Shenzhen, People's Republic of China,
| | - Ming Yang
- Shenzhen Jingmai Medical Scientific and Technique Company, Shenzhen, People's Republic of China
| | - Hongli Yang
- Clinical Medical Research Center, The Second Clinical Medical College (Shenzhen People Hospital), Jinan University, Shenzhen, People's Republic of China,
| | - Fangbin Zhou
- Clinical Medical Research Center, The Second Clinical Medical College (Shenzhen People Hospital), Jinan University, Shenzhen, People's Republic of China,
| | - Weixi Shen
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, People's Republic of China, ;
| |
Collapse
|
20
|
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: 4.5] [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
|
21
|
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: 16] [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
|
22
|
Brooks D, Zimmer A, Wakefield L, Lyle LT, Difilippantonio S, Tucci FC, Illiano S, Annunziata CM, Steeg PS. Limited fibrosis accompanies triple-negative breast cancer metastasis in multiple model systems and is not a preventive target. Oncotarget 2018; 9:23462-23481. [PMID: 29805748 PMCID: PMC5955109 DOI: 10.18632/oncotarget.25231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/04/2018] [Indexed: 12/16/2022] Open
Abstract
The lysophosphatidic acid receptor 1 (LPAR1) is mechanistically implicated in both tumor metastasis and tissue fibrosis. Previously, metastasis was increased when fulminant fibrosis was first induced in mice, suggesting a direct connection between these processes. The current report examined the extent of metastasis-induced fibrosis in breast cancer model systems, and tested the metastasis preventive efficacy and fibrosis attenuation of antagonists for LPAR1 and Idiopathic Pulmonary Fibrosis (IPF) in breast and ovarian cancer models. Staining analysis demonstrated only focal, low-moderate levels of fibrosis in lungs from eleven metastasis model systems. Two orally available LPAR1 antagonists, SAR100842 and EPGN9878, significantly inhibited breast cancer motility to LPA in vitro. Both compounds were negative for metastasis prevention and failed to reduce fibrosis in the experimental MDA-MB-231T and spontaneous murine 4T1 in vivo breast cancer metastasis models. SAR100842 demonstrated only occasional reductions in invasive metastases in the SKOV3 and OVCAR5 ovarian cancer experimental metastasis models. Two approved drugs for IPF, nintedanib and pirfenidone, were investigated. Both were ineffective at preventing MDA-MB-231T metastasis, with no attenuation of fibrosis. In summary, metastasis-induced fibrosis is only a minor component of metastasis in untreated progressive breast cancer. LPAR1 antagonists, despite in vitro evidence of specificity and efficacy, were ineffective in vivo as oral agents, as were approved IPF drugs. The data argue against LPAR1 and fibrosis as monotherapy targets for metastasis prevention in triple-negative breast cancer and ovarian cancer.
Collapse
Affiliation(s)
- Danielle Brooks
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alexandra Zimmer
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Lalage Wakefield
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - L. Tiffany Lyle
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | - Christina M. Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Patricia S. Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
23
|
Dolka I, Czopowicz M, Gruk-Jurka A, Wojtkowska A, Sapierzyński R, Jurka P. Diagnostic efficacy of smear cytology and Robinson's cytological grading of canine mammary tumors with respect to histopathology, cytomorphometry, metastases and overall survival. PLoS One 2018; 13:e0191595. [PMID: 29360854 PMCID: PMC5779680 DOI: 10.1371/journal.pone.0191595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 01/08/2018] [Indexed: 01/28/2023] Open
Abstract
Cytology is a simple, rapid, and inexpensive method used for pre-operative diagnosis of canine mammary tumors (CMTs) in veterinary practice. Studies related to human breast cancer showed the Robinson’s grading system—established for invasive ductal carcinoma, not otherwise specified (IDC, NOS) and used on cytological material—to not only closely correspond to the histopathological grading but also be helpful in assessing prognosis and selecting most suitable treatments before surgery. The objectives of this study were: to evaluate the accuracy of cytological diagnosis and cytological Robinson’s grading system compared to the histopathological examination of CMTs; to compare of cytological features and cytomorphometric parameters with tumor behavior, as well as cytological and histological grading; and to determine an association of the Robinson’s grading system and cytological background details with metastases, and patients’ survival. We report substantial diagnostic accuracy in detecting simple types and high grade tumors. Cytological diagnosis of tumor behavior showed relatively low sensitivity and specificity compared to human studies, and this might be caused by the heterogeneous morphology of CMTs. The presence of mucosecretory material and extracellular matrix was not significantly associated with tumor behavior. We report a positive correlation between both grading systems and cytological features (included in Robinson’s grading), the presence of necrotic debris, inflammation, and red blood cells. A negative correlation was determined only for the presence of extracellular matrix. The univariate and multivariate analyses confirmed a significantly higher risk of developing metastasis and shorter overall survival for dogs with tumors of grade 2 or 3 on cytology. In addition, these tumors were the most common cause of CMT-related deaths in dogs. Taken together, our findings suggest that the Robinson’s method of cytological grading applied for malignant CMTs evaluated in cytological smears regardless of tumor type can be adapted to veterinary cytology. Additionally, some background features seem to aid malignancy assessment.
