1
|
Gal S, Pavan MJ, Vidavsky N. Characterization of dolomite and calcite microcalcifications in human breast tissue. RSC Adv 2024; 14:28741-28752. [PMID: 39257663 PMCID: PMC11384324 DOI: 10.1039/d4ra04137b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
Pathological crystallization within soft tissues often yields biominerals with properties differing from those of their geological or synthetic counterparts. Microcalcifications (MCs) are abundant in breast tumors, particularly in non-invasive lesions, such as ductal carcinoma in situ (DCIS). Given the challenge of predicting DCIS progression into invasive cancer, it has been suggested that MCs can be leveraged to inform DCIS prognosis. The predominant type of breast MCs are those containing calcium phosphates (CaP), whose crystal properties are commonly held to correlate with malignancy. Less common are non-CaP minerals, which have received less attention, as they are associated mainly with benign lesions. Here, we conducted a retrospective study of tissue samples collected from patients who were originally diagnosed with DCIS and whose current medical status is known. We examined the elemental composition, morphology, and crystal phases of 398 MCs, aiming to investigate potential correlations between MC crystal properties and the progression of DCIS. Our findings revealed primarily non-CaP MCs, an observation that was likely made possible only by the tissue processing methodology employed, which did not involve harsh conditions. We found that non-CaP MCs were abundant in DCIS lesions, that they exhibited diverse morphologies and sizes, and that they were composed of calcite and dolomite. Dolomite formation in cancer has not been reported previously and may be linked to pH fluctuations in the tumor microenvironment. The small size of DCIS lesions often requires pathologists to use the entire sample, thus reducing the number of samples available for further research. Nonetheless, despite our limited sample size, the observed trend indicated an association of dolomite MCs with DCIS lesions that progressed into invasive cancer over time.
Collapse
Affiliation(s)
- Sahar Gal
- Department of Chemical Engineering, Ben-Gurion University of the Negev Beer-Sheva 8410501 Israel
| | - Mariela J Pavan
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev Beer-Sheva 8410501 Israel
| | - Netta Vidavsky
- Department of Chemical Engineering, Ben-Gurion University of the Negev Beer-Sheva 8410501 Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev Beer-Sheva 8410501 Israel
| |
Collapse
|
2
|
Li M, Chen J, Wang W, Hou C. Gastric Adenocarcinoma with Enteroblastic Differentiation in the Presence of Calcification: A Case Report and Review of the Literature. Int J Surg Pathol 2024; 32:845-850. [PMID: 37715642 DOI: 10.1177/10668969231195078] [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: 09/18/2023]
Abstract
Gastric adenocarcinoma with enteroblastic differentiation is a specific subtype of gastric cancer that is rare and highly malignant, usually presenting at an early stage with lymphovascular invasion, lymph node, and distant metastases, resulting in a poor prognosis. The pathology of this patient showed a classic tubular adenocarcinoma infiltrating into the mucosal layer, with the presence of cytoplasmic translucent tumor cells below the mucosal layer. It is noteworthy that this patient did not exhibit lymphovascular invasion, lymph node, and distant metastasis. Additionally, a large amount of calcification was observed; therefore, it remains unclear whether there exists any correlation between the two factors. To the best of our knowledge, this is the first case report demonstrating massive calcification in gastric adenocarcinoma with enteroblastic differentiation, which may have implications for future diagnosis of this rare subtype of gastric cancer.
Collapse
Affiliation(s)
- Mengyao Li
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, China
| | - Jiajun Chen
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Weihao Wang
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Chuanling Hou
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
3
|
Guergan S, Boeer B, Fugunt R, Helms G, Roehm C, Solomianik A, Neugebauer A, Nuessle D, Schuermann M, Brunecker K, Jurjut O, Boehme KA, Dammeier S, Enderle MD, Bettio S, Gonzalez-Menendez I, Staebler A, Brucker SY, Kraemer B, Wallwiener D, Fend F, Hahn M. Optical Emission Spectroscopy for the Real-Time Identification of Malignant Breast Tissue. Diagnostics (Basel) 2024; 14:338. [PMID: 38337854 PMCID: PMC10855719 DOI: 10.3390/diagnostics14030338] [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: 11/03/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Breast conserving resection with free margins is the gold standard treatment for early breast cancer recommended by guidelines worldwide. Therefore, reliable discrimination between normal and malignant tissue at the resection margins is essential. In this study, normal and abnormal tissue samples from breast cancer patients were characterized ex vivo by optical emission spectroscopy (OES) based on ionized atoms and molecules generated during electrosurgical treatment. The aim of the study was to determine spectroscopic features which are typical for healthy and neoplastic breast tissue allowing for future real-time tissue differentiation and margin assessment during breast cancer surgery. A total of 972 spectra generated by electrosurgical sparking on normal and abnormal tissue were used for support vector classifier (SVC) training. Specific spectroscopic features were selected for the classification of tissues in the included breast cancer patients. The average classification accuracy for all patients was 96.9%. Normal and abnormal breast tissue could be differentiated with a mean sensitivity of 94.8%, a specificity of 99.0%, a positive predictive value (PPV) of 99.1% and a negative predictive value (NPV) of 96.1%. For 66.6% patients all classifications reached 100%. Based on this convincing data, a future clinical application of OES-based tissue differentiation in breast cancer surgery seems to be feasible.
Collapse
Affiliation(s)
- Selin Guergan
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Bettina Boeer
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Regina Fugunt
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Gisela Helms
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Carmen Roehm
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Anna Solomianik
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Alexander Neugebauer
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Daniela Nuessle
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Mirjam Schuermann
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Kristin Brunecker
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Ovidiu Jurjut
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Karen A. Boehme
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Sascha Dammeier
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Markus D. Enderle
- Erbe Elektromedizin GmbH, Waldhoernlestr. 17, 72072 Tübingen, Germany; (A.N.); (D.N.); (M.S.); (O.J.); (K.A.B.); (S.D.); (M.D.E.)
| | - Sabrina Bettio
- Institute of Pathology and Neuropathology, Tuebingen University Hospital, 72076 Tübingen, Germany; (S.B.); (I.G.-M.); (A.S.); (F.F.)
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Tuebingen University Hospital, 72076 Tübingen, Germany; (S.B.); (I.G.-M.); (A.S.); (F.F.)
| | - Annette Staebler
- Institute of Pathology and Neuropathology, Tuebingen University Hospital, 72076 Tübingen, Germany; (S.B.); (I.G.-M.); (A.S.); (F.F.)
| | - Sara Y. Brucker
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Bernhard Kraemer
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Diethelm Wallwiener
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| | - Falko Fend
- Institute of Pathology and Neuropathology, Tuebingen University Hospital, 72076 Tübingen, Germany; (S.B.); (I.G.-M.); (A.S.); (F.F.)
| | - Markus Hahn
- Department of Women’s Health, Tuebingen University Hospital, 72076 Tübingen, Germany; (B.B.); (R.F.); (G.H.); (C.R.); (A.S.); (S.Y.B.); (B.K.); (D.W.); (M.H.)
| |
Collapse
|
4
|
Bonfiglio R, Sisto R, Casciardi S, Palumbo V, Scioli MP, Giacobbi E, Servadei F, Melino G, Mauriello A, Scimeca M. Aluminium bioaccumulation in colon cancer, impinging on epithelial-mesenchymal-transition and cell death. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168335. [PMID: 37939965 DOI: 10.1016/j.scitotenv.2023.168335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/10/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
We investigated the presence of aluminium (Al) in human colon cancer samples and its potential association with biological processes involved in cancer progression, such as epithelial to mesenchymal transition (EMT) and cell death. 25 consecutive colon samples were collected from patients undergoing colonic resection. Both neoplastic and normal mucosa were collected from each patient and subjected to histological, ultrastructural and immunohistochemical analyses. Moreover, colon samples from two Al-positive patients underwent multi-omic analyses, including whole genome sequencing and RNA sequencing (RNAseq). Morin staining, used to identify in situ aluminium bioaccumulation, showed the presence of Al in tumor areas of 24 % of patients. Transmission electron microscopy and energy-dispersive X-ray microanalysis confirmed the presence of Al specifically in intracytoplasmic electrondense nanodeposits adjacent to mitochondria of colon cancer cells. Immunohistochemical analyses for vimentin and nuclear β-catenin were performed to highlight the occurrence of the EMT phenomenon in association to Al bioaccumulation. Al-positive samples showed a significant increase in both the number of vimentin-positive and nuclear β-catenin-positive cancer cells compared to Al-negative samples. Moreover, Al-positive samples exhibited a significant decrease in the number of apoptotic cells, as well as the expression of the anti-apoptotic molecule BCL-2. Multi-omic analyses revealed a higher tumor mutational burden (TMB) in Al-positive colon cancers (n = 2) compared to a control cohort (n = 100). Additionally, somatic mutations in genes associated with EMT (GATA3) and apoptosis (TP53) were observed in Al-positive colon cancers. In conclusion, this study provides the first evidence of Al bioaccumulation in colon cancer and its potential role in modulating molecular pathways involved in cancer progression, such as EMT and apoptosis. Understanding the molecular mechanisms underlying Al toxicity might contribute to improve strategies for prevention, early detection, and targeted therapies for the management of colon cancer patients.
Collapse
Affiliation(s)
- Rita Bonfiglio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Renata Sisto
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Monte Porzio Catone, Rome 00078, Italy.
| | - Stefano Casciardi
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Monte Porzio Catone, Rome 00078, Italy.
| | - Valeria Palumbo
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Paola Scioli
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Erica Giacobbi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Francesca Servadei
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
5
|
Tian Y, Zhao L, Gui Z, Liu S, Liu C, Yu T, Zhang L. PI3K/AKT signaling activates HIF1α to modulate the biological effects of invasive breast cancer with microcalcification. NPJ Breast Cancer 2023; 9:93. [PMID: 37957150 PMCID: PMC10643473 DOI: 10.1038/s41523-023-00598-z] [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: 02/10/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Microcalcification (MC) is a valuable diagnostic indicator of breast cancer, and it is reported to be associated with increased tumor aggressiveness and poor prognosis. Nevertheless, the exact potential molecular mechanism is not completely understood. Here, we find that the mineralized invasive breast cancer (IBC) cells not only increased their proliferation and migration, but also showed the characteristic of doxorubicin resistance. The PI3K/AKT signaling pathway is associated with the generation of calcification in IBC, and it activates the transcription and translation of its downstream hypoxia-inducible factor 1α (HIF1α). Knockdown of HIF1α protein significantly downregulated cell proliferation and migration while calcification persists. Meanwhile, calcified breast cancer cells restored sensitivity to doxorubicin because of suppressed HIF1α expression. In addition, we provide initial data on the underlying value of HIF1α as a biomarker of doxorubicin resistance. These findings provide a new direction for exploring microcalcifications in IBC.
Collapse
Affiliation(s)
- Yao Tian
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Lu Zhao
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Zhengwei Gui
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Chenguang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Tianyao Yu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Lin Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China.
