1
|
Alloush F, Bahmad HF, Deb A, Ocejo S, Valencia AK, Abulaban A, Krishnamurthy K, Alghamdi S, Poppiti R. Influence of Cancerization of Lobules in Ductal Carcinoma In Situ of the Breast on the Pathological Outcomes in Mastectomy Specimens. Cancers (Basel) 2025; 17:1634. [PMID: 40427132 PMCID: PMC12110246 DOI: 10.3390/cancers17101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Cancerization of lobules (COL) is defined as the involvement of lobular acini by ductal carcinoma in situ (DCIS). Whether it represents a morphological variation in DCIS or a secondary extension of DCIS into lobules is debatable. The relation between COL and the probability of invasion is conflicting among different studies. We assessed if COL is a predictor of adverse pathological outcomes in mastectomy specimens. We reviewed the clinicopathological data of patients who underwent partial or total mastectomy for DCIS during a 3-year period (January 2015 until December 2017). Pathological parameters and follow-up data were collected. Whole-tissue hematoxylin and eosin (H&E) slides were reviewed and re-evaluated for COL. Cases with COL were stained immunohistochemically for E-cadherin and p120 to confirm the ductal phenotype of the neoplasms. In total, 171 mastectomies were identified including 65 specimens with pure DCIS and 106 specimens with DCIS with invasive carcinoma. COL was identified in 73 specimens. COL was significantly associated with adverse pathological outcomes including higher DCIS nuclear grade (p-value = 0.006), central (expansive "comedo") necrosis (p-value = 0.008), presence of DCIS within or less than 2 mm from the surgical resection margin(s) (p-value = 0.004), higher percentage of blocks/slides with DCIS (p-value < 0.001), and extensive intraductal component (EIC) (applicable in cases with invasion) (p-value < 0.001). Invasion was seen in approximately two-thirds of the cases regardless of the presence of COL, with no statistical significance. Ninety-eight patients achieved 60 months of follow-up, of which only one patient developed local DCIS recurrence and had COL and EIC. Four other patients developed metastatic disease related to the invasive component. While other studies have previously hypothesized that COL may be associated with a worse pathological outcome at mastectomy, our results show that it may indeed be a measure of a higher disease burden representing EIC; however, it is not associated with an increased risk of detecting invasive carcinoma.
Collapse
Affiliation(s)
- Ferial Alloush
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
| | - Hisham F. Bahmad
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Arunima Deb
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
| | - Stephanie Ocejo
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.O.); (A.-K.V.)
| | - Ann-Katrin Valencia
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (S.O.); (A.-K.V.)
| | - Amr Abulaban
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Kritika Krishnamurthy
- Department of Pathology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY 10467, USA;
| | - Sarah Alghamdi
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
- Department of Pathology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Robert Poppiti
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (F.A.); (A.D.); (S.A.); (R.P.)
- Department of Pathology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
2
|
Tapia-Uriol P, Becerra-Goicochea L, Campos-Valderrama V, del Valle-Mendoza J, Aguilar-Luis MA, Silva-Caso WG. Characterization of intrinsic subtypes of breast cancer and their relationship with staging: an observational study. Front Med (Lausanne) 2025; 12:1553910. [PMID: 40417678 PMCID: PMC12098373 DOI: 10.3389/fmed.2025.1553910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
Background Breast cancer is one of the main causes of morbidity and mortality among women around the world. In Peru, it has recently surpassed cervical cancer as the most commonly reported cancer. Studying the relationship between intrinsic breast cancer subtypes and disease staging can optimize diagnosis, prognosis, and treatment. Therefore, there is a need for better risk stratification, selection of personalized treatment, and improved early detection strategies. We conducted this study to address the lack of data on underrepresented populations such as the Peruvian population. The objective of the study was to analyze the distribution of intrinsic subtypes of breast cancer and their correlation with prognostic factors and demographic characteristics among women in Peru. Methods A descriptive, retrospective observational study was conducted, analyzing 67 cases of breast cancer of various intrinsic subtypes diagnosed at a referral hospital in Peru. Clinical, demographic, and pathological data were collected, including histological type, intrinsic subtype, tumor stage, and geographic origin of the patients. Intrinsic subtypes were classified through immunohistochemistry, and the data were processed to determine their distribution and correlation with prognostic factors such as disease stage. Results The mean age of the 67 patients included in the study was 54.2 years. The majority of cases originated from the city of Cajamarca (56.7%, n = 38). Invasive breast carcinoma of no special type was the most common histological type (62.7%, n = 42). Among the intrinsic subtypes, luminal B was the most common (31.3%, n = 21), followed by luminal A and triple-negative (22.4%, n = 15), both with the same frequency. Furthermore, 16.4% (n = 11) of patients presented with metastasis at the time of evaluation. A high frequency of tumors was observed in Tumor, Nodes, Metastasis (TNM) stages 3 and 4, accounting for 49.2% (n = 33). Conclusion This study describes the heterogeneity of breast cancer based on the identification of intrinsic subtypes within the analyzed population. The high frequency of luminal B, luminal A, and triple-negative subtypes is notable. The highest frequency of identified cases was in the advanced stages, highlighting the need for personalized treatments and improved early detection strategies.