Collapse
Affiliation(s)
- Izabella Dolka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Warsaw, Poland
- * E-mail:
| | - Michał Czopowicz
- Laboratory of Veterinary Epidemiology and Economics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Warsaw, Poland
| | - Anna Gruk-Jurka
- Department of Small Animal Diseases with Clinic, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW),Warsaw, Poland
| | - Agata Wojtkowska
- Department of Small Animal Diseases with Clinic, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW),Warsaw, Poland
| | - Rafał Sapierzyński
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Warsaw, Poland
| | - Piotr Jurka
- Department of Small Animal Diseases with Clinic, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (SGGW),Warsaw, Poland
| |
Collapse
|
24
|
Abstract
PURPOSE Recently we showed that a number of carboxylated near-infrared fluorescent (NIRF) cyanine dyes possess strong necrosis avid properties in vitro as well as in different mouse models of spontaneous and therapy-induced tumor necrosis, indicating their potential use for cancer diagnostic- and prognostic purposes. In the previous study, the detection of the cyanines was achieved by whole body optical imaging, a technique that, due to the limited penetration of near-infrared light, is not suitable for investigations deeper than 1 cm within the human body. Therefore, in order to facilitate clinical translation, the purpose of the present study was to generate a necrosis avid cyanine-based NIRF probe that could also be used for single photon emission computed tomography (SPECT). For this, the necrosis avid NIRF cyanine HQ4 was radiolabeled with 111indium, via the chelate diethylene triamine pentaacetic acid (DTPA). PROCEDURES The necrosis avid properties of the radiotracer [111In]DTPA-HQ4 were examined in vitro and in vivo in different breast tumor models in mice using SPECT and optical imaging. Moreover, biodistribution studies were performed to examine the pharmacokinetics of the probe in vivo. RESULTS Using optical imaging and radioactivity measurements, in vitro, we showed selective accumulation of [111In]DTPA-HQ4 in dead cells. Using SPECT and in biodistribution studies, the necrosis avidity of the radiotracer was confirmed in a 4T1 mouse breast cancer model of spontaneous tumor necrosis and in a MCF-7 human breast cancer model of chemotherapy-induced tumor necrosis. CONCLUSIONS The radiotracer [111In]DTPA-HQ4 possessed strong and selective necrosis avidity in vitro and in various mouse models of tumor necrosis in vivo, indicating its potential to be clinically applied for diagnostic purposes and to monitor anti-cancer treatment efficacy.