| |
Collapse
|
6
|
Yadav P, Makwana S, Bansal S, Soni S, Mahapatra MK, Bandyopadhayaya S, Tailor R, Shrivastava SK, Sharma LK, Mandal CC. Metformin prevents osteoblast-like potential and calcification in lung cancer A549 cells. J Biochem Mol Toxicol 2023; 37:e23454. [PMID: 37409753 DOI: 10.1002/jbt.23454] [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: 01/14/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
In spite of recent advances made in understanding its progression, cancer is still a leading cause of death across the nations. Molecular pathophysiology of these cancer cells largely differs depending on cancer types and even within the same tumor. Pathological mineralization/calcification is seen in various tissues including breast, prostate, and lung cancer. Osteoblast-like cells derived after trans-differentiation of mesenchymal cells usually drive calcium deposition in various tissues. This study aims to explore the presence of osteoblast-like potential in lung cancer cells and its prevention. ALP assay, ALP staining, nodule formation, RT-PCR, RT-qPCR, and western blot analysis experiments were carried out in lung cancer A549 cells to achieve said objective. Expressions of various osteoblast markers (e.g., ALP, OPN, RUNX2, and Osterix) along with osteoinducer genes (BMP-2 and BMP-4) were observed in A549 cells. Moreover, ALP activity and ability leading to nodule formation revealed the presence of osteoblast-like potential in lung cancer cells. Here, BMP-2 treatment increased expressions of osteoblast transcription factors such as RUNX2 and Osterix, enhanced ALP activity, and augmented calcification in this cell line. It was also observed that antidiabetic metformin inhibited BMP-2 mediated increase in osteoblast-like potential and calcification in these cancer cells. The current study noted that metformin blocked BMP-2 mediated increase in epithelial to mesenchymal transition (EMT) in A549 cells. The above findings for the first time unravel that A549 cells possess osteoblast-like potential which drives lung cancer calcification. Metformin might prevent BMP-2 induced osteoblast-like phenotype of the lung cancer cells with concomitant inhibition of EMT to inhibit lung cancer tissue calcification.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sweta Makwana
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Shivani Bansal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Manas K Mahapatra
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Rashmi Tailor
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sandeep K Shrivastava
- Centre for Innovation, Research & Development, Dr. B. Lal Clinical Laboratory Pvt Ltd., Jaipur, Rajasthan, India
| | - Lokendra K Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
7
|
Yacouba A, Bellali S, Haddad G, Mavros N, Fontanini A, Dubourg G, Lagier JC, Raoult D, Bou Khalil J. Use of scanning electron microscopy and energy dispersive X-ray for urine analysis: A preliminary investigation. Microsc Res Tech 2023; 86:1249-1257. [PMID: 36773029 DOI: 10.1002/jemt.24301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/12/2023]
Abstract
Scanning electron microscopy (SEM) and energy dispersive X-ray (EDX) are powerful tools to study the ultrastructure of numerous specimens and to determine their elemental composition, respectively. However, results have not yet been reported on their application to urine samples in routine clinical laboratory practice. Herein we investigate urine sediment by using SEM and EDX to detect and identify different urine components. A total of 206 urine samples from patients with and without urinary tract infections were analyzed using SEM and EDX. Microorganisms, crystals, epithelial cells, leukocytes, and erythrocytes were targeted in urine sediment samples. The identification of urine components was based on their morphology, size, contrast, and elemental composition. SEM-analysis allowed us to identify and classify microorganisms in urine sediments into the categories of gram-negative bacilli, cluster cocci, chain cocci, gram-negative bacilli, gram-positive bacilli, and yeasts. In addition, various types of epithelial cells such as renal, transitional, and squamous epithelial cells were found. Furthermore, leukocytes and erythrocytes were well identified, with the detection of various morphological forms of erythrocytes, such as dysmorphic and isomorphic erythrocytes. Using SEM-EDX analysis, calcium oxalate was the most frequently-identified crystal (92.0%), with prominent peaks of C, O, and Ca elements, followed by struvite (6%), with peaks of Mg, P, O, and N. These preliminary data suggest that the two complementary SEM-EDX analyses can be used to detect and identify microorganisms and crystals in urine samples. Further studies are still needed to apply SEM-EDX to urine sediment analysis. SEM-EDX analyses provided comparative results with the routine results, with accurate identification, high resolution and deep focus compared to the routine urinalysis SEM-analysis allowed us to identify and classify microorganisms in urine sediments into the categories of gram-negative bacilli, cluster cocci, chain cocci, gram-negative bacilli, gram-positive bacilli and yeasts. SEM-EDX analysis enabled the accurate identification of crystals based on both morphology and elemental composition.
Collapse
Affiliation(s)
- Abdourahamane Yacouba
- IRD, AP-HM, MEPHI, Aix Marseille University, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- Université Abdou Moumouni, Niamey, Niger
| | | | - Gabriel Haddad
- IRD, AP-HM, MEPHI, Aix Marseille University, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | | | | | - Grégory Dubourg
- IRD, AP-HM, MEPHI, Aix Marseille University, Marseille, France
| | | | | | | |
Collapse
|
8
|
Cohen A, Gotnayer L, Gal S, Aranovich D, Vidavsky N. Multicellular spheroids containing synthetic mineral particles: an advanced 3D tumor model system to investigate breast precancer malignancy potential according to the mineral type. J Mater Chem B 2023; 11:8033-8045. [PMID: 37534429 DOI: 10.1039/d3tb00439b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Mineral particles that form in soft tissues in association with disease conditions are heterogeneous in their composition and physiochemical properties. Hence, it is challenging to study the effect of mineral type on disease progression in a high-throughput and realistic manner. For example, most early breast precancer lesions, termed ductal carcinoma in situ (DCIS), contain microcalcifications (MCs), calcium-containing pathological minerals. The most common type of MCs is calcium phosphate crystals, mainly carbonated apatite; it is associated with either benign or malignant lesions. In vitro studies indicate that the crystal properties of apatite MCs can affect breast cancer progression. A less common type of MCs is calcium oxalate dihydrate (COD), which is almost always found in benign lesions. We developed a 3D tumor model of multicellular spheroids of human precancer cells containing synthetic MC analogs that link the crystal properties of MCs with the progression of breast precancer to invasive cancer. Using this 3D model, we show that apatite crystals induce Her2 overexpression in DCIS cells. This tumor-triggering effect is increased when the carbonate fraction in the MCs decreases. COD crystals, in contrast, decrease Her2 expression in the spheroids, even compared with a control group with no added MC analogs. Furthermore, COD decreases cell proliferation and migration in DCIS monolayers compared to untreated cells and cells incubated with apatite crystals. This finding suggests that COD is not randomly located only in benign lesions-it may actively contribute to suppressing precancer progression in its surroundings. Our model provides an easy-to-manipulate platform to better understand the interactions between mineral particles and their biological microenvironment. A better understanding of the effect of the crystal properties of MCs on precancer progression will potentially provide new directions for better precancer prognosis and treatment.
Collapse
Affiliation(s)
- Amit Cohen
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Lotem Gotnayer
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Sahar Gal
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Dina Aranovich
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Netta Vidavsky
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
9
|
Du H, Yao MMS, Liu S, Chen L, Chan WP, Feng M. Automatic Calcification Morphology and Distribution Classification for Breast Mammograms With Multi-Task Graph Convolutional Neural Network. IEEE J Biomed Health Inform 2023; 27:3782-3793. [PMID: 37027577 DOI: 10.1109/jbhi.2023.3249404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The morphology and distribution of microcalcifications are the most important descriptors for radiologists to diagnose breast cancer based on mammograms. However, it is very challenging and time-consuming for radiologists to characterize these descriptors manually, and there also lacks of effective and automatic solutions for this problem. We observed that the distribution and morphology descriptors are determined by the radiologists based on the spatial and visual relationships among calcifications. Thus, we hypothesize that this information can be effectively modelled by learning a relationship-aware representation using graph convolutional networks (GCNs). In this study, we propose a multi-task deep GCN method for automatic characterization of both the morphology and distribution of microcalcifications in mammograms. Our proposed method transforms morphology and distribution characterization into node and graph classification problem and learns the representations concurrently. We trained and validated the proposed method in an in-house dataset and public DDSM dataset with 195 and 583 cases,respectively. The proposed method reaches good and stable results with distribution AUC at 0.812 ± 0.043 and 0.873 ± 0.019, morphology AUC at 0.663 ± 0.016 and 0.700 ± 0.044 for both in-house and public datasets. In both datasets, our proposed method demonstrates statistically significant improvements compared to the baseline models. The performance improvements brought by our proposed multi-task mechanism can be attributed to the association between the distribution and morphology of calcifications in mammograms, which is interpretable using graphical visualizations and consistent with the definitions of descriptors in the standard BI-RADS guideline. In short, we explore, for the first time, the application of GCNs in microcalcification characterization that suggests the potential of using graph learning for more robust understanding of medical images.
Collapse
|
10
|
Hu Y, Mao L, Wang M, Li Z, Li M, Wang C, Ji L, Zeng H, Zhang X. New insights into breast microcalcification for poor prognosis: NACT cohort and bone metastasis evaluation cohort. J Cancer Res Clin Oncol 2023; 149:7285-7297. [PMID: 36917189 DOI: 10.1007/s00432-023-04668-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/23/2023] [Indexed: 03/15/2023]
Abstract
OBJECTIVES The study aimed to analyze the poor prognosis of microcalcification in breast cancer (BC), including the pathological complete response (pCR) to neoadjuvant chemotherapy (NACT) and the risk of bone metastases. MATERIALS AND METHODS 313 breast cancer patients received NACT to evaluate pCR and 1182 patients from a multicenter database to assess bone metastases were retrospectively included. Two groups were divided according to the presence or absence of mammography microcalcification. Clinical data, image characteristics, neoadjuvant treatment response, bone involvement, and follow-up information were recorded. The pCR and bone metastases were compared between subgroups using the Mann-Whitney and χ2 tests and logistic regression, respectively. RESULTS Mammographic microcalcification was associated with a lower pCR than uncalcified BC in the NACT cohort (20.6% vs 31.6%, P = 0.029). Univariate and multivariate analysis suggested that calcification was a risk factor for poor NACT response [OR = 1.780, 95%CI (1.065-2.974), P = 0.028], [OR = 2.352, 95%CI (1.186-4.667), P = 0.014]. Microcalcification was more likely to be necrosis on MRI than those without microcalcification (53.0% vs 31.7%, P < 0.001), multivariate analysis indicated that tumor necrosis was also a risk factor for poor NACT response [OR = 2.325, 95%CI (1.100-4.911), P = 0.027]. Age, menopausal status, breast density, mass, molecular, and pathology type were not significantly associated with non-pCR risk assessment. In a multicenter cohort of 1182 patients with pathologically confirmed BC, those with microcalcifications had a higher proportion of bone metastases compared to non-calcified BC (11.6% vs 4.9%, P < 0.001). Univariate and multivariate analysis showed that microcalcification was an independent risk factor for bone metastasis [OR = 2.550, 95%CI (1.620-4.012), P < 0.001], [OR = 2.268(1.263-4.071), P = 0.006)]. Osteolytic bone metastases predominated but there was no statistical difference between the two groups (78.9% vs 60.7%, P = 0.099). Calcified BC was mainly involved in axial bone, but was more likely to involve the whole-body bone than non-calcified BC (33.8% vs 10.7%, P = 0.021). CONCLUSION This study provides important insights into the poor prognosis of microcalcification, not only in terms of poor response to NACT but also the risk factor of bone metastases.
Collapse
Affiliation(s)
- Yangling Hu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lijuan Mao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mengyi Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhenqiu Li
- Department of Radiology, The Panyu Fifth Hospital, Guangzhou, China
| | - Meizhi Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoyang Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lin Ji
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui Zeng
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiaoling Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
11
|
Porcelli F, Verri M, De Santis S, Crescenzi A, Bianchi A, Felici AC, Sotgiu G, Romano S, Orsini M. Considerations on chemical composition of psammoma bodies: Automated detection strategy by infrared microspectroscopy in ovarian and thyroid cancer tissues. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 298:122792. [PMID: 37156176 DOI: 10.1016/j.saa.2023.122792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/04/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Ectopic calcifications are observed in many soft tissues and are associated with several diseases, including cancer. The mechanism of their formation and the correlation with disease progression are often unclear. Detailed knowledge of the chemical composition of these inorganic formations can be very helpful in better understanding their relationship with unhealthy tissue. In addition, information on microcalcifications can be very useful for early diagnosis and provide insight into prognosis. In this work the chemical composition of psammoma bodies (PBs) found in tissues of human ovarian serous tumors was examined. The analysis using Micro Fourier Transform Infrared Spectroscopy (micro-FTIR) revealed that these microcalcifications contain amorphous calcium carbonate phosphate. Moreover, some PB grains showed the presence of phospholipids. This interesting result corroborates the proposed formation mechanism reported in many studies according to which ovarian cancer cells switch to a calcifying phenotype by inducing the deposition of calcifications. In addition, other techniques as X-ray Fluorescence Spectroscopy (XRF), Inductively Coupled Plasma Optical Emission Spectroscopy(ICP-OES) and Scanning electron microscopy (SEM) with Energy Dispersive X-ray Spectroscopy (EDX) were performed on the PBs from ovary tissues to determine the elements present. The PBs found in ovarian serous cancer showed a composition comparable to PBs isolated from papillary thyroid. Based on the chemical similarity of IR spectra, using micro-FTIR spectroscopy combined with multivariate analysis, an automatic recognition method was constructed. With this prediction model it was possible to identify PBs microcalcifications in tissues of both ovarian cancers, regardless of tumor grade, and thyroid cancer with high sensitivity. Such approach could become a valuable tool for routine macrocalcification detection because it eliminates sample staining, and the subjectivity of conventional histopathological analysis.
Collapse
Affiliation(s)
- Francesco Porcelli
- Department of Industrial, Electronic and Mechanical Engineering, Roma Tre University, Via Vito Volterra 62, 00146 Rome, Italy
| | - Martina Verri
- Pathology Unit, University Hospital Campus Bio-Medico, Rome, Italy
| | - Serena De Santis
- Department of Industrial, Electronic and Mechanical Engineering, Roma Tre University, Via Vito Volterra 62, 00146 Rome, Italy
| | - Anna Crescenzi
- Pathology Unit, University Hospital Campus Bio-Medico, Rome, Italy
| | | | - Anna Candida Felici
- Basic and Applied Sciences for Engineering, Sapienza University, Via A. Scarpa 16, Rome, Italy
| | - Giovanni Sotgiu
- Department of Industrial, Electronic and Mechanical Engineering, Roma Tre University, Via Vito Volterra 62, 00146 Rome, Italy
| | - Susanna Romano
- Department of Industrial, Electronic and Mechanical Engineering, Roma Tre University, Via Vito Volterra 62, 00146 Rome, Italy
| | - Monica Orsini
- Department of Industrial, Electronic and Mechanical Engineering, Roma Tre University, Via Vito Volterra 62, 00146 Rome, Italy.
| |
Collapse
|
12
|
Bouzy P, Lyburn ID, Pinder SE, Scott R, Mansfield J, Moger J, Greenwood C, Bouybayoune I, Cornford E, Rogers K, Stone N. Exploration of utility of combined optical photothermal infrared and Raman imaging for investigating the chemical composition of microcalcifications in breast cancer. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:1620-1630. [PMID: 36880909 PMCID: PMC10065137 DOI: 10.1039/d2ay01197b] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/21/2023] [Indexed: 06/02/2023]
Abstract
Microcalcifications play an important role in cancer detection. They are evaluated by their radiological and histological characteristics but it is challenging to find a link between their morphology, their composition and the nature of a specific type of breast lesion. Whilst there are some mammographic features that are either typically benign or typically malignant often the appearances are indeterminate. Here, we explore a large range of vibrational spectroscopic and multiphoton imaging techniques in order to gain more information about the composition of the microcalcifications. For the first time, we validated the presence of carbonate ions in the microcalcifications by O-PTIR and Raman spectroscopy at the same time, the same location and the same high resolution (0.5 μm). Furthermore, the use of multiphoton imaging allowed us to create stimulated Raman histology (SRH) images which mimic histological images with all chemical information. In conclusion, we established a protocol for efficiently analysing the microcalcifications by iteratively refining the area of interest.