Collapse
Affiliation(s)
- Paola Tapia-Uriol
- Oncology Unit, Cajamarca Regional Teaching Hospital, Cajamarca, Peru
| | - Lorena Becerra-Goicochea
- Oncology Unit, Cajamarca Regional Teaching Hospital, Cajamarca, Peru
- Professional Academic School of Obstetrics, National University of Cajamarca, Cajamarca, Peru
| | | | - Juana del Valle-Mendoza
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| | - Miguel Angel Aguilar-Luis
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| | - Wilmer Gianfranco Silva-Caso
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| |
Collapse
|
3
|
Su L, Xie Q, Yi A, Zhang Q, Chen J. Ultrasound characteristics comparison and development of a predictive nomogram for intraductal papilloma and ductal carcinoma in situ: a retrospective cohort study. Front Oncol 2025; 15:1454951. [PMID: 40313250 PMCID: PMC12043480 DOI: 10.3389/fonc.2025.1454951] [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: 07/19/2024] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Background Intraductal Papilloma (IDP) and Ductal Carcinoma In Situ (DCIS) are significant benign and pre-invasive breast lesions, respectively. This study aimed to investigate ultrasound features and develop a predictive nomogram for discriminating between IDP and DCIS. Methods Conducted at Quanzhou First Hospital over a three-year period, 389 patients were enrolled with detailed ultrasound examinations and confirmed pathological diagnoses. IDP was classified into Grades 3, 4, and 5, whereas DCIS presented with a mass-like morphology. Patients meeting the inclusion criteria underwent rigorous analysis, with exclusion criteria eliminating those with incomplete imaging data or confounding comorbidities. Ultrasound characteristics, including lesion size, shape, margin, and echogenicity, etc., were systematically evaluated and compared between the two groups. Univariate and multivariate logistic regression analyses were conducted to identify significant risk factors. Subsequently, based on these characteristics, both static and dynamic nomograms were developed. The performance of the nomograms was evaluated using the area under the receiver operating characteristic curve (AUC), calibration plots, and decision curve analysis (DCA). Results The study cohort included 272 patients in the training set and 117 in the validation set. Significant differences were observed between IDP and DCIS in age, size, shape, aspect ratio, margin, duct dilatation, and microcalcification (P < 0.05). Logistic regression analyses identified age, size, aspect ratio, margin, microcalcification, and duct dilatation as independent risk factors. Compared to DCIS, IDP is associated with younger age, smaller size, clearer margins, fewer microcalcifications, and more ductal dilation. The performance of the nomogram developed to predict IDP and DCIS showed an AUC of 0.918 in the training set and 0.888 in the validation set. The calibration curve indicates a strong fit of the predictive model in the validation set, with the Hosmer-Lemeshow test showing high consistency between predicted and actual probabilities (training set, P = 0.875; validation set, P = 0.751). Additionally, DCA confirms the clinical utility of the model. Conclusion The nomogram incorporating key predictors provides a valuable tool for differentiating between IDP and DCIS based on ultrasound characteristics. This approach aids in clinical decision-making and potentially reduces unnecessary biopsies.
Collapse
Affiliation(s)
- Liyang Su
- Department of Ultrasonography, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | | | | | | | | |
Collapse
|
4
|
Sibomana O. FDA approves Datroway: a novel therapy for HR-positive, HER2-negative metastatic breast cancer. Ann Med Surg (Lond) 2025; 87:2521-2522. [PMID: 40212175 PMCID: PMC11981328 DOI: 10.1097/ms9.0000000000003122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/21/2025] [Indexed: 04/13/2025] Open
Abstract
The Food and Drug Administration (FDA) has approved datopotamab deruxtecan (Datroway) for the treatment of unresectable or metastatic HR-positive, HER2-negative breast cancer. This antibody-drug conjugate comprises a monoclonal antibody (mAb) that targets TROP2, a cytotoxic agent DXd, and a linker. The mAb binds to TROP2 on cancer cells, facilitating internalization, after which DXd is released, inducing cell death. In the TROPION-Breast01 trial, datopotamab deruxtecan demonstrated superiority over conventional chemotherapy. It provides a promising alternative for patients who have failed prior endocrine or chemotherapy, offering enhanced efficacy than traditional treatments. However, additional studies are required to thoroughly assess the drug's safety and efficacy, as well as to establish the most current information regarding its optimal dosage and administration guidelines.