Collapse
|
25
|
Shimada H, Hasebe T, Sugiyama M, Shibasaki S, Sugitani I, Ueda S, Gotoh Y, Yasuda M, Arai E, Osaki A, Saeki T. Fibrotic focus: An important parameter for accurate prediction of a high level of tumor-associated macrophage infiltration in invasive ductal carcinoma of the breast. Pathol Int 2017; 67:331-341. [PMID: 28590017 DOI: 10.1111/pin.12550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/22/2017] [Indexed: 01/11/2023]
Abstract
Our group and others have previously reported that a fibrotic focus is a very useful histological factor for the accurate prediction of the outcome of patients with invasive ductal carcinoma of the breast. We classified 258 cases of invasive ductal carcinoma into those with and those without a fibrotic focus to investigate whether the presence of a fibrotic focus was significantly associated with the degree of tumor-associated macrophage (CD68, CD163 or CD204-positive) infiltration or whether the presence of tumor-associated macrophage infiltration heightened the malignant potential of invasive ductal carcinoma with a fibrotic focus. Multiple regression analyses demonstrated that a fibrotic focus was the only factor that was significantly associated with a high level of CD68-, CD163- or CD204-positive tumor-associated macrophage infiltration. The combined assessment of the presence or absence of a fibrotic focus and a high or a low level of CD204-positive tumor-associated macrophage infiltration clearly demonstrated that CD204-positive tumor-associated macrophage infiltration had a significant prognostic power only for patients with invasive ductal carcinoma with a fibrotic focus in multivariate analyses; CD204-positive tumor-associated macrophages might only exert a significant effect on tumor progression when a fibrotic focus is present within the invasive ductal carcinoma of the breast.
Collapse
Affiliation(s)
- Hiroko Shimada
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan.,Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Takahiro Hasebe
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Michiko Sugiyama
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan.,Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Satomi Shibasaki
- Community Health Science Center, Saitama Medical University, 29,, Morohongou, Moroyama Town, Iruma district, 350-0495, Saitama, Japan
| | - Ikuko Sugitani
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Shigeto Ueda
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Yoshiya Gotoh
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Eiichi Arai
- Department of Pathology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Akihiko Osaki
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| | - Toshiaki Saeki
- Department of Breast Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka City, 350-1298, Saitama, Japan
| |
Collapse
|
26
|
Xie B, Stammes MA, van Driel PBAA, Cruz LJ, Knol-Blankevoort VT, Löwik MAM, Mezzanotte L, Que I, Chan A, van den Wijngaard JPHM, Siebes M, Gottschalk S, Razansky D, Ntziachristos V, Keereweer S, Horobin RW, Hoehn M, Kaijzel EL, van Beek ER, Snoeks TJA, Löwik CWGM. Necrosis avid near infrared fluorescent cyanines for imaging cell death and their use to monitor therapeutic efficacy in mouse tumor models. Oncotarget 2016; 6:39036-49. [PMID: 26472022 PMCID: PMC4770755 DOI: 10.18632/oncotarget.5498] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/30/2015] [Indexed: 01/25/2023] Open
Abstract
Quantification of tumor necrosis in cancer patients is of diagnostic value as the amount of necrosis is correlated with disease prognosis and it could also be used to predict early efficacy of anti-cancer treatments. In the present study, we identified two near infrared fluorescent (NIRF) carboxylated cyanines, HQ5 and IRDye 800CW (800CW), which possess strong necrosis avidity. In vitro studies showed that both dyes selectively bind to cytoplasmic proteins of dead cells that have lost membrane integrity. Affinity for cytoplasmic proteins was confirmed using quantitative structure activity relations modeling. In vivo results, using NIRF and optoacoustic imaging, confirmed the necrosis avid properties of HQ5 and 800CW in a mouse 4T1 breast cancer tumor model of spontaneous necrosis. Finally, in a mouse EL4 lymphoma tumor model, already 24 h post chemotherapy, a significant increase in 800CW fluorescence intensity was observed in treated compared to untreated tumors. In conclusion, we show, for the first time, that the NIRF carboxylated cyanines HQ5 and 800CW possess strong necrosis avid properties in vitro and in vivo. When translated to the clinic, these dyes may be used for diagnostic or prognostic purposes and for monitoring in vivo tumor response early after the start of treatment.