Collapse
Affiliation(s)
- Pascaline Bouzy
- School of Physics and Astronomy, University of Exeter, Exeter, UK.
| | - Iain D Lyburn
- Cranfield Forensic Institute, Cranfield University, Shrivenham, UK
- Gloucestershire Hospitals NHS Foundation Trust, UK
| | - Sarah E Pinder
- King's College London, Comprehensive Cancer Centre at Guy's Hospital, London, UK
| | - Robert Scott
- Cranfield Forensic Institute, Cranfield University, Shrivenham, UK
| | | | - Julian Moger
- School of Physics and Astronomy, University of Exeter, Exeter, UK.
| | - Charlene Greenwood
- School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, UK
| | - Ihssane Bouybayoune
- King's College London, Comprehensive Cancer Centre at Guy's Hospital, London, UK
| | | | - Keith Rogers
- Cranfield Forensic Institute, Cranfield University, Shrivenham, UK
| | - Nick Stone
- School of Physics and Astronomy, University of Exeter, Exeter, UK.
- Gloucestershire Hospitals NHS Foundation Trust, UK
| |
Collapse
|
13
|
Tian Y, Zhao L, Gui Z, Liu S, Liu C, Yu T, Zhang L. Clinical and pathological features analysis of invasive breast cancer with microcalcification. Cancer Med 2023. [DOI: 10.1002/cam4.5848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 03/09/2023] [Accepted: 03/12/2023] [Indexed: 03/29/2023] Open
|
14
|
Gotnayer L, Aranovich D, Fraenkel M, Yoel U, Vidavsky N. Zinc in microscopic calcifications isolated from thyroid fine needle aspiration may serve as a biomarker of thyroid nodule malignancy: A promising proof-of-concept. Acta Biomater 2023; 161:275-284. [PMID: 36931418 DOI: 10.1016/j.actbio.2023.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Thyroid nodules (TNs) are common neck ultrasonography (US) findings, yet only 5-10% of these nodules harbor thyroid cancer (TC). When US characteristics are consistent with an intermediate or high suspicion for TN malignancy, fine needle aspiration for cytology (FNAC) is indicated. The main limitation of FNAC is that cytological results can be indeterminate in up to 30% of cases, necessitating reevaluation through repeated FNAC, expensive molecular testing, or diagnostic thyroid lobe resection. As such, there is a need for further refinement of current diagnostic algorithms for TNs without subjecting patients to additional invasive procedures. As calcifications detected during thyroid US are considered a high-risk feature for malignancy, we used the material remaining following routine thyroid FNAC to isolate microscopic calcifications (MCs). We then characterized the elemental composition, morphology, and crystal phases of these MCs, ultimately revealing differences between the MCs from benign and malignant TNs. Specifically, thyroid MCs were identified as calcium phosphate crystals containing varying levels of magnesium, sodium, iron, and zinc. MCs obtained from malignant TNs, mainly papillary thyroid carcinoma, were composed of sub-micrometer spherical particles, whereas MCs from benign TNs consisted of faceted particles. While samples from most patients with a final diagnosis of malignant TNs (50% of them with indeterminate cytology) harbored zinc-containing MCs, zinc was largely absent in MCs from benign TNs (23% with indeterminate or non-diagnostic cytology). Together, these data suggest that the presence of zinc in MCs isolated from samples collected during routine FNAC may potentially offer value as a biomarker of TN malignancy. STATEMENT OF SIGNIFICANCE: As up to 40% of patients assessed for thyroid malignancy do not receive a definite diagnosis following thyroid nodule (TN) fine needle aspiration (FNA), there is a pressing need to improve the accuracy of current diagnostic algorithms. Chemical analyses of microscopic calcifications (MCs) may serve as a diagnostic target. We developed a straightforward protocol to chemically characterize MCs from excess material collected from TNs during routine FNA and found that these MCs differed between benign and malignant TNs. Specifically, zinc in TN-derived MCs may indicate a higher nodule malignancy risk, thus increasing the diagnostic accuracy of the FNA procedure, reducing the need for recurrent biopsies and diagnostic surgical procedures, and decreasing the costs, uncertainty, and stress faced by affected patients.
Collapse
Affiliation(s)
- Lotem Gotnayer
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Dina Aranovich
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Merav Fraenkel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; Endocrinology, Soroka University Medical Center, Beer Sheva, Israel
| | - Uri Yoel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel; Endocrinology, Soroka University Medical Center, Beer Sheva, Israel.
| | - Netta Vidavsky
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel; Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
15
|
Chen H, Bao L, Yu L, Sun H, Tan Y, Wei P, Zheng Z. Value of multimodal imaging in the diagnosis of breast sclerosing adenosis associated with malignant lesions. JOURNAL OF CLINICAL ULTRASOUND : JCU 2023; 51:485-493. [PMID: 36250329 DOI: 10.1002/jcu.23376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
AIM To explore the diagnostic value of multimodal imaging techniques, including automatic breast volume scanner (ABVS), mammography (MG), and magnetic resonance (MRI) in breast sclerosing adenosis (SA) associated with malignant lesions. METHODS From January 2018 to October 2020, 76 patients (88 lesions) with pathologically confirmed as SA associated with malignant or benign lesions were retrospective analyzed. All patients completed ABVS examination, 58 patients (67 lesions) with MG and 50 patients (62 lesions) with MRI were also completed before biopsy or surgical excision, of which, six patients (eight lesions) diagnosed as Breast Imaging Reporting and Data System (BI-RADS) category 3 by all imaging examinations underwent surgical excision without biopsy, other 70 patients (80 lesions) with BI-RADS category 4 or above by any imaging examination completed biopsy, including 65 patients (75 lesions) were further surgical excised and the other five patients (five lesions) were just followed up. All lesions were retrospectively described and classified, and were divided into benign group and malignant group according to their pathological results. Image features of different examination methods between the two groups were compared and analyzed. A ROC curve was established using the sensitivity of BI-RADS categories to predict malignant lesions in different imaging techniques as the ordinate and 1-specificity as the abscissa. RESULTS 88 lesions including 26 purely SA and 45 SA associated with benign lesions were classified as benign group, and the remaining 17 SA associated with malignant lesions were classified as malignant group. On ABVS, 40 mass lesions, their heterogeneous echo, not circumscribed margin and coronal convergence signs were statistically significant for malignant lesions (p < .05), but the remain 48 nonmass lesions lack specific sonographic features. On MG, 12 showed negative results, 55 showed with microcalcification, mass, structural distortion, and asymmetric density shadow, of which 11 lesions had the above two signs at the same time, but only microcalcification had statistical difference between the two groups. 35 mass enhanced lesions and 27 nonmass enhanced lesions on MRI, but there were no significant difference between their pathological results. Time signal intensity curves showed no differences, but ADC value <1.10 × 10-3 mm2 /s is more significant in malignant lesions (p < .05). The area under the ROC curve (AUC) of BI-RADS classification of ABVS, MG, and MRI in the diagnosis of malignant lesions were 0.611, 0.474, and 0.751, respectively, and the AUC of the combined diagnosis of the three was 0.761. CONCLUSION Mass lesions with heterogeneous echo, not circumscribed margin and coronal convergence sign on ABVS, microcalcification on MG and the ADC value <1.10 × 10-3 mm2 /s on MRI are significant signs for SA associated with malignant lesions. The combined diagnosis of the three methods was the highest, and the following were MRI, ABVS, and MG. Therefore, be cognizant of significant characteristics in SA associated with malignancy showed in different imaging examinations can improve the preoperative evaluation of SA and better provide basis for subsequent clinical decision-making.
Collapse
Affiliation(s)
- Haiping Chen
- Department of Ultrasound, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Echocardiography and Vascular Ultrasound Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyun Bao
- Department of Ultrasound, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifang Yu
- Department of Ultrasound, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Sun
- Department of Ultrasound, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanjuan Tan
- Department of Ultrasound, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peiying Wei
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhelan Zheng
- Department of Echocardiography and Vascular Ultrasound Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
16
|
Kunitake JA, Sudilovsky D, Johnson LM, Loh HC, Choi S, Morris PG, Jochelson MS, Iyengar NM, Morrow M, Masic A, Fischbach C, Estroff LA. Biomineralogical signatures of breast microcalcifications. SCIENCE ADVANCES 2023; 9:eade3152. [PMID: 36812311 PMCID: PMC9946357 DOI: 10.1126/sciadv.ade3152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Microcalcifications, primarily biogenic apatite, occur in cancerous and benign breast pathologies and are key mammographic indicators. Outside the clinic, numerous microcalcification compositional metrics (e.g., carbonate and metal content) are linked to malignancy, yet microcalcification formation is dependent on microenvironmental conditions, which are notoriously heterogeneous in breast cancer. We interrogate multiscale heterogeneity in 93 calcifications from 21 breast cancer patients using an omics-inspired approach: For each microcalcification, we define a "biomineralogical signature" combining metrics derived from Raman microscopy and energy-dispersive spectroscopy. We observe that (i) calcifications cluster into physiologically relevant groups reflecting tissue type and local malignancy; (ii) carbonate content exhibits substantial intratumor heterogeneity; (iii) trace metals including zinc, iron, and aluminum are enhanced in malignant-localized calcifications; and (iv) the lipid-to-protein ratio within calcifications is lower in patients with poor composite outcome, suggesting that there is potential clinical value in expanding research on calcification diagnostic metrics to include "mineral-entrapped" organic matrix.
Collapse
Affiliation(s)
| | - Daniel Sudilovsky
- Department of Pathology and Laboratory Medicine, Cayuga Medical Center at Ithaca, Ithaca, NY 14850, USA
- Pathology Department, Kingman Regional Medical Center, Kingman, AZ 86409, USA
- Pathology Department, Western Arizona Medical Center, Bullhead City, AZ 86442, USA
- Pathology Department, Yuma Regional Medical Center, Yuma, AZ 85364, USA
| | - Lynn M. Johnson
- Cornell Statistical Consulting Unit, Cornell University, Ithaca, NY 14850, USA
| | - Hyun-Chae Loh
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Patrick G. Morris
- Medical Oncology Service, Beaumont Hospital, Dublin, Ireland
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Maxine S. Jochelson
- Department of Radiology, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY 10065, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Monica Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Admir Masic
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Corresponding author. (L.A.E.); (C.F.); (A.M.)
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14850, USA
- Corresponding author. (L.A.E.); (C.F.); (A.M.)
| | - Lara A. Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14850, USA
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14850, USA
- Corresponding author. (L.A.E.); (C.F.); (A.M.)
| |
Collapse
|
17
|
Piddubnyi A, Kolomiiets O, Danilchenko S, Stepanenko A, Moskalenko Y, Moskalenko R. The Prospects of Using Structural Phase Analysis of Microcalcifications in Breast Cancer Diagnostics. Diagnostics (Basel) 2023; 13:diagnostics13040737. [PMID: 36832224 PMCID: PMC9955541 DOI: 10.3390/diagnostics13040737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
The detection of microcalcifications in the breast by mammography is of great importance for the early diagnostics of breast cancer. This study aimed to establish the basic morphological and crystal-chemical properties of microscopic calcifications and their impact on breast cancer tissue. During the retrospective study, 55 out of 469 breast cancer samples had microcalcifications. The expression of the estrogen and progesterone receptors and Her2-neu showed no significant difference from the non-calcified samples. An in-depth study of 60 tumor samples revealed a higher expression of osteopontin in the calcified breast cancer samples (p ˂ 0.01). The mineral deposits had a hydroxyapatite composition. Within the group of calcified breast cancer samples, we detected six cases of colocalization of oxalate microcalcifications together with biominerals of the usual "hydroxyapatite" phase composition. The simultaneous presence of calcium oxalate and hydroxyapatite was accompanied by a different spatial localization of microcalcifications. Thus, the phase compositions of microcalcifications could not be used as criteria for the differential diagnostics of breast tumors.