Collapse
Affiliation(s)
- Olivier Sibomana
- Department of General Medicine and Surgery, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Department of Research, OLGnova, Kigali, Rwanda
| |
Collapse
|
5
|
Rohan TE, Wang Y, Couch F, Feigelson HS, Greenlee RT, Honda S, Stark A, Chitale D, Zhang C, Xue X, Ginsberg M, Loudig O. Clinicopathologic characteristics of ductal carcinoma in situ and risk of subsequent invasive breast cancer: a multicenter, population-based cohort study. Breast Cancer Res Treat 2025; 210:615-625. [PMID: 39832051 PMCID: PMC11953078 DOI: 10.1007/s10549-024-07599-x] [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: 10/23/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE To study the association between clinicopathologic characteristics of ductal carcinoma in situ (DCIS) and risk of subsequent invasive breast cancer (IBC). METHODS We conducted a case-control study nested in a multicenter, population-based cohort of 8175 women aged ≥ 18 years with DCIS diagnosed between 1987 and 2016 and followed for a median duration of 83 months. Cases (n = 497) were women with a first diagnosis of DCIS who developed a subsequent IBC ≥ 6 months later; controls (2/case; n = 959) were matched to cases on age at and calendar year of DCIS diagnosis. Univariable and multivariable conditional logistic regression models were used to examine the associations between the DCIS characteristics of interest (non-screen detection of DCIS, tumor size, positive margins, grade of DCIS, necrosis, architectural pattern, microcalcification, and estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status) and risk of IBC. RESULTS In the total study population, the associations were largely null. In subgroup analyses, there were strong position associations with punctate necrosis (pre/perimenopausal women), detection by physical exam (postmenopausal women), architectural patterns other than the main types (breast-conserving surgery [BCS]), and DCIS margins (ipsilateral cases), and inverse associations with HER2 positivity (BCS) and microcalcification (mastectomy); however, the associated confidence intervals were mostly very wide. CONCLUSION The results of this study provide limited support for associations of the DCIS clinicopathologic characteristics studied here and risk of IBC.
Collapse
Affiliation(s)
- Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Fergus Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Robert T Greenlee
- Center for Clinical Epidemiology and Population Health, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Stacey Honda
- Center for Integrated Healthcare, Kaiser Permanente, Hawaii Permanente Medical Group, Honolulu, HI, USA
| | - Azadeh Stark
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, MI, USA
- Breast Oncology Program and Department of Pathology, Henry Ford Health System, Detroit, MI, USA
| | - Dhananjay Chitale
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, MI, USA
- Breast Oncology Program and Department of Pathology, Henry Ford Health System, Detroit, MI, USA
| | - Chenxin Zhang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mindy Ginsberg
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Olivier Loudig
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, Nutley, NJ, USA
| |
Collapse
|
6
|
Palma M. Advancing Breast Cancer Treatment: The Role of Immunotherapy and Cancer Vaccines in Overcoming Therapeutic Challenges. Vaccines (Basel) 2025; 13:344. [PMID: 40333213 PMCID: PMC12030785 DOI: 10.3390/vaccines13040344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body's immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody-drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines-including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines-aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
| |
Collapse
|
7
|
Ali Q, Niaz R, Soomro R. Unraveling Ductal Carcinoma In Situ in an Unscreened Population: Understanding Its Natural History Through Advanced Presentation and Management. Cureus 2025; 17:e80950. [PMID: 40255704 PMCID: PMC12009591 DOI: 10.7759/cureus.80950] [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] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a non-invasive breast cancer with debated management strategies, particularly in unscreened populations where delayed detection often leads to advanced presentations. Understanding DCIS in this context is crucial for improving risk stratification, treatment, and outcomes. OBJECTIVE This study aims to explore the clinicopathologic features, progression, and outcomes of DCIS in an unscreened population, comparing findings with national and international studies. METHODS We conducted a retrospective analysis of 172 patients diagnosed with isolated DCIS at Liaquat National Hospital and Medical College, Karachi, Pakistan, from January 2019 to December 2023. Data collected included demographics, presenting symptoms, imaging findings, biopsy methods, histopathologic features, and treatment details. Statistical analysis was performed using SPSS Statistics for Windows, Version 25 (Released 2017; SPSS Inc., Chicago, United States), with p ≤ 0.05 considered significant. RESULTS Of 4690 breast cancer cases, 3.6% were isolated DCIS. The median age was 51 years, with 66% postmenopausal. The most common symptom was a palpable lump (68%), with only 3.5% detected via screening. High-grade DCIS was prevalent (41.7%), with comedo necrosis in 23.7%. Tumor size exceeded 5 cm in 25.8% of cases. Breast-conserving surgery (BCS) was performed in 39.9% of patients, with a 15.4% re-surgery rate. Mastectomy and sentinel lymph node biopsy were required in 56.6% of cases. The upgrade rate to invasive carcinoma was 39.9%, higher than global averages. Estrogen receptor positivity was noted in 70.9% of patients. CONCLUSION DCIS in unscreened populations presents more aggressively, with larger, higher-grade tumors and a significant risk of progression to invasive disease. The findings emphasize the need for targeted screening and tailored management strategies to improve outcomes. Future research should focus on optimizing diagnostic and therapeutic approaches in such high-risk groups.