Collapse
Affiliation(s)
- Bangwen Xie
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke A Stammes
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands.,In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Pieter B A A van Driel
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands
| | - Luis J Cruz
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vicky T Knol-Blankevoort
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands
| | - Martijn A M Löwik
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Laura Mezzanotte
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ivo Que
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alan Chan
- Percuros BV, Enschede, The Netherlands
| | - Jeroen P H M van den Wijngaard
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Siebes
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sven Gottschalk
- Faculty of Medicine, Technical University of Munich, Munich, Germany.,Institute for Biological and Medical Imaging, Helmholtz Center Munich, Munich, Germany
| | - Daniel Razansky
- Faculty of Medicine, Technical University of Munich, Munich, Germany.,Institute for Biological and Medical Imaging, Helmholtz Center Munich, Munich, Germany
| | - Vasilis Ntziachristos
- Faculty of Medicine, Technical University of Munich, Munich, Germany.,Institute for Biological and Medical Imaging, Helmholtz Center Munich, Munich, Germany
| | - Stijn Keereweer
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Richard W Horobin
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, The University of Glasgow, Glasgow, Scotland, UK
| | - Mathias Hoehn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands.,In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Eric L Kaijzel
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ermond R van Beek
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Medres, Cologne, Germany
| | - Thomas J A Snoeks
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clemens W G M Löwik
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
27
|
Stammes MA, Maeda A, Bu J, Scollard DA, Kulbatski I, Medeiros PJ, Sinisi R, Dubikovskaya EA, Snoeks TJA, van Beek ER, Chan AB, Löwik CWGM, DaCosta RS. The Necrosis-Avid Small Molecule HQ4-DTPA as a Multimodal Imaging Agent for Monitoring Radiation Therapy-Induced Tumor Cell Death. Front Oncol 2016; 6:221. [PMID: 27818949 PMCID: PMC5073092 DOI: 10.3389/fonc.2016.00221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/05/2016] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Most effective antitumor therapies induce tumor cell death. Non-invasive, rapid and accurate quantitative imaging of cell death is essential for monitoring early response to antitumor therapies. To facilitate this, we previously developed a biocompatible necrosis-avid near-infrared fluorescence (NIRF) imaging probe, HQ4, which was radiolabeled with 111Indium-chloride (111In-Cl3) via the chelate diethylene triamine pentaacetic acid (DTPA), to enable clinical translation. The aim of the present study was to evaluate the application of HQ4-DTPA for monitoring tumor cell death induced by radiation therapy. Apart from its NIRF and radioactive properties, HQ4-DTPA was also tested as a photoacoustic imaging probe to evaluate its performance as a multimodal contrast agent for superficial and deep tissue imaging. MATERIALS AND METHODS Radiation-induced tumor cell death was examined in a xenograft mouse model of human breast cancer (MCF-7). Tumors were irradiated with three fractions of 9 Gy each. HQ4-DTPA was injected intravenously after the last irradiation, NIRF and photoacoustic imaging of the tumors were performed at 12, 20, and 40 h after injection. Changes in probe accumulation in the tumors were measured in vivo, and ex vivo histological analysis of excised tumors was performed at experimental endpoints. In addition, biodistribution of radiolabeled [111In]DTPA-HQ4 was assessed using hybrid single-photon emission computed tomography-computed tomography (SPECT-CT) at the same time points. RESULTS In vivo NIRF imaging demonstrated a significant difference in probe accumulation between control and irradiated tumors at all time points after injection. A similar trend was observed using in vivo photoacoustic imaging, which was validated by ex vivo tissue fluorescence and photoacoustic imaging. Serial quantitative radioactivity measurements of probe biodistribution further demonstrated increased probe accumulation in irradiated tumors. CONCLUSION HQ4-DTPA has high specificity for dead cells in vivo, potentiating its use as a contrast agent for determining the relative level of tumor cell death following radiation therapy using NIRF, photoacoustic imaging and SPECT in vivo. Initial preclinical results are promising and indicate the need for further evaluation in larger cohorts. If successful, such studies may help develop a new multimodal method for non-invasive and dynamic deep tissue imaging of treatment-induced cell death to quantitatively assess therapeutic response in patients.