Collapse
Affiliation(s)
- Artem Piddubnyi
- Department of Pathology, Sumy State University, 40022 Sumy, Ukraine
- Ukrainian-Swedish Research Center SUMEYA, Sumy State University, 40022 Sumy, Ukraine
| | - Olena Kolomiiets
- Department of Pathology, Sumy State University, 40022 Sumy, Ukraine
| | | | - Andriy Stepanenko
- Department of Electronics, General and Applied Physics, Sumy State University, 40007 Sumy, Ukraine
| | - Yuliia Moskalenko
- Department of Oncology and Radiology, Sumy State University, 40022 Sumy, Ukraine
| | - Roman Moskalenko
- Department of Pathology, Sumy State University, 40022 Sumy, Ukraine
- Ukrainian-Swedish Research Center SUMEYA, Sumy State University, 40022 Sumy, Ukraine
- Correspondence: ; Tel.: +38-(09)-79802731
| |
Collapse
|
18
|
Contrast-Enhanced Spectral Mammography in the Evaluation of Breast Microcalcifications: Controversies and Diagnostic Management. Healthcare (Basel) 2023; 11:healthcare11040511. [PMID: 36833045 PMCID: PMC9956946 DOI: 10.3390/healthcare11040511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The aim of this study was to evaluate the diagnostic performance of contrast-enhanced spectral mammography (CESM) in predicting breast lesion malignancy due to microcalcifications compared to lesions that present with other radiological findings. Three hundred and twenty-one patients with 377 breast lesions that underwent CESM and histological assessment were included. All the lesions were scored using a 4-point qualitative scale according to the degree of contrast enhancement at the CESM examination. The histological results were considered the gold standard. In the first analysis, enhancement degree scores of 2 and 3 were considered predictive of malignity. The sensitivity (SE) and positive predictive value (PPV) were significative lower for patients with lesions with microcalcifications without other radiological findings (SE = 53.3% vs. 82.2%, p-value < 0.001 and PPV = 84.2% vs. 95.2%, p-value = 0.049, respectively). On the contrary, the specificity (SP) and negative predictive value (NPV) were significative higher among lesions with microcalcifications without other radiological findings (SP = 95.8% vs. 84.2%, p-value = 0.026 and NPV = 82.9% vs. 55.2%, p-value < 0.001, respectively). In a second analysis, degree scores of 1, 2, and 3 were considered predictive of malignity. The SE (80.0% vs. 96.8%, p-value < 0.001) and PPV (70.6% vs. 88.3%, p-value: 0.005) were significantly lower among lesions with microcalcifications without other radiological findings, while the SP (85.9% vs. 50.9%, p-value < 0.001) was higher. The enhancement of microcalcifications has low sensitivity in predicting malignancy. However, in certain controversial cases, the absence of CESM enhancement due to its high negative predictive value can help to reduce the number of biopsies for benign lesions.
Collapse
|
19
|
Zhu Y, Yin WF, Yu P, Zhang C, Sun MH, Kong LY, Yang L. Meso-Hannokinol inhibits breast cancer bone metastasis via the ROS/JNK/ZEB1 axis. Phytother Res 2023. [PMID: 36726293 DOI: 10.1002/ptr.7732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/25/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Distal metastases from breast cancer, especially bone metastases, are extremely common in the late stages of the disease and are associated with a poor prognosis. EMT is a biomarker of the early process of bone metastasis, and MMP-9 and MMP-13 are important osteoclastic activators. Previously, we found that meso-Hannokinol (HA) could significantly inhibit EMT and MMP-9 and MMP-13 expressions in breast cancer cells. On this basis, we further explored the role of HA in breast cancer bone metastasis. In vivo, we established a breast cancer bone metastasis model by intracardially injecting breast cancer cells. Intraperitoneal injections of HA significantly reduced breast cancer cell metastasis to the leg bone in mice and osteolytic lesions caused by breast cancer. In vitro, HA inhibited the migration and invasion of breast cancer cells and suppressed the expressions of EMT, MMP-9, MMP-13, and other osteoclastic activators. HA inhibited EMT and MMP-9 by activating the ROS/JNK pathway as demonstrated by siJNK and SP600125 inhibition of JNK phosphorylation and NAC scavenging of ROS accumulation. Moreover, HA promoted bone formation and inhibited bone resorption in vitro. In conclusion, our findings suggest that HA may be an excellent candidate for treating breast cancer bone metastasis.
Collapse
Affiliation(s)
- Yuan Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wei-Feng Yin
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Pei Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ming-Hui Sun
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
Cui H, Sun Y, Zhao D, Zhang X, Kong H, Hu N, Wang P, Zuo X, Fan W, Yao Y, Fu B, Tian J, Wu M, Gao Y, Ning S, Zhang L. Radiogenomic analysis of prediction HER2 status in breast cancer by linking ultrasound radiomic feature module with biological functions. J Transl Med 2023; 21:44. [PMID: 36694240 PMCID: PMC9875533 DOI: 10.1186/s12967-022-03840-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 12/19/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) overexpressed associated with poor prognosis in breast cancer and HER2 has been defined as a therapeutic target for breast cancer treatment. We aimed to explore the molecular biological information in ultrasound radiomic features (URFs) of HER2-positive breast cancer using radiogenomic analysis. Moreover, a radiomics model was developed to predict the status of HER2 in breast cancer. METHODS This retrospective study included 489 patients who were diagnosed with breast cancer. URFs were extracted from a radiomics analysis set using PyRadiomics. The correlations between differential URFs and HER2-related genes were calculated using Pearson correlation analysis. Functional enrichment of the identified URFs-correlated HER2 positive-specific genes was performed. Lastly, the radiomics model was developed based on the URF-module mined from auxiliary differential URFs to assess the HER2 status of breast cancer. RESULTS Eight differential URFs (p < 0.05) were identified among the 86 URFs extracted by Pyradiomics. 25 genes that were found to be the most closely associated with URFs. Then, the relevant biological functions of each differential URF were obtained through functional enrichment analysis. Among them, Zone Entropy is related to immune cell activity, which regulate the generation of calcification in breast cancer. The radiomics model based on the Logistic classifier and URF-module showed good discriminative ability (AUC = 0.80, 95% CI). CONCLUSION We searched for the URFs of HER2-positive breast cancer, and explored the underlying genes and biological functions of these URFs. Furthermore, the radiomics model based on the Logistic classifier and URF-module relatively accurately predicted the HER2 status in breast cancer.
Collapse
Affiliation(s)
- Hao Cui
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Yue Sun
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Dantong Zhao
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Xudong Zhang
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Hanqing Kong
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Nana Hu
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Panting Wang
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Xiaoxuan Zuo
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Wei Fan
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Yuan Yao
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Baiyang Fu
- grid.412463.60000 0004 1762 6325Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Jiawei Tian
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Meixin Wu
- grid.412463.60000 0004 1762 6325Department of Clinical Medicine, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang, 150086 China
| | - Yue Gao
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Shangwei Ning
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Lei Zhang
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| |
Collapse
|
21
|
Simmons L, Feng L, Fatemi-Ardekani A, Noseworthy MD. The Role of Calcium in Non-Invasively Imaging Breast Cancer: An Overview of Current and Modern Imaging Techniques. Crit Rev Biomed Eng 2023; 51:43-62. [PMID: 37602447 DOI: 10.1615/critrevbiomedeng.2023047683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
The landscape of breast cancer diagnostics has significantly evolved over the past decade. With these changes, it is possible to provide a comprehensive assessment of both benign and malignant breast calcifications. The biochemistry of breast cancer and calcifications are thoroughly examined to describe the potential to characterize better different calcium salts composed of calcium carbonate, calcium oxalate, or calcium hydroxyapatite and their associated prognostic implications. Conventional mammographic imaging techniques are compared to available ones, including breast tomosynthesis and contrast-enhanced mammography. Additional methods in computed tomography and magnetic resonance imaging are discussed. The concept of using magnetic resonance imaging particularly magnetic susceptibility to characterize the biochemical characteristics of calcifications is described. As we know magnetic resonance imaging is safe and there is no ionization radiation. Experimental findings through magnetic resonance susceptibility imaging techniques are discussed to illustrate the potential for integrating this technique to provide a quantitative assessment of magnetic susceptibility. Under the right magnetic resonance imaging conditions, a distinct phase variability was isolated amongst different types of calcium salts.
Collapse
Affiliation(s)
- Lyndsay Simmons
- Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, ON, Canada; Mohawk College, Institute for Applied Health Sciences, Hamilton, ON, Canada; Imaging Research Centre, St. Joseph's Healthcare Hamilton, 50 Charlton Ave. E., Hamilton, ON, Canada
| | - Lisa Feng
- Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, ON, Canada
| | - Ali Fatemi-Ardekani
- Medical Physics, Merit Health, Southeast Cancer Network; Department of Physics, Jackson State University
| | - Michael D Noseworthy
- Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, ON, Canada; Imaging Research Centre, St. Joseph's Healthcare Hamilton, 50 Charlton Ave. E., Hamilton, ON, Canada; Department of Electrical and Computer Engineering, McMaster University, 280 Main Street W., Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton ON, Canada; Department of Radiology, McMaster University, 1280 Main St. W., Hamilton, ON, Canada
| |
Collapse
|
22
|
Petay M, Cherfan M, Bouderlique E, Reguer S, Mathurin J, Dazzi A, L’Heronde M, Daudon M, Letavernier E, Deniset-Besseau A, Bazin D. Multiscale approach to provide a better physicochemical description of women breast microcalcifications. CR CHIM 2022. [DOI: 10.5802/crchim.210] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Mauriello S, Treglia M, Pallocci M, Bonfiglio R, Giacobbi E, Passalacqua P, Cammarano A, D’Ovidio C, Marsella LT, Scimeca M. Antigenicity Preservation Is Related to Tissue Characteristics and the Post-Mortem Interval: Immunohistochemical Study and Literature Review. Healthcare (Basel) 2022; 10:healthcare10081495. [PMID: 36011152 PMCID: PMC9408092 DOI: 10.3390/healthcare10081495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
The main aim of this study was to investigate the post-mortem proteolytic degradation process of selected tissue antigens and correlate it to the post-mortem interval. During the autopsy of 12 cadavers (time interval ranging 1 day–2 years after death) samples of skin, liver, kidney, and spleen were collected. All samples were formalin-fixed and paraffin-embedded. Four µm paraffin sections were used for hematoxylin–eosin staining and immunohistochemical analysis (Ki67, Vimentin, Pan cytokeratin, and CD20). Data reported here show that immunohistochemical reactivity preservation was related to the characteristics of the tissues. In particular, the most resistant tissue was the skin, where the autolysis phenomena were not appreciable before 5 days. On the contrary, the liver and the spleen underwent early autolysis, while the kidney displayed an early autolysis of the tubules and a late one of the glomeruli. As concerns specific antigens, immunoreactivity was lost earliest for nuclear antigens as compared to cytoplasmic ones. In conclusion, our results demonstrate that immunohistochemical detection of specific antigens may be useful in estimating the post-mortem interval, especially when we need to know whether the post-mortem interval is a few days or more than 7–10 days.
Collapse
Affiliation(s)
- Silvestro Mauriello
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Michele Treglia
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Margherita Pallocci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
- Correspondence:
| | - Rita Bonfiglio
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Erica Giacobbi
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Pierluigi Passalacqua
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Andrea Cammarano
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Cristian D’Ovidio
- Department of Medicine and Aging Sciences, University of Chieti-Pescara “G. D’Annunzio”, Section of Legal Medicine, 66100 Chieti, Italy
| | - Luigi Tonino Marsella
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
24
|
Abstract
Metastasis is responsible for a large majority of death from malignant solid tumors. Bone is one of the most frequently affected organs in cancer metastasis, especially in breast and prostate cancer. Development of bone metastasis requires cancer cells to successfully complete a number of challenging steps, including local invasion and intravasation, survival in circulation, extravasation and initial seeding, and finally, formation of metastatic colonies after a period of dormancy or indolent growth. During this process, cancer cells often undergo a series of cellular and molecular changes to gain cellular plasticity that helps them adapt to various environments they encounter along the journey of metastasis. Understanding the mechanisms behind cellular plasticity and adaptation during the formation of bone metastasis is crucial for the development of novel therapies.
Collapse
Affiliation(s)
- Cao Fang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
25
|
Brahimetaj R, Willekens I, Massart A, Forsyth R, Cornelis J, Mey JD, Jansen B. Improved automated early detection of breast cancer based on high resolution 3D micro-CT microcalcification images. BMC Cancer 2022; 22:162. [PMID: 35148703 PMCID: PMC8832731 DOI: 10.1186/s12885-021-09133-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The detection of suspicious microcalcifications on mammography represents one of the earliest signs of a malignant breast tumor. Assessing microcalcifications' characteristics based on their appearance on 2D breast imaging modalities is in many cases challenging for radiologists. The aims of this study were to: (a) analyse the association of shape and texture properties of breast microcalcifications (extracted by scanning breast tissue with a high resolution 3D scanner) with malignancy, (b) evaluate microcalcifications' potential to diagnose benign/malignant patients. METHODS Biopsy samples of 94 female patients with suspicious microcalcifications detected during a mammography, were scanned using a micro-CT scanner at a resolution of 9 μm. Several preprocessing techniques were applied on 3504 extracted microcalcifications. A high amount of radiomic features were extracted in an attempt to capture differences among microcalcifications occurring in benign and malignant lesions. Machine learning algorithms were used to diagnose: (a) individual microcalcifications, (b) samples. For the samples, several methodologies to combine individual microcalcification results into sample results were evaluated. RESULTS We could classify individual microcalcifications with 77.32% accuracy, 61.15% sensitivity and 89.76% specificity. At the sample level diagnosis, we achieved an accuracy of 84.04%, sensitivity of 86.27% and specificity of 81.39%. CONCLUSIONS By studying microcalcifications' characteristics at a level of details beyond what is currently possible by using conventional breast imaging modalities, our classification results demonstrated a strong association between breast microcalcifications and malignancies. Microcalcification's texture features extracted in transform domains, have higher discriminating power to classify benign/malignant individual microcalcifications and samples compared to pure shape-features.