Collapse
Affiliation(s)
- Quratulain Ali
- Department of Breast Surgery, Liaquat National Hospital and Medical College, Karachi, PAK
| | - Rabia Niaz
- Department of Breast Surgery, Liaquat National Hospital and Medical College, Karachi, PAK
| | - Rufina Soomro
- Department of Breast Surgery, Liaquat National Hospital and Medical College, Karachi, PAK
| |
Collapse
|
8
|
Li W, Zhao Y, Fei X, Wu Y, Zhan W, Zhou W, Xia S, Song Y, Zhou J. Image Features and Diagnostic Value of Contrast-Enhanced Ultrasound for Ductal Carcinoma In Situ of the Breast: Preliminary Findings. ULTRASONIC IMAGING 2025; 47:59-67. [PMID: 39506270 DOI: 10.1177/01617346241292032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
To explore the image features and the diagnostic value of contrast-enhanced ultrasound (CEUS) for ductal carcinoma in situ (DCIS) of the breast. A total of 96 female patients with a solitary and histologically proven DCIS were analyzed retrospectively, and 100 female cases of invasive ductal carcinoma (IDC) lesions were used as the control group. The Breast Imaging Reporting and Data System (BI-RADS) category of breast lesions was assessed according to conventional ultrasound features. The DCIS lesions were classified into mass type and non-mass type. The CEUS characteristics of these breast lesions were retrospectively analyzed qualitatively and quantitatively. The final gold standard was biopsy or surgery with histo-pathological examination. Comparing the ultrasound images of DCIS with that of IDC, there were significant differences in echo pattern, calcification morphology, and calcification distribution (p < .05 for all). There was a significant difference between DCIS and IDC in enhancement intensity, perfusion defects, peripheral high enhancement, intratumoral vessels, and arrival time (AT) (p < .05 for all). In the logistic multivariate regression analysis, two indicators linked with DCIS were recognized: perfusion defects (p = .002) and peripheral high enhancement (p < .001). In forecasting DCIS, the logistic regression equation resulted in an AUC of 0.689, a specificity of 0.720, and a sensitivity of 0.563. CEUS showed differences in enhancement characteristics between DCIS and IDC, with perfusion defects and peripheral high enhancement being associated with DCIS.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Ultrasound, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yingyan Zhao
- Department of Ultrasound, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaochun Fei
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ying Wu
- Department of Breast Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shujun Xia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanyan Song
- Department of Biostatistics, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianqiao Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- College of Health Science and Technology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
10
|
Schandiz H, Farkas L, Park D, Liu Y, Andersen SN, Sauer T, Geisler J. High Ki67 expression, HER2 overexpression, and low progesterone receptor levels in high-grade DCIS: significant associations with clinical practice implications. Front Oncol 2025; 15:1467664. [PMID: 39959664 PMCID: PMC11826238 DOI: 10.3389/fonc.2025.1467664] [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: 07/20/2024] [Accepted: 01/02/2025] [Indexed: 02/18/2025] Open
Abstract
Simple summary We investigated the role of Ki67, a ubiquitous marker in cancer, within the context of ductal carcinoma in situ (DCIS), a precursor of invasive breast cancer. Through rigorous analysis of histopathological and immunopathological samples from a substantial cohort, this study revealed robust correlations between heightened Ki67 expression, diminished progesterone (PR) levels, and HER2 overexpression, indicative of aggressive DCIS phenotypes. These findings offer novel insights into the surrogate immunomolecular subtyping landscape of DCIS, potentially refining risk stratification and therapeutic approaches. This elucidation underscores the translational significance of Ki67 as a prognostic and predictive biomarker in DCIS, with implications for personalized treatment paradigms and patient outcomes. Background The Ki67 proliferation index is widely used in various tumors, including invasive breast carcinoma (IBC). However, its prognostic utility is often constrained by technical complexity. Its diagnostic and clinical significance