Collapse
Affiliation(s)
- Marieke A. Stammes
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Percuros BV, Enschede, Netherlands
| | - Azusa Maeda
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jiachuan Bu
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | | | - Iris Kulbatski
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Philip J. Medeiros
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Riccardo Sinisi
- LCBIM, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Elena A. Dubikovskaya
- LCBIM, Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Thomas J. A. Snoeks
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Ermond R. van Beek
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Ralph S. DaCosta
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Techna Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
28
|
de Mascarel I, Debled M, Brouste V, Mauriac L, Sierankowski G, Velasco V, Croce S, Chibon F, Boudeau J, Debant A, MacGrogan G. Comprehensive prognostic analysis in breast cancer integrating clinical, tumoral, micro-environmental and immunohistochemical criteria. SPRINGERPLUS 2015; 4:528. [PMID: 26405647 PMCID: PMC4576021 DOI: 10.1186/s40064-015-1297-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 09/01/2015] [Indexed: 12/31/2022]
Abstract
Significant morphological, clinical and biological prognostic factors vary according to molecular subtypes of breast tumors, yet comprehensive analysis of such factors linked to survival in each group is lacking. Clinicopathological and micro-environmental criteria, estrogen (ER), progesterone (PR) receptors, HER2, Ki67, basal markers, CD24, CD44, ALDH1, BCL2, E-Cadherin and Trio were assessed in 1070 primary operable breast cancers from a single center according to five main molecular subtypes and associations with distant metastasis-free survival (DMFS) were examined. There were 682 (64 %) luminal A (LA), 166 (16 %) Luminal B HER2 negative (LBH−), 47 (4 %) Luminal B HER2 positive (LBH+), 108 (10 %) triple negative (TN) and 67 (6 %) HER2-enriched tumors (H2+). Median follow-up was 13.7 years. At 5 years, DMFS in LA (90 %) was better than in LBH− (80.9 %), hazard ratio (HR) = 2.22 [1.44–3.43] P < 0.001; LBH+ (74.5 %), HR = 3.14 [1.69–5.84] P < 0.001, TN (71.5 %) HR = 3.63 [2.34–5.63], P < 0.001; and H2+ (65.2 %), HR = 4.69 [2.90–7.59], P < 0.001. In multivariable analysis, factors associated with shorter DMFS varied according to molecular subtype, with tumor size being associated with shorter DMFS in the LBH−, LBH+ and TN groups and the Rho GEF Trio and BCL2 phenotypes in TN tumors only. These findings help to define new clinicophenotypic models and to identify new therapeutic strategies in the specific molecular subgroups.
Collapse
Affiliation(s)
- Isabelle de Mascarel
- Department of BioPathology, Institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France
| | - Marc Debled
- Department of Medical Oncology, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| | - Véronique Brouste
- Clinical and Epidemiological Research Unit, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| | - Louis Mauriac
- Department of Medical Oncology, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| | - Ghislaine Sierankowski
- Department of BioPathology, Institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France
| | - Valérie Velasco
- Department of BioPathology, Institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France
| | - Sabrina Croce
- Department of BioPathology, Institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) U916, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| | - Frédéric Chibon
- Department of BioPathology, Institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) U916, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| | - Jêrome Boudeau
- Centre de Recherche en Biochimie Macromoléculaire, CRBM-CNRS UMR 5237, Universités Montpellier I et II, Montpellier, France
| | - Anne Debant
- Centre de Recherche en Biochimie Macromoléculaire, CRBM-CNRS UMR 5237, Universités Montpellier I et II, Montpellier, France
| | - Gaëtan MacGrogan
- Department of BioPathology, Institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux, France ; Institut National de la Santé et de la Recherche Médicale (INSERM) U916, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| |
Collapse
|
29
|
Onishi N, Kanao S, Kataoka M, Iima M, Sakaguchi R, Kawai M, Kataoka TR, Mikami Y, Toi M, Togashi K. Apparent diffusion coefficient as a potential surrogate marker for Ki-67 index in mucinous breast carcinoma. J Magn Reson Imaging 2014; 41:610-5. [DOI: 10.1002/jmri.24615] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/17/2014] [Indexed: 11/11/2022] Open
Affiliation(s)
- Natsuko Onishi
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Shotaro Kanao