Collapse
Affiliation(s)
- Redona Brahimetaj
- Department of Electronics and Informatics (ETRO), Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, B-1050 Belgium
| | - Inneke Willekens
- Radiology Department, Universitair Ziekenhuis (UZ) Brussels, Laarbeeklaan 101, Brussels, 1090 Belgium
| | - Annelien Massart
- Radiology Department, Universitair Ziekenhuis (UZ) Brussels, Laarbeeklaan 101, Brussels, 1090 Belgium
| | - Ramses Forsyth
- Pathology Department, Universitair Ziekenhuis (UZ) Brussels, Laarbeeklaan 101, Brussels, 1090 Belgium
| | - Jan Cornelis
- Department of Electronics and Informatics (ETRO), Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, B-1050 Belgium
| | - Johan De Mey
- Radiology Department, Universitair Ziekenhuis (UZ) Brussels, Laarbeeklaan 101, Brussels, 1090 Belgium
| | - Bart Jansen
- Department of Electronics and Informatics (ETRO), Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, B-1050 Belgium
- IMEC, Kapeldreef 75, Leuven, B-3001 Belgium
| |
Collapse
|
26
|
Logullo A, Prigenzi K, Nimir C, Franco A, Campos M. Breast microcalcifications: Past, present and future (Review). Mol Clin Oncol 2022; 16:81. [PMID: 35251632 PMCID: PMC8892454 DOI: 10.3892/mco.2022.2514] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/19/2021] [Indexed: 11/08/2022] Open
Abstract
Mammary microcalcifications (MCs) are calcium deposits that are considered as robust markers of breast cancer when identified on mammography. MCs are frequently associated with premalignant and malignant lesions. The aim of the present review was to describe the MC types and associated radiological and pathological aspects in detail, provide insights and approaches to the topic, and describe specific clinical scenarios. The primary MC types are composed of calcium oxalate, hydroxyapatite and hydroxyapatite associated with magnesium. The first type is usually associated with benign conditions, while the others remain primarily associated with malignancy. Radiologically, MCs are classified as benign or suspicious. MCs may represent an active pathological mineralization process rather than a passive process, such as degeneration or necrosis. Practical management of breast specimens requires finely calibrated radiological pathological procedures. Understanding the molecular and structural development of MCs may contribute to breast lesion detection and treatment.
Collapse
Affiliation(s)
- Angela Logullo
- Department of Pathology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo 04023‑062, Brazil
| | - Karla Prigenzi
- Department of Pathology, Femme Laboratories, São Paulo 04004‑030, Brazil
| | - Cristiane Nimir
- Department of Pathology, Femme Laboratories, São Paulo 04004‑030, Brazil
| | - Andreia Franco
- Department of Pathology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo 04023‑062, Brazil
| | - Mario Campos
- Breast Imaging Service, Femme Laboratories, São Paulo 04004‑030, Brazil
| |
Collapse
|
27
|
Wang S, Jiang H, Zheng C, Gu M, Zheng X. Secretion of BMP-2 by tumor-associated macrophages (TAM) promotes microcalcifications in breast cancer. BMC Cancer 2022; 22:34. [PMID: 34983451 PMCID: PMC8729115 DOI: 10.1186/s12885-021-09150-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/23/2021] [Indexed: 01/14/2023] Open
Abstract
Introduction Breast microcalcifications is a characteristic feature in diagnostic imaging and a prognostic factor of breast cancer. However, the underlying mechanisms of breast microcalcifications formation are not fully understood. Previous studies have shown that upregulation of bone morphogenetic protein 2 (BMP-2) is associated with the occurrence of microcalcifications and tumor-associated macrophages (TAMs) in the tumor microenvironment can secrete BMP-2. The aim of this study is to elucidate the role of secretion of BMP-2 by TAMs in promoting microcalcifications of breast cancer through immunohistochemical staining and co-culturing of breast cancer cells with TAMs. Methods A total of 272 patients diagnosed with primary invasive breast cancer from January 2010 to January 2012 in the First Hospital of China Medical University were included in this study. Immunohistochemical staining of CD68 (marker of entire macrophages), CD168 (marker of the M2-like macrophages) and BMP-2 were performed on 4-μm tissue microarray (TMA) sections. Following induction, THP-1 cells were differentiated to M2-like TAMs and were then co-cultured with breast cancer cells (MCF-7). Calcifications and BMP-2 expression were analyzed by Alizarin Red S staining and western blot, respectively. Results Immunohistochemical analysis showed that the expression of CD168 was significantly increased in tissues with microcalcifications and was correlated with the expression of BMP-2 and poor prognosis. The formation of cellular microcalcifications and BMP-2 expression were significantly increased in MCF-7 cells co-cultured with TAMs compared with MCF-7 cells alone. Conclusions These findings support the hypothesis that TAMs secrete BMP-2 to induce microcalcifications in breast cancer cells and influence prognosis via multiple pathways including BMP-2 and its downstream factors. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09150-3.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Haiyang Jiang
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Caiwei Zheng
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ming Gu
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xinyu Zheng
- Department of Breast Surgery, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, China. .,Lab 1, Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
28
|
Mohapatra S, Das P, Nayak R, Mishra A, Nayak B. Diagnostic accuracy of mammography in characterizing breast masses using the 5 th edition of BI-RADS: A retrospective study. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_224_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
29
|
Caldarone A, Piccotti F, Morasso C, Truffi M, Sottotetti F, Guerra C, Albasini S, Agozzino M, Villani L, Corsi F. Raman analysis of microcalcifications in male breast cancer. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 263:120185. [PMID: 34298281 DOI: 10.1016/j.saa.2021.120185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/18/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
Microcalcifications (MCs) are important disease markers for breast cancer. Many studies were conducted on their characterization in female breast cancer (FBC), but no information is available on their composition in male breast cancer (MBC). Raman spectroscopy (RS) is a molecular spectroscopy that can rapidly explore the biochemical composition of MCs without requiring any staining protocol. In this study, we optimized an algorithm to identify the mineral components present in MCs from Raman images. The algorithm was then used to study and compare MCs identified on breast cancer pieces from male and female patients. In total, we analyzed 41 MCs from 5 invasive MBC patients and 149 MCs from 13 invasive FBC patients. Results show that hydroxyapatite is the most abundant type of calcium both in MBC and FBC. However, some differences in the amount and distribution of calcium minerals are present between the two groups. Besides, we observed that MCs in MBC have a higher amount of organic material (collagen) than FBC. To the best of our knowledge, this study provides the first overview of the composition of MCs present in MBC patients; and suggests that these patients have specific features that differentiate them from the previously studied FBC. Our result support thus the need for studies designed explicitly to the understanding of MBC.
Collapse
Affiliation(s)
| | - Francesca Piccotti
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Federico Sottotetti
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Chiara Guerra
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Sara Albasini
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Manuela Agozzino
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Laura Villani
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy
| | - Fabio Corsi
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100 Pavia, Italy; Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy.
| |
Collapse
|
30
|
Bansal GJ, Emanuel L, Kanagasabai S. Malignancy risk of indeterminate mammographic calcification in symptomatic breast clinics. Postgrad Med J 2021; 99:postgradmedj-2021-140835. [PMID: 34815330 DOI: 10.1136/postgradmedj-2021-140835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/30/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND To explore the potential risk factors predicting malignancy in patients with indeterminate incidental mammographic microcalcification and to evaluate the short-term risk of developing malignancy. METHODS Between January 2011 and December 2015, one hundred and fifty (150) consecutive patients with indeterminate mammographic microcalcifications who had undergone stereotactic biopsy were evaluated. Clinical and mammographic features were recorded and compared with histopathological biopsy results. In patients with malignancy, postsurgical findings and surgical upgrade, if any, were recorded. Linear regression analysis (SPSS V.25) was used to evaluate significant variables predicting malignancy. OR with 95% CIs was calculated for all variables. All patients were followed up for a maximum of 10 years. The mean age of the patients was 52 years (range 33-79 years). RESULTS There were a total of 55 (37%) malignant results in this study cohort. Age was an independent predictor of breast malignancy with an OR (95% CI) of 1.10 (1.03 to 1.16). Mammographic microcalcification size, pleomorphic morphology, multiple clusters and linear/segmental distribution were significantly associated with malignancy with OR (CI) of 1.03 (1.002 to 1.06), 6.06 (2.24 to 16.66), 6.35 (1.44 to 27.90) and 4.66 (1.07 to 20.19). The regional distribution of microcalcification had an OR of 3.09 (0.92 to 10.3), but this was not statistically significant. Patients with previous breast biopsies had a lower risk of breast malignancy than patients with no prior biopsy (p=0.034). CONCLUSION Multiple clusters, linear/segmental distribution, pleomorphic morphology, size of mammographic microcalcifications and increasing age were independent predictors of malignancy. Having a previous breast biopsy did not increase malignancy risk.
Collapse
Affiliation(s)
- Gaurav J Bansal
- The Breast Centre, University Hospital of Llandough, Cardiff and Vale University Health Board, Cardiff, UK
| | - Lauren Emanuel
- The Breast Centre, University Hospital of Llandough, Cardiff and Vale University Health Board, Cardiff, UK
| | - Sesha Kanagasabai
- The Breast Centre, University Hospital of Llandough, Cardiff and Vale University Health Board, Cardiff, UK
| |
Collapse
|
31
|
Montanaro M, Scimeca M, Toschi N, Bonanno E, Giacobbi E, Servadei F, Ippoliti A, Santeusanio G, Mauriello A, Anemona L. Effects of Risk Factors on In Situ Expression of Proinflammatory Markers Correlated to Carotid Plaque Instability. Appl Immunohistochem Mol Morphol 2021; 29:741-749. [PMID: 34039839 DOI: 10.1097/pai.0000000000000947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/13/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Several studies demonstrated a role of active chronic inflammatory infiltrate in carotid plaques progression suggesting a possible link between cardiovascular risk factors and inflammation-related plaque instability. The aim of this study is therefore to evaluate the possible effects of cardiovascular risk factors on in situ expression of proinflammatory markers associated with carotid plaque instability. METHODS AND RESULTS A tissue microarray containing carotid plaques from 36 symptomatic (major stroke or transient ischemic attack) and 37 asymptomatic patients was built. Serial sections were employed to evaluate the expression of some inflammatory markers by immunohistochemistry [CD3, CD4a, CD8, CD20, CD86, CD163, interleukin (IL)-2, IL-6, IL-17]. Immunohistochemical data were analyzed to study the possible associations between in situ expression of inflammatory biomarker and the main cardiovascular risk factors. Our data demonstrated that plaque instability is associated with the high in situ expression of some cytokines, such as IL-2, IL-6, IL-17. Besides the female sex, none of the risk factors analyzed showed a significant association between the in situ expression of these markers and unstable plaques. A significant increase of IL-6-positive and IL-17-positive cells was observed in unstable atheromatous plaques of female patients, as compared with unstable plaques of male patients. CONCLUSIONS Plaque destabilization is certainly correlated with the presence of the major cardiovascular risk factors, however, our results showed that, with the exception of sex, their action in the evolutive process of plaque instability seems rather nonspecific, favoring a general release of proinflammatory cytokines.
Collapse
Affiliation(s)
| | - Manuel Scimeca
- Departments of Experimental Medicine
- University of San Raffaele
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Nicola Toschi
- Biomedicine and Prevention, University of Rome "Tor Vergata"
- Imaging Martinos Center for Biomedical Imaging
- Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Bouzy P, O'Grady S, Madupalli H, Tecklenburg M, Rogers K, Palombo F, Morgan MP, Stone N. A time-course Raman spectroscopic analysis of spontaneous in vitro microcalcifications in a breast cancer cell line. J Transl Med 2021; 101:1267-1280. [PMID: 34117364 PMCID: PMC8367820 DOI: 10.1038/s41374-021-00619-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 01/05/2023] Open
Abstract
Microcalcifications are early markers of breast cancer and can provide valuable prognostic information to support clinical decision-making. Current detection of calcifications in breast tissue is based on X-ray mammography, which involves the use of ionizing radiation with potentially detrimental effects, or MRI scans, which have limited spatial resolution. Additionally, these techniques are not capable of discriminating between microcalcifications from benign and malignant lesions. Several studies show that vibrational spectroscopic techniques are capable of discriminating and classifying breast lesions, with a pathology grade based on the chemical composition of the microcalcifications. However, the occurrence of microcalcifications in the breast and the underlying mineralization process are still not fully understood. Using a previously established model of in vitro mineralization, the MDA-MB-231 human breast cancer cell line was induced using two osteogenic agents, inorganic phosphate (Pi) and β-glycerophosphate (βG), and direct monitoring of the mineralization process was conducted using Raman micro-spectroscopy. MDA-MB-231 cells cultured in a medium supplemented with Pi presented more rapid mineralization (by day 3) than cells exposed to βG (by day 11). A redshift of the phosphate stretching peak for cells supplemented with βG revealed the presence of different precursor phases (octacalcium phosphate) during apatite crystal formation. These results demonstrate that Raman micro-spectroscopy is a powerful tool for nondestructive analysis of mineral species and can provide valuable information for evaluating mineralization dynamics and any associated breast cancer progression, if utilized in pathological samples.