in ductal carcinoma in situ (DCIS) remains uncertain. We studied Ki67 immunohistochemistry interobserver diagnostic agreement at different cutoff values in high-grade DCIS. Additionally, we investigated the associations between Ki67 expression, PR levels, and human epidermal growth factor receptor 2 (HER2) in high-grade DCIS among various subtypes (Luminal (Lum) A, LumB HER2-, LumB HER2+, HER2-enriched, and triple-negative)). Methods Using histopathological specimens from 484 patients diagnosed with DCIS between 1996 and 2018, we implemented the 2013 St. Gallen recommendations for surrogate immunomolecular subtyping of IBC. Subtypes were classified, and the Ki67 interobserver diagnostic agreement between Counting Pathologist 1 (CP1) and CP2 was calculated using Cohen's kappa coefficient at various cutoff values. Results The Cohen's kappa coefficient for interobserver agreement between CP1 and CP2 was κ = 0.586, indicating moderate agreement. Ki67 levels varied significantly among subtypes (p < 0.0001), with a median Ki67% being higher in cases with invasive components (p = 0.0351). Low PR combined with high Ki67% was significantly associated with HER2 overexpression (p = 0.0107). Conclusions Interobserver agreement for the Ki67 count was moderate. Ki67 expression showed considerable variability in high-grade DCIS. Low PR levels combined with high Ki67 expression were linked to HER2 overexpression, showing possible clinical implications for identifying high-risk DCIS.
Collapse
Affiliation(s)
- Hossein Schandiz
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lorant Farkas
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Daehoon Park
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Yan Liu
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), AHUS, Lørenskog, Norway
| | - Solveig N. Andersen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Torill Sauer
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Mitsa G, Florianova L, Lafleur J, Aguilar-Mahecha A, Zahedi RP, del Rincon SV, Basik M, Borchers CH, Batist G. Clinical Proteomics Reveals Vulnerabilities in Noninvasive Breast Ductal Carcinoma and Drives Personalized Treatment Strategies. CANCER RESEARCH COMMUNICATIONS 2025; 5:138-149. [PMID: 39723668 PMCID: PMC11755405 DOI: 10.1158/2767-9764.crc-24-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/04/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
SIGNIFICANCE This study provides real-world evidence for DCIS, a disease for which currently no molecular tools or biomarkers exist, and gives an unbiased, comprehensive, and deep proteomic profile, identifying >380 actionable targets.
Collapse
Affiliation(s)
- Georgia Mitsa
- Division of Experimental Medicine, McGill University, Montréal, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital and McGill University, Montréal, Canada
| | - Livia Florianova
- Department of Pathology, McGill University, Montréal, Canada
- Division of Pathology, Jewish General Hospital, Montréal, Canada
| | - Josiane Lafleur
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Canada
| | - Adriana Aguilar-Mahecha
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Canada
| | - Rene P. Zahedi
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Canada
- CancerCare Manitoba Research Institute, Winnipeg, Canada
| | - Sonia V. del Rincon
- Division of Experimental Medicine, McGill University, Montréal, Canada
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Canada
| | - Mark Basik
- Division of Experimental Medicine, McGill University, Montréal, Canada
- Department of Medicine, McGill University, Montréal, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Canada
- Department of Surgery, McGill University, Montréal, Canada
| | - Christoph H. Borchers
- Division of Experimental Medicine, McGill University, Montréal, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital and McGill University, Montréal, Canada
- Department of Pathology, McGill University, Montréal, Canada
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Canada
| | - Gerald Batist
- Division of Experimental Medicine, McGill University, Montréal, Canada
- Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Canada
- Department of Surgery, McGill University, Montréal, Canada
- Exactis Innovation, Montréal, Canada
| |
Collapse
|
12
|