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Masako Kataoka
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Mami Iima
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Rena Sakaguchi
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Makiko Kawai
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Tatsuki R. Kataoka
- Department of Diagnostic Pathology; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Yoshiki Mikami
- Department of Diagnostic Pathology; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Masakazu Toi
- Department of Breast Surgery; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine; Kyoto University Graduate School of Medicine; Kyoto Japan
| |
Collapse
|
30
|
Sung JS, Jochelson MS, Brennan S, Joo S, Wen YH, Moskowitz C, Zheng J, Dershaw DD, Morris EA. MR imaging features of triple-negative breast cancers. Breast J 2013; 19:643-9. [PMID: 24015869 DOI: 10.1111/tbj.12182] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Triple-negative (TN) breast cancers, which are associated with a more aggressive clinical course and poorer prognosis, often present with benign imaging features on mammography and ultrasound. The purpose of this study was to compare the magnetic resonance imaging features of TN breast cancers with estrogen (ER) and progesterone (PR) positive, human epidermal growth factor receptor (HER2) negative cancers. Retrospective review identified 140 patients with TN breast cancer who underwent a preoperative breast MRI between 2003 and 2008. Comparison was made to 181 patients with ER+/PR+/HER2- cancer. Breast MRIs were independently reviewed by two radiologists blinded to the pathology. Discrepancies were resolved by a third radiologist. TN cancers presented with a larger tumor size (p = 0.002), higher histologic grade (<0.001), and were more likely to be unifocal (p = 0.018) compared with ER+/PR+/HER2- tumors. MRI features associated with TN tumors included mass enhancement (p = 0.026), areas of intratumoral high T2 signal intensity (p < 0.001), lobulated shape (p < 0.001), rim enhancement (p < 0.001), and smooth margins (p = 0.005). Among the TN tumors with marked necrosis, 26% showed a large central acellular zone of necrosis.
Collapse
Affiliation(s)
- Janice S Sung
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gazinska P, Grigoriadis A, Brown JP, Millis RR, Mera A, Gillett CE, Holmberg LH, Tutt AN, Pinder SE. Comparison of basal-like triple-negative breast cancer defined by morphology, immunohistochemistry and transcriptional profiles. Mod Pathol 2013; 26:955-66. [PMID: 23392436 DOI: 10.1038/modpathol.2012.244] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/20/2012] [Accepted: 11/22/2012] [Indexed: 11/09/2022]
Abstract
Basal-like invasive breast cancer is an important clinical group because of its association with a triple-negative phenotype defined by the lack of expression of estrogen, progesterone and human epidermal growth factor receptors 2, relative lack of therapeutic options and poor prognosis. However, depending on the method used to define these lesions, morphological assessment, immunohistochemical markers or gene expression, a different set of tumors is captured. The aim of this study was to investigate the consequences of using different methodological approaches to define basal-like lesions among triple-negative breast carcinomas with regard to their clinicopathological features and patient outcome. The cohort consisted of 142 invasive breast cancers with a triple-negative receptor status. First, each was reviewed histologically and those with morphological basal-like features were characterized as 'Path-Basal'. Second, the 'Core Basal' immunohistochemical lesions, defined as cytokeratin 5/6 and/or epidermal growth factor receptor 1 positive, within the triple-negative breast cancers were identified, and third their classification based on gene expression profiling was retrieved and those in the molecular 'PAM50 basal-like' subtype recorded. A total of 116 basal-like breast cancers were identified among the 142 triple-negative breast cancers by at least one of these three classifications (80%), but only 13 samples were defined as basal-like with all three methods. None of these 13 tumors were associated with lymphovascular invasion. The 34 morphological 'Path-Basal' lesions were significantly associated with a lack of nodal metastases. Comparing the estimates of death in the three classifications, the highest risk of death was seen for the 'Core Basal' group. In this study, we highlight that the definition of basal-like breast cancer based on different methodologies varies significantly and does not identify the same lesions. This incomplete overlap of cases emphasizes the need for consistent or new approaches to improve precise identification.