Collapse
Affiliation(s)
- Pascaline Bouzy
- School of Physics and Astronomy, University of Exeter, Exeter, UK
| | - Shane O'Grady
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, 2, Ireland
| | - Honey Madupalli
- Department of Chemistry and Biochemistry and Science of Advanced Materials Program, Central Michigan University, Mt. Pleasant, MI, USA
| | - Mary Tecklenburg
- Department of Chemistry and Biochemistry and Science of Advanced Materials Program, Central Michigan University, Mt. Pleasant, MI, USA
| | - Keith Rogers
- Cranfield Forensic Institute, Cranfield University, Shrivenham, UK
| | | | - Maria P Morgan
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, 2, Ireland
| | - Nicholas Stone
- School of Physics and Astronomy, University of Exeter, Exeter, UK.
| |
Collapse
|
33
|
Azam S, Eriksson M, Sjölander A, Gabrielson M, Hellgren R, Czene K, Hall P. Mammographic microcalcifications and risk of breast cancer. Br J Cancer 2021; 125:759-765. [PMID: 34127810 PMCID: PMC8405644 DOI: 10.1038/s41416-021-01459-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/18/2021] [Accepted: 06/02/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Mammographic microcalcifications are considered early signs of breast cancer (BC). We examined the association between microcalcification clusters and the risk of overall and subtype-specific BC. Furthermore, we studied how mammographic density (MD) influences the association between microcalcification clusters and BC risk. METHODS We used a prospective cohort (n = 53,273) of Swedish women with comprehensive information on BC risk factors and mammograms. The total number of microcalcification clusters and MD were measured using a computer-aided detection system and the STRATUS method, respectively. Cox regressions and logistic regressions were used to analyse the data. RESULTS Overall, 676 women were diagnosed with BC. Women with ≥3 microcalcification clusters had a hazard ratio [HR] of 2.17 (95% confidence interval [CI] = 1.57-3.01) compared to women with no clusters. The estimated risk was more pronounced in premenopausal women (HR = 2.93; 95% CI = 1.67-5.16). For postmenopausal women, microcalcification clusters and MD had a similar influence on BC risk. No interaction was observed between microcalcification clusters and MD. Microcalcification clusters were significantly associated with in situ breast cancer (odds ratio: 2.03; 95% CI = 1.13-3.63). CONCLUSIONS Microcalcification clusters are an independent risk factor for BC, with a higher estimated risk in premenopausal women. In postmenopausal women, microcalcification clusters have a similar association with BC as baseline MD.
Collapse
Affiliation(s)
- Shadi Azam
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Arvid Sjölander
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Marike Gabrielson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Roxanna Hellgren
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.,Department of Mammography, South General Hospital, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.,Department of Oncology, South General Hospital, Stockholm, Sweden
| |
Collapse
|
34
|
Zubareva EY, Senchukova MA. Prognostic and predictive significance of osteopontin in malignant neoplasms. ADVANCES IN MOLECULAR ONCOLOGY 2021. [DOI: 10.17650/2313-805x-2021-8-2-23-28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Osteopontin is an extracellular matrix protein which is produced by different types of cells and plays an important functional role in many biological processes. This review discusses the main functions of osteopontin, its role in the progression and chemoresistance of malignant neoplasms, in the regulation of epithelial-mesenchymal transition, angiogenesis, and the body’s immune response to the tumor. The article considers the currently known mechanisms by which osteopontin affects to the survival, mobility and invasion of tumor cells, to tumor sensitivity to drug treatment, as well as the prospects for a integrated study of the predictive significance of osteopontin, markers of hypoxia, angiogenesis, epithelial- mesenchymal transition, and immunological tolerance.
Collapse
Affiliation(s)
- E. Yu. Zubareva
- Orenburg Regional Clinical Oncological Dispensary; Orenburg State Medical University
| | - M. A. Senchukova
- Orenburg Regional Clinical Oncological Dispensary; Orenburg State Medical University
| |
Collapse
|
35
|
Unravelling the Encapsulation of DNA and Other Biomolecules in HAp Microcalcifications of Human Breast Cancer Tissues by Raman Imaging. Cancers (Basel) 2021; 13:cancers13112658. [PMID: 34071374 PMCID: PMC8198780 DOI: 10.3390/cancers13112658] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 01/01/2023] Open
Abstract
Simple Summary Although microcalcifications can be considered one of the first indicators of suspicious cancer lesions, depending on their morphology and distribution, the formation of hydroxyapatite calcifications and their relationship with malignancy remains unknown. In this work, we investigate in depth the biochemical composition of breast cancer microcalcifications, combining Raman spectroscopy imaging and advanced multivariate analysis. We demonstrate that DNA is naturally adsorbed and encapsulated inside hydroxyapatite found in breast cancer tissue. Furthermore, we also show the encapsulation of other relevant biomolecules such as lipids, proteins, cytochrome C and polysaccharides. The demonstration of the natural DNA biomineralization in cancer tissues represents an unprecedented advance in the field, as it can pave the way to understanding the role of hydroxyapatite in malignant tissues. Abstract Microcalcifications are detected through mammography screening and, depending on their morphology and distribution (BI-RADS classification), they can be considered one of the first indicators of suspicious cancer lesions. However, the formation of hydroxyapatite (HAp) calcifications and their relationship with malignancy remains unknown. In this work, we report the most detailed three-dimensional biochemical analysis of breast cancer microcalcifications to date, combining 3D Raman spectroscopy imaging and advanced multivariate analysis in order to investigate in depth the molecular composition of HAp calcifications found in 26 breast cancer tissue biopsies. We demonstrate that DNA has been naturally adsorbed and encapsulated inside HAp microcalcifications. Furthermore, we also show the encapsulation of other relevant biomolecules in HAp calcifications, such as lipids, proteins, cytochrome C and polysaccharides. The demonstration of natural DNA biomineralization, particularly in the tumor microenvironment, represents an unprecedented advance in the field, as it can pave the way to understanding the role of HAp in malignant tissues.
Collapse
|
36
|
Louro J, Román M, Posso M, Vázquez I, Saladié F, Rodriguez-Arana A, Quintana MJ, Domingo L, Baré M, Marcos-Gragera R, Vernet-Tomas M, Sala M, Castells X. Developing and validating an individualized breast cancer risk prediction model for women attending breast cancer screening. PLoS One 2021; 16:e0248930. [PMID: 33755692 PMCID: PMC7987139 DOI: 10.1371/journal.pone.0248930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Several studies have proposed personalized strategies based on women's individual breast cancer risk to improve the effectiveness of breast cancer screening. We designed and internally validated an individualized risk prediction model for women eligible for mammography screening. METHODS Retrospective cohort study of 121,969 women aged 50 to 69 years, screened at the long-standing population-based screening program in Spain between 1995 and 2015 and followed up until 2017. We used partly conditional Cox proportional hazards regression to estimate the adjusted hazard ratios (aHR) and individual risks for age, family history of breast cancer, previous benign breast disease, and previous mammographic features. We internally validated our model with the expected-to-observed ratio and the area under the receiver operating characteristic curve. RESULTS During a mean follow-up of 7.5 years, 2,058 women were diagnosed with breast cancer. All three risk factors were strongly associated with breast cancer risk, with the highest risk being found among women with family history of breast cancer (aHR: 1.67), a proliferative benign breast disease (aHR: 3.02) and previous calcifications (aHR: 2.52). The model was well calibrated overall (expected-to-observed ratio ranging from 0.99 at 2 years to 1.02 at 20 years) but slightly overestimated the risk in women with proliferative benign breast disease. The area under the receiver operating characteristic curve ranged from 58.7% to 64.7%, depending of the time horizon selected. CONCLUSIONS We developed a risk prediction model to estimate the short- and long-term risk of breast cancer in women eligible for mammography screening using information routinely reported at screening participation. The model could help to guiding individualized screening strategies aimed at improving the risk-benefit balance of mammography screening programs.
Collapse
Affiliation(s)
- Javier Louro
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Servei d’Epidemiologia i Avaluació, Hospital del Mar, Barcelona, Spain
- European Higher Education Area (EHEA) Doctoral Programme in Methodology of Biomedical Research and Public Health in Department of Pediatrics, Obstetrics and Gynecology, Preventive Medicine and Public Health, Universitat Autónoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Marta Román
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Servei d’Epidemiologia i Avaluació, Hospital del Mar, Barcelona, Spain
- * E-mail:
| | - Margarita Posso
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Servei d’Epidemiologia i Avaluació, Hospital del Mar, Barcelona, Spain
| | | | - Francina Saladié
- Cancer Epidemiology and Prevention Service, Hospital Universitari Sant Joan de Reus, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | | | - M. Jesús Quintana
- Department of Clinical Epidemiology and Public Health, University Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Laia Domingo
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Servei d’Epidemiologia i Avaluació, Hospital del Mar, Barcelona, Spain
| | - Marisa Baré
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Clinical Epidemiology and Cancer Screening, Parc Taulí University Hospital, Sabadell, Spain
| | - Rafael Marcos-Gragera
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Department of Health, Epidemiology Unit and Girona Cancer Registry, Oncology Coordination Plan, Autonomous Government of Catalonia, Catalan Institute of Oncology, Girona, Spain
| | | | - Maria Sala
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Servei d’Epidemiologia i Avaluació, Hospital del Mar, Barcelona, Spain
| | - Xavier Castells
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
- Servei d’Epidemiologia i Avaluació, Hospital del Mar, Barcelona, Spain
| | | |
Collapse
|
37
|
Arizmendi-Izazaga A, Navarro-Tito N, Jiménez-Wences H, Mendoza-Catalán MA, Martínez-Carrillo DN, Zacapala-Gómez AE, Olea-Flores M, Dircio-Maldonado R, Torres-Rojas FI, Soto-Flores DG, Illades-Aguiar B, Ortiz-Ortiz J. Metabolic Reprogramming in Cancer: Role of HPV 16 Variants. Pathogens 2021; 10:pathogens10030347. [PMID: 33809480 PMCID: PMC7999907 DOI: 10.3390/pathogens10030347] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/13/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic reprogramming is considered one of the hallmarks in cancer and is characterized by increased glycolysis and lactate production, even in the presence of oxygen, which leads the cancer cells to a process called “aerobic glycolysis” or “Warburg effect”. The E6 and E7 oncoproteins of human papillomavirus 16 (HPV 16) favor the Warburg effect through their interaction with a molecule that regulates cellular metabolism, such as p53, retinoblastoma protein (pRb), c-Myc, and hypoxia inducible factor 1α (HIF-1α). Besides, the impact of the E6 and E7 variants of HPV 16 on metabolic reprogramming through proteins such as HIF-1α may be related to their oncogenicity by favoring cellular metabolism modifications to satisfy the energy demands necessary for viral persistence and cancer development. This review will discuss the role of HPV 16 E6 and E7 variants in metabolic reprogramming and their contribution to developing and preserving the malignant phenotype of cancers associated with HPV 16 infection.
Collapse
Affiliation(s)
- Adán Arizmendi-Izazaga
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (N.N.-T.); (M.O.-F.)
| | - Hilda Jiménez-Wences
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Miguel A. Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
| | - Dinorah N. Martínez-Carrillo
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
| | - Ana E. Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (N.N.-T.); (M.O.-F.)
| | - Roberto Dircio-Maldonado
- Laboratorio de Investigación Clínica, Facultad de Ciencias, Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico;
- Laboratorio de Diagnóstico e Investigación en Salud, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Francisco I. Torres-Rojas
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Diana G. Soto-Flores
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
- Laboratorio de Diagnóstico e Investigación en Salud, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (A.A.-I.); (M.A.M.-C.); (A.E.Z.-G.); (F.I.T.-R.); (D.G.S.-F.); (B.I.-A.)
- Laboratorio de Investigación en Biomoléculas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas S/N, Ciudad Universitaria, Colonia La Haciendita, Chilpancingo C.P. 39090, Guerrero, Mexico; (H.J.-W.); (D.N.M.-C.)