Ayoub Y, Cheung SM, Maglan B, Senn N, Chan KS, He J. Differentiation of histological calcification classifications in breast cancer using ultrashort echo time and chemical shift-encoded imaging MRI. Front Oncol 2024; 14:1475090. [PMID: 39741975 PMCID: PMC11685069 DOI: 10.3389/fonc.2024.1475090] [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: 08/06/2024] [Accepted: 11/25/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Ductal carcinoma in situ (DCIS) accounts for 25% of newly diagnosed breast cancer cases with only 14%-53% developing into invasive ductal carcinoma (IDC), but currently overtreated due to inadequate accuracy of mammography. Subtypes of calcification, discernible from histology, has been suggested to have prognostic value in DCIS, while the lipid composition of saturated and unsaturated fatty acids may be altered in de novo synthesis with potential sensitivity to the difference between DCIS and IDC. We therefore set out to examine calcification using ultra short echo time (UTE) MRI and lipid composition using chemical shift-encoded imaging (CSEI), as markers for histological calcification classification, in the initial ex vivo step towards in vivo application. Methods Twenty female patients, with mean age (range) of 57 (35-78) years, participated in the study. Intra- and peri-tumoural degree of calcification and peri-tumoural lipid composition were acquired on MRI using UTE and CSEI, respectively. Ex vivo imaging was conducted on the freshly excised breast tumour specimens immediately after surgery. Histopathological analysis was conducted to determine the calcification status, Nottingham Prognostic Index (NPI), and proliferative activity marker Ki-67. Results Intra-tumoural degree of calcification in malignant classification (1.05 ± 0.13) was significantly higher (p = 0.012) against no calcification classification (0.84 ± 0.09). Peri-tumoural degree of calcification in malignant classification (1.64 ± 0.10) was significantly higher (p = 0.033) against no calcification classification (1.41 ± 0.18). Peri-tumoural MUFA in malignant classification (0.40 ± 0.01) was significantly higher (p = 0.039) against no calcification classification (0.38 ± 0.02). Ki-67 showed significant negative correlation against peri-tumoural MUFA (p = 0.043, ρ = -0.457), significant positive correlation against SFA (p = 0.008, ρ = 0.577), and significant negative correlation against PUFA (p = 0.002, ρ = -0.653). Conclusion The intra- and peri-tumoural degree of calcification and peri-tumoural MUFA are sensitive to histological calcification classes supporting future investigation into DCIS prognosis.
Collapse
Affiliation(s)
- Yazan Ayoub
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Sai Man Cheung
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Boddor Maglan
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Nicholas Senn
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Kwok-Shing Chan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Jiabao He
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| |
Collapse
|
13
|
Galappaththi SPL, Smith KR, Alsatari ES, Hunter R, Dyess DL, Turbat-Herrera EA, Dasgupta S. The Genomic and Biologic Landscapes of Breast Cancer and Racial Differences. Int J Mol Sci 2024; 25:13165. [PMID: 39684874 DOI: 10.3390/ijms252313165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer is a significant health challenge worldwide and is the most frequently diagnosed cancer among women globally. This review provides a comprehensive overview of breast cancer biology, genomics, and microbial dysbiosis, focusing on its various subtypes and racial differences. Breast cancer is primarily classified into carcinomas and sarcomas, with carcinomas constituting most cases. Epidemiology and breast cancer risk factors are important for public health intervention. Staging and grading, based on the TNM and Nottingham grading systems, respectively, are crucial to determining the clinical outcome and treatment decisions. Histopathological subtypes include in situ and invasive carcinomas, such as invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC). The review explores molecular subtypes, including Luminal A, Luminal B, Basal-like (Triple Negative), and HER2-enriched, and delves into breast cancer's histological and molecular progression patterns. Recent research findings related to nuclear and mitochondrial genetic alterations, epigenetic reprogramming, and the role of microbiome dysbiosis in breast cancer and racial differences are also reported. The review also provides an update on breast cancer's current diagnostics and treatment modalities.