Collapse
Affiliation(s)
- Patrycja Gazinska
- Breakthrough Breast Cancer Research Unit, King's College London, School of Medicine, Division of Cancer Studies, Bermondsey Wing, Guy's Hospital, London SE1 9RT, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hasebe T. Tumor-stromal interactions in breast tumor progression--significance of histological heterogeneity of tumor-stromal fibroblasts. Expert Opin Ther Targets 2013; 17:449-60. [PMID: 23297753 DOI: 10.1517/14728222.2013.757305] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Existing pathological diagnostic protocols for breast cancer do not fully reflect the biological characteristics of tumor stromata. To improve the pathological diagnosis of breast cancer, a new pathological diagnostic method capable of assessing the degree of breast cancer malignancy based on the histological features of the tumor stroma is needed. AREAS COVERED The presence of a fibrotic focus (FF), which consists of fibroblasts or collagen fibers, and the presence of atypical tumor-stromal fibroblasts are significantly associated with nodal metastasis or distant-organ metastasis in patients with invasive ductal carcinoma (IDC) of the breast. FF is the only factor that is significantly associated with an increase in tumor angiogenesis. The importance of FF and atypical tumor-stromal fibroblasts clearly indicates that the malignant potential of IDC does not depend only on the biological characteristics of the tumor cell, but also on those of the tumor stroma. EXPERT OPINION The biological characteristics of fibroblasts forming an FF or atypical tumor-stromal fibroblasts probably differ from those of fibroblasts located outside an FF or ordinary tumor-stromal fibroblasts. Thus, similar to tumor cells, the heterogeneity of tumor-stromal fibroblasts probably influences the outcome of patients with IDC of the breast.
Collapse
Affiliation(s)
- Takahiro Hasebe
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, 277-8577, Chiba, Japan.
| |
Collapse
|
33
|
Wen YH, Ho A, Patil S, Akram M, Catalano J, Eaton A, Norton L, Benezra R, Brogi E. Id4 protein is highly expressed in triple-negative breast carcinomas: possible implications for BRCA1 downregulation. Breast Cancer Res Treat 2012; 135:93-102. [PMID: 22538771 DOI: 10.1007/s10549-012-2070-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 04/16/2012] [Indexed: 01/08/2023]
Abstract
BRCA1 germline mutation carriers usually develop ER, PR and HER2 negative breast carcinoma. Somatic BRCA1 mutations are rare in sporadic breast cancers, but other mechanisms could impair BRCA1 functions in these tumors, particularly in triple-negative breast carcinomas (TNBCs). Id4, a helix-loop-helix DNA binding factor, blocks BRCA1 gene transcription in vitro and could downregulate BRCA1 in vivo. We compared Id4 immunoreactivity in 101 TNBCs versus 113 non-TNBCs, and correlated the results with tumor morphology and immunoreactivity for CK5/6, CK14, EGFR, and androgen receptor (AR). Id4 was present in 76 out of 101 (75 %) TNBCs: 40 (40 %) TNBCs displayed Id4 positivity in >50 % of neoplastic cells, 23 (23 %) in 5-50 %, and 13 (13 %) in <5 %. In contrast, only 6 (5 %) of 113 non-TNBCs showed focal Id4 positivity, limited to fewer than 5 % of the tumor (p < 0.0001). Id4 expression significantly associated with high histologic grade (p = 0.0002) and mitotic rate (p = 0.006). Id4 decorated all 12 TNBCs with large central acellular zone of necrosis in our series, with positive staining in 10-90 % of the cells. Id4 signal strongly correlated with cytokeratin CK14 reactivity (p < 0.0001), but not with CK5/6 and EGFR. All apocrine carcinomas in our series were positive for AR and most for EGFR, but they were negative for CK5/6, CK14, and Id4, with only two exceptions. Our results document substantial expression of Id4 in most TNBCs, which could result in functional downregulation of BRCA1 pathways in these tumors.