- Correspondence: ; Tel.: +52-747-471-0901
| |
Collapse
|
38
|
Azam S, Eriksson M, Sjölander A, Gabrielson M, Hellgren R, Czene K, Hall P. Predictors of mammographic microcalcifications. Int J Cancer 2021; 148:1132-1143. [PMID: 32949149 PMCID: PMC7821182 DOI: 10.1002/ijc.33302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/28/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022]
Abstract
We examined the association between established risk factors for breast cancer and microcalcification clusters and their asymmetry. A cohort study of 53 273 Swedish women aged 30 to 80 years, with comprehensive information on breast cancer risk factors and mammograms, was conducted. Total number of microcalcification clusters and the average mammographic density area were measured using a Computer Aided Detection system and the STRATUS method, respectively. A polygenic risk score for breast cancer, including 313 single nucleotide polymorphisms, was calculated for those women genotyped (N = 7387). Odds ratios (ORs) and 95% confidence intervals (CIs), with adjustment for potential confounders, were estimated. Age was strongly associated with microcalcification clusters. Both high mammographic density (>40 cm2 ), and high polygenic risk score (80-100 percentile) were associated with microcalcification clusters, OR = 2.08 (95% CI = 1.93-2.25) and OR = 1.22 (95% CI = 1.06-1.48), respectively. Among reproductive risk factors, life-time breastfeeding duration >1 year was associated with microcalcification clusters OR = 1.22 (95% CI = 1.03-1.46). The association was confined to postmenopausal women. Among lifestyle risk factors, women with a body mass index ≥30 kg/m2 had the lowest risk of microcalcification clusters OR = 0.79 (95% CI = 0.73-0.85) and the association was stronger among premenopausal women. Our results suggest that age, mammographic density, genetic predictors of breast cancer, having more than two children, longer duration of breast-feeding are significantly associated with increased risk of microcalcification clusters. However, most lifestyle risk factors for breast cancer seem to protect against presence of microcalcification clusters. More research is needed to study biological mechanisms behind microcalcifications formation.
Collapse
Affiliation(s)
- Shadi Azam
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Mikael Eriksson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Arvid Sjölander
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Marike Gabrielson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Roxanna Hellgren
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- Department of MammographySouth General HospitalStockholmSweden
| | - Kamila Czene
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Per Hall
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- Department of OncologySouth General HospitalStockholmSweden
| |
Collapse
|
39
|
Vidavsky N, Kunitake JAMR, Estroff LA. Multiple Pathways for Pathological Calcification in the Human Body. Adv Healthc Mater 2021; 10:e2001271. [PMID: 33274854 PMCID: PMC8724004 DOI: 10.1002/adhm.202001271] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/16/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization of skeletal components (e.g., bone and teeth) is generally accepted to occur under strict cellular regulation, leading to mineral-organic composites with hierarchical structures and properties optimized for their designated function. Such cellular regulation includes promoting mineralization at desired sites as well as inhibiting mineralization in soft tissues and other undesirable locations. In contrast, pathological mineralization, with potentially harmful health effects, can occur as a result of tissue or metabolic abnormalities, disease, or implantation of certain biomaterials. This progress report defines mineralization pathway components and identifies the commonalities (and differences) between physiological (e.g., bone remodeling) and pathological calcification formation pathways, based, in part, upon the extent of cellular control within the system. These concepts are discussed in representative examples of calcium phosphate-based pathological mineralization in cancer (breast, thyroid, ovarian, and meningioma) and in cardiovascular disease. In-depth mechanistic understanding of pathological mineralization requires utilizing state-of-the-art materials science imaging and characterization techniques, focusing not only on the final deposits, but also on the earlier stages of crystal nucleation, growth, and aggregation. Such mechanistic understanding will further enable the use of pathological calcifications in diagnosis and prognosis, as well as possibly provide insights into preventative treatments for detrimental mineralization in disease.
Collapse
Affiliation(s)
- Netta Vidavsky
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Jennie A M R Kunitake
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY, 14853, USA
| |
Collapse
|
40
|
The impact of oral contraceptive use on breast cancer risk: State of the art and future perspectives in the era of 4P medicine. Semin Cancer Biol 2021; 72:11-18. [PMID: 33454355 DOI: 10.1016/j.semcancer.2020.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
Breast cancer is the most commonly occurring cancer in women, the second most frequent cancer overall, and it causes the greatest number of cancer-related deaths among women. The significant increased concern of breast cancer worldwide may be attributed to the prolonged life expectancy and the adoption of the western lifestyle with its related risks factors. A woman's risk for breast cancer is linked to her reproductive history and with her lifetime hormonal exposure. Among the known risk factors for breast cancer, several studies investigated the possible role of the assumption of hormonal "pills" in both breast cancer incidence and development. Nevertheless, data about the association between the assumption of oral contraceptives and breast cancer incidence are still controversial and not conclusive. Given the public health importance of breast cancer and the popularity of hormonal "pills" as contraceptive, the impact of oral contraceptive use on breast cancer risk assumes relevance from both a clinical and a social point of view. Therefore, in this review we wanted to illustrate this issue by addressing the following major themes: a) the role of sex steroid hormones in female breast development and carcinogenesis; b) the clinical impact of hormonal oral contraception according to the state of the art literature; c) the actual scientific debate and future perspectives.
Collapse
|
41
|
Molecular Aspects and Prognostic Significance of Microcalcifications in Human Pathology: A Narrative Review. Int J Mol Sci 2020; 22:ijms22010120. [PMID: 33374380 PMCID: PMC7795544 DOI: 10.3390/ijms22010120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
The presence of calcium deposits in human lesions is largely used as imaging biomarkers of human diseases such as breast cancer. Indeed, the presence of micro- or macrocalcifications is frequently associated with the development of both benign and malignant lesions. Nevertheless, the molecular mechanisms involved in the formation of these calcium deposits, as well as the prognostic significance of their presence in human tissues, have not been completely elucidated. Therefore, a better characterization of the biological process related to the formation of calcifications in different tissues and organs, as well as the understanding of the prognostic significance of the presence of these calcium deposits into human tissues could significantly improve the management of patients characterized by microcalcifications associated lesions. Starting from these considerations, this narrative review highlights the most recent histopathological and molecular data concerning the formation of calcifications in breast, thyroid, lung, and ovarian diseases. Evidence reported here could deeply change the current point of view concerning the role of ectopic calcifications in the progression of human diseases and also in the patients’ management. In fact, the presence of calcifications can suggest an unfavorable prognosis due to dysregulation of normal tissues homeostasis.
Collapse
|
42
|
Calabrò ML, Lazzari N, Rigotto G, Tonello M, Sommariva A. Role of Epithelial-Mesenchymal Plasticity in Pseudomyxoma Peritonei: Implications for Locoregional Treatments. Int J Mol Sci 2020; 21:ijms21239120. [PMID: 33266161 PMCID: PMC7731245 DOI: 10.3390/ijms21239120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanisms by which neoplastic cells disseminate from the primary tumor to metastatic sites, so-called metastatic organotropism, remain poorly understood. Epithelial-mesenchymal transition (EMT) plays a role in cancer development and progression by converting static epithelial cells into the migratory and microenvironment-interacting mesenchymal cells, and by the modulation of chemoresistance and stemness of tumor cells. Several findings highlight that pathways involved in EMT and its reverse process (mesenchymal-epithelial transition, MET), now collectively called epithelial-mesenchymal plasticity (EMP), play a role in peritoneal metastases. So far, the relevance of factors linked to EMP in a unique peritoneal malignancy such as pseudomyxoma peritonei (PMP) has not been fully elucidated. In this review, we focus on the role of epithelial-mesenchymal dynamics in the metastatic process involving mucinous neoplastic dissemination in the peritoneum. In particular, we discuss the role of expression profiles and phenotypic transitions found in PMP in light of the recent concept of EMP. A better understanding of EMP-associated mechanisms driving peritoneal metastasis will help to provide a more targeted approach for PMP patients selected for locoregional interventions involving cytoreductive surgery and hyperthermic intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (N.L.); (G.R.)
- Correspondence:
| | - Nayana Lazzari
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (N.L.); (G.R.)
| | - Giulia Rigotto
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (N.L.); (G.R.)
| | - Marco Tonello
- Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy;
| | - Antonio Sommariva
- Advanced Surgical Oncology, Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy;
| |
Collapse
|
43
|
Tsai HT, Huang CS, Tu CC, Liu CY, Huang CJ, Ho YS, Tu SH, Tseng LM, Huang CC. Multi-gene signature of microcalcification and risk prediction among Taiwanese breast cancer. Sci Rep 2020; 10:18276. [PMID: 33106505 PMCID: PMC7588423 DOI: 10.1038/s41598-020-74982-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022] Open
Abstract
Microcalcification is one of the most common radiological and pathological features of breast ductal carcinoma in situ (DCIS), and to a lesser extent, invasive ductal carcinoma. We evaluated messenger RNA (mRNA) transcriptional profiles associated with ectopic mammary mineralization. A total of 109 breast cancers were assayed with oligonucleotide microarrays. The associations of mRNA abundance with microcalcifications and relevant clinical features were evaluated. Microcalcifications were present in 86 (79%) patients by pathological examination, and 81 (94%) were with coexistent DCIS, while only 13 (57%) of 23 patients without microcalcification, the invasive diseases were accompanied with DCIS (χ2-test, P < 0.001). There were 69 genes with differential mRNA abundance between breast cancers with and without microcalcifications, and 11 were associated with high-grade (comedo) type DCIS. Enriched Gene Ontology categories included glycosaminoglycan and aminoglycan metabolic processes and protein ubiquitination, indicating an active secretory process. The intersection (18 genes) of microcalcificaion-associated and DCIS-associated genes provided the best predictive accuracy of 82% with Bayesian compound covariate predictor. Ten genes were further selected for prognostic index score construction, and five-year relapse free survival was 91% for low-risk and 83% for high-risk group (log-rank test, P = 0.10). Our study suggested that microcalcification is not only the earliest detectable radiological sign for mammography screening but the phenomenon itself may reflect the underling events during mammary carcinogenesis. Future studies to evaluate the prognostic significance of microcalcifications are warranted.
Collapse
Affiliation(s)
- Hsin-Tien Tsai
- Division of General Surgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Ching-Shui Huang
- Division of General Surgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Chiang Tu
- Department of Surgery, Fu-Jen Catholic University Hospital, New Taipei, Taiwan.,School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Chih-Yi Liu
- Division of Pathology, Cathay General Hospital Sijhih, New Taipei, Taiwan
| | - Chi-Jung Huang
- Department of Medical Research, Cathay General Hospital, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,Department of Medical Laboratory, Taipei Medical University Hospital, Taipei, Taiwan.,School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shih-Hsin Tu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,Division of Breast Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei City, 11217, Taiwan, ROC. .,School of Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Chi-Cheng Huang
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei City, 11217, Taiwan, ROC.
| |
Collapse
|
44
|
De Santis S, Sotgiu G, Crescenzi A, Taffon C, Felici AC, Orsini M. On the chemical composition of psammoma bodies microcalcifications in thyroid cancer tissues. J Pharm Biomed Anal 2020; 190:113534. [PMID: 32841781 DOI: 10.1016/j.jpba.2020.113534] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023]
Abstract
Recently the knowledge of chemical composition of pathological mineralizations is an important topic extensively studied because it could give more in-depth information to understand pathologies themselves and to improve prevention methods. In this work, psammoma bodies (PBs) microcalcifications in thyroid cancer tissue are investigated by different and complementary analytical methods as: micro-Fourier transformed spectroscopy, X-ray fluorescence spectroscopy, Inductively Coupled plasma Optical Emission Spectroscopy (ICP-OES) and scanning electron microscopy (SEM) with energy dispersive X-ray spectroscopy imaging (EDX). For the first time the micro-FTIR analysis of the only inorganic phase isolated from PBs was reported. Signals of the recorded spectrum showed that the main component of the calcifications is the amorphous carbonated calcium phosphate, and the IR spectrum of thyroid PBs is strongly consistent with that of PBs in human ovarian tumors. The XRF and the ICP analysis detected also the presence of iron ad zinc in thyroid PBs. These results are validated by scanning electron microscopy (SEM) with energy dispersive X-ray spectroscopy imaging (EDX) carried out on tissue samples of the papillary thyroid carcinoma. By these analytical methods magnesium and sodium were detected within PBs while the presence of iron was confirmed by the Perls test. Summarizing the results of applied analytical methods, the main detected elements within the thyroid psammoma bodies are Ca, P, Mg, Na, Fe and Zn. Magnesium and sodium are found in malignant breast cancer microcalcifications, thus they seem correlated to neoplastic transformation. The Fe and Zn elements could give information about the origin of these pathological microcalcifications.