Collapse
Affiliation(s)
- Sapthala P Loku Galappaththi
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Kelly R Smith
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Enas S Alsatari
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Rachel Hunter
- Department of Surgery, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Donna L Dyess
- Department of Surgery, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Elba A Turbat-Herrera
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Santanu Dasgupta
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
- Department of Biochemistry and Molecular Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
14
|
Debeljak M, Cho S, Downs BM, Considine M, Avin-McKelvey B, Wang Y, Perez PN, Grizzle WE, Hoadley KA, Lynch CF, Hernandez BY, van Diest PJ, Cozen W, Hamilton AS, Hawes D, Gabrielson E, Cimino-Mathews A, Florea LD, Cope L, Umbricht CB. Multimodal genome-wide survey of progressing and non-progressing breast ductal carcinoma in-situ. Breast Cancer Res 2024; 26:178. [PMID: 39633428 PMCID: PMC11616160 DOI: 10.1186/s13058-024-01927-1] [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: 08/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Ductal carcinoma in-situ (DCIS) is a pre-invasive form of invasive breast cancer (IBC). Due to improved breast cancer screening, it now accounts for ~ 25% of all breast cancers. While the treatment success rates are over 90%, this comes at the cost of considerable morbidity, considering that the majority of DCIS never become invasive and our understanding of the molecular changes occurring in DCIS that predispose to invasive disease is limited. The aim of this study is to characterize molecular changes that occur in DCIS, with the goal of improving DCIS risk stratification. METHODS We identified and obtained a total of 197 breast tissue samples from 5 institutions (93 DCIS progressors, 93 DCIS non-progressors, and 11 adjacent normal breast tissues) that had at least 10-year follow-up. We isolated DNA and RNA from archival tissue blocks and characterized genome-wide mRNA expression, DNA methylation, DNA copy number variation, and RNA splicing variation. RESULTS We obtained all four genomic data sets in 122 of the 197 samples. Our intrinsic expression subtype-stratified analyses identified multiple molecular differences both between DCIS subtypes and between DCIS and IBC. While there was heterogeneity in molecular signatures and outcomes within intrinsic subtypes, several gene sets that differed significantly between progressing and non-progressing DCIS were identified by Gene Set Enrichment Analysis. CONCLUSION DCIS is a molecularly highly heterogenous disease with variable outcomes, and the molecular events determining DCIS disease progression remain poorly defined. Our genome-wide multi-omic survey documents DCIS-associated alterations and reveals molecular heterogeneity within the intrinsic DCIS subtypes. Further studies investigating intrinsic subtype-stratified characteristics and molecular signatures are needed to determine if these may be exploitable for risk assessment and mitigation of DCIS progression. The highly significant associations of specific gene sets with IBC progression revealed by our Gene Set Enrichment Analysis may lend themselves to the development of a prognostic molecular score, to be validated on independent DCIS cohorts.
Collapse
Affiliation(s)
- Marija Debeljak
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Soonweng Cho
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bradley M Downs
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Considine
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Yongchun Wang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phillip N Perez
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William E Grizzle
- Department of Pathology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles F Lynch
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Brenda Y Hernandez
- Population Sciences in the Pacific-Program, University of Hawaii Cancer Research Center, Honolulu, HI, USA
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wendy Cozen
- Department of Medicine, School of Medicine, Susan and Henry Samueli College of Health Sciences, University of California at Irvine, Irvine, CA, USA
| | - Ann S Hamilton
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Debra Hawes
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ashley Cimino-Mathews
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liliana D Florea
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie Cope
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher B Umbricht
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Johns Hopkins University School of Medicine, Ross Building, Room 743, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
15
|
Li W, Zhang H, Xu J, Maimaitijiang A, Su Z, Fan Z, Li J. The Biological Roles of ZKSCAN3 (ZNF306) in the Hallmarks of Cancer: From Mechanisms to Therapeutics. Int J Mol Sci 2024; 25:11532. [PMID: 39519085 PMCID: PMC11546961 DOI: 10.3390/ijms252111532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
ZKSCAN3 (also known as ZNF306) plays a pivotal role in the regulation of various cellular processes that are fundamental to the development of cancer. It has been widely acknowledged as a key contributor to cancer progression, with its overexpression consistently reported in a broad spectrum of malignancies. Importantly, clinical studies have demonstrated a significant association between elevated ZKSCAN3 levels and adverse prognosis, as well as resistance to therapeutic drugs. Specifically, ZKSCAN3 promotes tumor progression by enhancing multiple hallmark features of cancer and promoting the acquisition of cancer-specific phenotypes. These effects manifest as increased tumor cell proliferation, invasion, and metastasis, accompanied by inhibiting tumor cell apoptosis and modulating autophagy. Consequently, ZKSCAN3 emerges as a promising prognostic marker, and targeting its inhibition represents a potential strategy for anti-tumor therapy. In this review, we provide an updated perspective on the role of ZKSCAN3 in governing tumor characteristics and the underlying molecular mechanisms. Furthermore, we underscore the clinical relevance of ZKSCAN3 and its potential implications for tumor prognosis and therapeutic strategies.