Collapse
Affiliation(s)
- Yong Hannah Wen
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Important Histologic Outcome Predictors for Patients With Invasive Ductal Carcinoma of the Breast. Am J Surg Pathol 2011; 35:1484-97. [DOI: 10.1097/pas.0b013e318224ca28] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Caruso R, Fedele F, Lucianò R, Branca G, Parisi C, Paparo D, Parisi A. Mitotic catastrophe in malignant epithelial tumors: the pathologist's viewpoint. Ultrastruct Pathol 2011; 35:66-71. [PMID: 21299346 DOI: 10.3109/01913123.2010.543753] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mitotic catastrophe is a common phenomenon occurring in tumor cells with impaired p53 function exposed to various cytotoxic and genotoxic agents. The defective p53 checkpoint causes improper segregation of chromosomes, resulting in aberrant mitosis, multiple micronuclei, multinucleate giant cells, and eventual necrosis-like death and centrosome aberration. Although various descriptions explaining mitotic catastrophe exist, there is still no generally accepted definition of this phenomenon. However, the syndrome of mitotic catastrophe may be a unifying morphological concept of particular interest to cancer research, as it integrally links cell death to checkpoints of the cell cycle. Morphological findings compatible with mitotic catastrophe may be found in pleomorphic, giant cell carcinomas--neoplasms characterized by a poor prognosis. The inclusion of mitotic catastrophe as part of the microscopic evaluation of tumors will add further insight to the pathobiology of tumor progression and in novel therapeutic designs. Finally, the possibility of assimilating mitotic catastrophe into a prognostic score is discussed.
Collapse
Affiliation(s)
- Rosario Caruso
- Department of Human Pathology, University of Messina, Messina, Italy.
| | | | | | | | | | | | | |
Collapse
|
36
|
Vandorpe T, Smeets A, Van Calster B, Van Hoorde K, Leunen K, Amant F, Moerman P, Deraedt K, Brouckaert O, Van Huffel S, Wildiers H, Christiaens MR, Neven P. Lobular and non-lobular breast cancers differ regarding axillary lymph node metastasis: a cross-sectional study on 4,292 consecutive patients. Breast Cancer Res Treat 2011; 128:429-35. [PMID: 21562708 DOI: 10.1007/s10549-011-1565-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 04/28/2011] [Indexed: 11/24/2022]
Abstract
Invasive lobular carcinoma (ILC) accounts for 8-14% of all breast cancers and carries distinct prognostic and biologic implications. The goal of our study was to investigate the impact of lobular histology on axillary lymph node (ALN) involvement. This is a cross-sectional study of 4,292 consecutive patients surgically treated for breast carcinoma at the University Hospitals Leuven. Logistic regression analysis was used to relate ILC to lymph node involvement while controlling for the following clinicopathologic features: tumor size, multifocal disease, tumor grade, lobular subtype and the combined steroid, and Her-2 status. Odds ratios (ORs) and 95% confidence intervals (CIS) were computed. A subgroup analysis was performed for patients that underwent a sentinel lymph node (SLN) procedure. The observed incidence of ILC was 13%. ILCs were larger, were more often grade II, multifocal, steroid receptor positive and Her-2 negative, and tended to be present in older patients. Incidence of ALN involvement was 42.0% for ILCs versus 38.3% for other tumors (OR 1.17, 95% CI 0.97-1.40). For the SLN subgroup, ILCs were less often ALN positive than non-ILCs (20.5% versus 28.3%, OR 0.66, 95% CI: 0.41-1.00). In the multivariable analysis, the lobular subtype was identified as less likely to have ALN involvement (adjusted OR 0.66, 95% CI 0.53-0.82). The analysis for the SLN subgroup showed comparable results (adjusted OR 0.49, 95% CI 0.30-0.78). This study has demonstrated that the lobular subtype is an independent predictor of lymph node involvement with ILC having a lower incidence of involved lymph nodes. The mildly higher incidence of ALN metastasis in lobular cancers in univariable analysis is not due to the lobular subtype, but due to confounding factors that interact with lymph node involvement.
Collapse
Affiliation(s)
- T Vandorpe
- Multidisciplinary Breast Centre, University Hospitals Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Viale G. Integrating molecular profiling, histological type and other variables: Defining the fingerprint of responsiveness to treatment. Breast 2009; 18 Suppl 3:S32-6. [DOI: 10.1016/s0960-9776(09)70269-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|