Collapse
Affiliation(s)
- Serena De Santis
- Department of Engineering, Roma Tre University, via Vito Volterra 62, Roma Italy.
| | - Giovanni Sotgiu
- Department of Engineering, Roma Tre University, via Vito Volterra 62, Roma Italy
| | - Anna Crescenzi
- Pathology Unit, University Hospital Campus Bio-Medico, Rome Italy
| | - Chiara Taffon
- Pathology Unit, University Hospital Campus Bio-Medico, Rome Italy
| | - Anna Candita Felici
- Basic and Applied Sciences for Engineering, Sapienza University, Via A. Scarpa 16, Roma Italy
| | - Monica Orsini
- Department of Engineering, Roma Tre University, via Vito Volterra 62, Roma Italy
| |
Collapse
|
45
|
Martins ML, Pinto TS, Gomes AM, Parra JPRLL, Franchi GC, Zambuzzi WF, Rodrigues CG. Immobilization of Paclitaxel on Hydroxyapatite for Breast Cancer Investigations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:8723-8732. [PMID: 32643936 DOI: 10.1021/acs.langmuir.0c00868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A simple method for immobilization of the chemotherapy drug paclitaxel (PTX) on hydroxyapatite nanoparticles (n-HAP) using the biopolymer chitosan as a trapping agent is described focusing on applications involving breast cancer cells. n-HAP with two distinct crystallinity profiles were used: with predominant crystallization along the long axis and with a more homogeneous crystallization in all directions. In the first scenario, the interactions between chitosan and both the OH and PO43- groups on the surface of the nanoparticles are favored and lead to a more efficient attachment of the drug. In this case, PTX is found to remain mostly attached to the n-HAP for at least 24 h, while being dispersed in aqueous solution. During this time, the activity of the drug is inhibited as corroborated by in vitro assays with breast cancer cells. With that, the in vitro experiments revealed distinct effects from the drug-loaded nanoparticles on the cells depending on the experimental conditions. In a short term, that is, in 24 h, the cells exhibit higher viability than those challenged with nonloaded materials. Nevertheless, after 72 h, even a small content of PTX in the presence of n-HAP can reduce the cells' viability via stimulation of the apoptotic phenotype and suppression of survival stimuli.
Collapse
Affiliation(s)
- Murillo L Martins
- Post-Graduation Program in Industrial and Systems Engineering, Pontifical Catholic University of Goiás, Goiânia 74175-120, Goiás, Brazil
| | - Thais S Pinto
- Laboratory of Bioassays and Cellular Dynamics (LaBIO), Chemistry and Biochemistry Department, Bioscience Institute of Botucatu (IBB), State University of São Paulo (UNESP), Botucatu 18618-000, São Paulo, Brazil
| | - Anderson M Gomes
- Laboratory of Bioassays and Cellular Dynamics (LaBIO), Chemistry and Biochemistry Department, Bioscience Institute of Botucatu (IBB), State University of São Paulo (UNESP), Botucatu 18618-000, São Paulo, Brazil
| | - João P R L L Parra
- Laboratory of Bioassays and Cellular Dynamics (LaBIO), Chemistry and Biochemistry Department, Bioscience Institute of Botucatu (IBB), State University of São Paulo (UNESP), Botucatu 18618-000, São Paulo, Brazil
| | - Gilberto C Franchi
- Onco-Hematological Child Research Center (CIPOI), Faculty of Medical Sciences, University of Campinas-UNICAMP, Campinas 13083-970, São Paulo, Brazil
| | - Willian F Zambuzzi
- Laboratory of Bioassays and Cellular Dynamics (LaBIO), Chemistry and Biochemistry Department, Bioscience Institute of Botucatu (IBB), State University of São Paulo (UNESP), Botucatu 18618-000, São Paulo, Brazil
| | - Cloves G Rodrigues
- Post-Graduation Program in Industrial and Systems Engineering, Pontifical Catholic University of Goiás, Goiânia 74175-120, Goiás, Brazil
- School of Exact Sciences and Computing, Pontifical Catholic University of Goiás, Goiânia 74175-120, Goiás, Brazil
| |
Collapse
|
46
|
Fushimi A, Takeyama H, Tachibana T, Manome Y. Osteogenic cocktail induces calcifications in human breast cancer cell line via placental alkaline phosphatase expression. Sci Rep 2020; 10:12669. [PMID: 32728117 PMCID: PMC7391638 DOI: 10.1038/s41598-020-69622-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/14/2020] [Indexed: 01/27/2023] Open
Abstract
Breast cancer is frequently characterized by calcifications in mammography. The mechanism for calcifications in breast cancer is not completely known. Understanding this mechanism will improve diagnostic accuracy. Herein, we demonstrated that calcifications occur and that alkaline phosphatase enzyme activity increases in MDA-MB-231 cells cultured using an osteogenic cocktail-containing medium. Microarray transcript analysis showed that the PI3K-Akt signaling pathway was significantly involved, with recruitment of placental alkaline phosphatase. Calcifications and alkaline phosphatase enzyme activity were suppressed by silencing placental alkaline phosphatase using a small interfering RNA. Inhibition of the PI3K-Akt signaling pathway suppressed phospho-c-Jun and placental alkaline phosphatase and resulted in absence of calcifications. These findings reveal that breast cancer cells acquire alkaline phosphatase enzyme activity via placental alkaline phosphatase expression and suggest that breast calcification formation is closely associated with the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Atsushi Fushimi
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, Japan. .,Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan.
| | - Hiroshi Takeyama
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshiaki Tachibana
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, Japan
| | - Yoshinobu Manome
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, Japan
| |
Collapse
|
47
|
BMP-2 Variants in Breast Epithelial to Mesenchymal Transition and Microcalcifications Origin. Cells 2020; 9:cells9061381. [PMID: 32498363 PMCID: PMC7348762 DOI: 10.3390/cells9061381] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
This study aims to investigate the possible different roles of the BMP-2 variants, cytoplasmic and nuclear variant, in both epithelial to mesenchymal transition and in microcalcifications origin in human breast cancers. To this end, the in situ expression of cytoplasmic and nuclear BMP-2 was associated with the expression of the main epithelial to mesenchymal transition biomarkers (e-cadherin and vimentin) and molecules involved in bone metabolisms (RUNX2, RANKL, SDF-1) by immunohistochemistry. In addition, the expression of cytoplasmic and nuclear BMP-2 was associated with the presence of microcalcifications. Our data showed a significant association among the number of cytoplasmic BMP-2-positive cells and the number of both vimentin (positive association) and e-cadherin (negative association) positive breast cells. Conversely, no associations were found concerning the nuclear BMP-2-positive breast cells. Surprisingly, the opposite result was obtained by analyzing the variants of BMP-2 and both the expression of RANKL and SDF-1 and the presence of microcalcifications. Specifically, the presence of microcalcifications was related to the expression of nuclear BMP-2 variant rather than the cytoplasmic one, as well as a strong association between the number of nuclear BMP-2 and the expression of the main breast osteoblast-like cells (BOLCs) biomarkers. To further corroborate these data, an in vitro experiment for demonstrating the co-expression of nBMP-2 and RANKL or vimentin or SDF-1 in breast cancer cells that acquire the capability to produce microcalcifications was developed. These investigations confirmed the association between the nBMP-2 expression and both RANKL and SDF-1. The data supports the idea that whilst cytoplasmic BMP-2 can be involved in epithelial to mesenchymal transition phenomenon, the nuclear variant is related to the essential mechanisms for the formation of breast microcalcifications. In conclusion, from these experimental and translational perspectives, the complexity of BMP-2 signaling will require a detailed understanding of the involvement of specific BMP-2 variants in breast cancers.
Collapse
|
48
|
Chen Y, Zhou JH, Fan HX, Luo Y, Peng YL, Ma BY. Ultrasound Diagnosis of Breast Lymphoma and the Identification of Breast Infiltrating Ductal Carcinoma. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2020; 39:1203-1211. [PMID: 31891216 DOI: 10.1002/jum.15209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/20/2019] [Accepted: 12/08/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVES By analyzing the B-mode ultrasound and color Doppler flow imaging characteristics of breast lymphoma (BL) and breast infiltrating ductal carcinoma (BIDC), we expected to discriminate these diseases. METHODS Thirty-two patients with BL and 30 with BIDC confirmed pathologically were selected. The BL group was divided into nodular and diffuse groups. We analyzed and compared the general and imaging characteristics of the BL subgroups and the BIDC group. RESULTS The mean maximum diameter of BL was 54.93 ± 43.74 cm, and that of BIDC was 23.90 ± 6.79 cm (P < .05). The differences between the nodular BL and BIDC groups in a circumscribed margin (60.00% versus 20.00%), calcification (20.00% versus 53.33%), aggregation characteristics (0.00% versus 53.33%), and density (73.33% versus 10.00%) were statistically significant (P < .05). The differences between the diffuse BL and BIDC groups in calcification (6.67% versus 53.33%), aggregation characteristics (6.67% versus 53.33%) and density (40.00% versus 10.00%) were statistically significant (P < .05). The difference in a circumscribed margin (60% versus 13.33%) between the BL subgroups was statistically significant (P < .05). The blood flow signal in BL lesions was richer than that in BIDC lesions (P < .05). CONCLUSIONS Extrasuperior-quadrant single lesions in the BL group were larger than those in the BIDC group. The edges of the lesions in the nodular BL group were circumscribed and dense. Lesions in the diffuse BL group did not have a circumscribed margin, calcification, aggregation characteristics, or density. The blood flow signal in BL lesions was richer than that in BIDC lesions.
Collapse
Affiliation(s)
- Yang Chen
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory for Neuroinformation of the Ministry of Education, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie-Hong Zhou
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Hong-Xia Fan
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Luo
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Yu-Lan Peng
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Bu-Yun Ma
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Shin KS, Laohajaratsang M, Men S, Figueroa B, Dintzis SM, Fu D. Quantitative chemical imaging of breast calcifications in association with neoplastic processes. Am J Cancer Res 2020; 10:5865-5878. [PMID: 32483424 PMCID: PMC7254998 DOI: 10.7150/thno.43325] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022] Open
Abstract
Calcifications play an essential role in early breast cancer detection and diagnosis. However, information regarding the chemical composition of calcifications identified on mammography and histology is limited. Detailed spectroscopy reveals an association between the chemical composition of calcifications and breast cancer, warranting the development of novel analytical tools to better define calcification types. Previous investigations average calcification composition across broad tissue sections with no spatially resolved information or provide qualitative visualization, which prevents a robust linking of specific spatially resolved changes in calcification chemistry with the pathologic process. Method: To visualize breast calcification chemical composition at high spatial resolution, we apply hyperspectral stimulated Raman scattering (SRS) microscopy to study breast calcifications associated with a spectrum of breast changes ranging from benign to neoplastic processes, including atypical ductal hyperplasia, ductal carcinoma in situ, and invasive ductal carcinoma. The carbonate content of individual breast calcifications is quantified using a simple ratiometric analysis. Results: Our findings reveal that intra-sample calcification carbonate content is closely associated with local pathological processes. Single calcification analysis supports previous studies demonstrating decreasing average carbonate level with increasing malignant potential. Sensitivity and specificity reach >85% when carbonate content level is used as the single differentiator in separating benign from neoplastic processes. However, the average carbonate content is limiting when trying to separate specific diagnostic categories, such as fibroadenoma and invasive ductal carcinoma. Second harmonic generation (SHG) data can provide critical information to bridge this gap. Conclusion: SRS, combined with SHG, can be a valuable tool in better understanding calcifications in carcinogenesis, diagnosis, and possible prognosis. This study not only reveals previously unknown large variations of breast microcalcifications in association with local malignancy but also corroborates the clinical value of linking microcalcification chemistry to breast malignancy. More importantly, it represents an important step in the development of a label-free imaging strategy for breast cancer diagnosis with tremendous potential to address major challenges in diagnostic discordance in pathology.
Collapse
|
50
|
Saxena N, Chandra NC. Cholesterol: A Prelate in Cell Nucleus and its Serendipity. Curr Mol Med 2020; 20:692-707. [PMID: 32282300 DOI: 10.2174/1566524020666200413112030] [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: 10/08/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 11/22/2022]
Abstract
Cholesterol is a chameleon bio-molecule in cellular multiplex. It acts as a prelate in almost every cellular compartment with its site specific characteristics viz. regulation of structural veracity and scaffold fluidity of bio-membranes, insulation of electrical transmission in nerves, controlling of genes by making steroid endocrines, acting as precursors of metabolic regulators and many more with its emerging prophecy in the cell nucleus to drive new cell formation. Besides the crucial legacy in cellular functionality, cholesterol is ostracized as a member of LDL particle, which has been proved responsible to clog blood vessels. LDL particles get deposited in the blood vessels because of their poor clearance owing to the non-functioning LDL receptor on the vessel wall and surrounding tissues. Blocking of blood vessel promotes heart attack and stroke. On the other hand, cholesterol has been targeted as pro-cancerous molecule. At this phase again cholesterol is biphasic. Although cholesterol is essential to construct nuclear membrane and its lipid-rafts; in cancer tumour cells, cholesterol is not under the control of intracellular feedback regulation and gets accumulated within cell nucleus by crossing nuclear membrane and promoting cell proliferation. In precancerous stage, the immune cells also die because of the lack of requisite concentration of intracellular and intranuclear cholesterol pool. The existence of cholesterol within the cell nucleus has been found in the nuclear membrane, epichromosomal location and nucleoplasm. The existence of cholesterol in the microdomain of nuclear raft has been reported to be linked with gene transcription, cell proliferation and apoptosis. Hydrolysis of cholesterol esters in chromosomal domain is linked with new cell generation. Apparently, Cholesterol is now a prelate in cell nucleus too ------ A serendipity in cellular haven.
Collapse
Affiliation(s)
- Nimisha Saxena
- Department of Biochemistry, KDMCH & Research Center, Akbarpur, Mathura - 281406, India
| | - Nimai Chand Chandra
- Department of Biochemistry, All India Institute of Medical Sciences, Phulwarisharif, Patna - 801507, India
| |
Collapse
|