Collapse
Affiliation(s)
- Wenfang Li
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Han Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jianxiong Xu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Ayitila Maimaitijiang
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Zhengding Su
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Zhongxiong Fan
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
16
|
John E, Lesluyes T, Baker TM, Tarabichi M, Gillenwater A, Wang JR, Van Loo P, Zhao X. Reconstructing oral cavity tumor evolution through brush biopsy. Sci Rep 2024; 14:22591. [PMID: 39343812 PMCID: PMC11439926 DOI: 10.1038/s41598-024-72946-3] [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: 02/11/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
Oral potentially malignant disorders (OPMDs) with genomic alterations have a heightened risk of evolving into oral squamous cell carcinoma (OSCC). Currently, genomic data are typically obtained through invasive tissue biopsy. However, brush biopsy is a non-invasive method that has been utilized for identifying dysplastic cells in OPMD but its effectiveness in reflecting the genomic landscape of OPMDs remains uncertain. This pilot study investigates the potential of brush biopsy samples in accurately reconstructing the genomic profile and tumor evolution in a patient with both OPMD and OSCC. We analyzed single nucleotide variants (SNVs), copy number aberrations (CNAs), and subclonal architectures in paired tissue and brush biopsy samples. The results showed that brush biopsy effectively captured 90% of SNVs and had similar CNA profiles as those seen in its paired tissue biopsies in all lesions. It was specific, as normal buccal mucosa did not share these genomic alterations. Interestingly, brush biopsy revealed shared SNVs and CNAs between the distinct OPMD and OSCC lesions from the same patient, indicating a common ancestral origin. Subclonal reconstruction confirmed this shared ancestry, followed by divergent evolution of the lesions. These findings highlight the potential of brush biopsies in accurately representing the genomic profile of OPL and OSCC, proving insight into reconstructing tumor evolution.
Collapse
Affiliation(s)
- Evit John
- Department of Genetics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, FCT 10.6008, 77030, TX, Houston, USA
| | | | - Toby M Baker
- Department of Genetics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, FCT 10.6008, 77030, TX, Houston, USA
- The Francis Crick Institute, London, UK
| | - Maxime Tarabichi
- Institute for Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Ann Gillenwater
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, TX, Houston, USA
| | - Jennifer R Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, TX, Houston, USA
| | - Peter Van Loo
- Department of Genetics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, FCT 10.6008, 77030, TX, Houston, USA
- The Francis Crick Institute, London, UK
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, TX, Houston, USA
| | - Xiao Zhao
- Department of Genetics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, FCT 10.6008, 77030, TX, Houston, USA.
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, TX, Houston, USA.
| |
Collapse
|
17
|
Nicosia L, Mariano L, Latronico A, Bozzini AC, Bellerba F, Gaeta A, Pesapane F, Mazzarol G, Fusco N, Corso G, Sangalli C, Gialain C, Lazzeroni M, Raimondi S, Cassano E. Exploring non-surgical alternatives for low to intermediate-grade in situ ductal carcinoma of the breast using vacuum-assisted excision: the VACIS protocol. Front Med (Lausanne) 2024; 11:1467738. [PMID: 39380737 PMCID: PMC11458405 DOI: 10.3389/fmed.2024.1467738] [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: 07/20/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
Background Surgery is still the standard treatment for breast lesions such as in situ ductal carcinoma (DCIS); however, its survival benefit is minimal, particularly for low-grade DCIS. Surgical complications and related depression status can adversely affect patients' quality of life. Approximately 25% of breast cancer (BC) cases are in situ forms, with DCIS making up 90% of these. Low and intermediate-grade DCIS often grow slowly and do not always progress clinically significant diseases. Identifying non-invasive lesions could help prevent overtreatment. In this context, new diagnostic tools like vacuum-assisted excision (VAE) could enhance the management of these conditions. Methods The prospective VACIS study explores the role of VAE in ensuring the absence of pathology at subsequent surgery and reducing the diagnostic underestimation of breast biopsies for microcalcifications. Patients with suspicious breast microcalcifications up to 15 mm, who are candidates for stereotactic biopsy, will be enrolled and randomised into two groups. The control group will complete the biopsy with typical sampling, aiming to collect some microcalcifications from the target, while the experimental group will focus on the complete removal of the biopsy target (confirmed by mammography on the biopsy table), followed by a second sequence of cleaning samples. Radiograms will confirm lesion removal. Pathologic outcomes at surgery will be compared between the groups, and the percentage of underestimation will be assessed. The sample size is calculated to be 70 patients per group, using statistical tests and multivariate logistic models to detect a significant difference in the absence of pathology. Data collected will include patient age, lesion characteristics, and details of the biopsy, pathology and surgery. Discussion Current surgical treatments for low-and sometimes intermediate-grade DCIS offer limited survival benefits and may hurt patients' quality of life due to surgery-related complications and associated depression. These lesions often grow slowly and might not become clinically significant, suggesting a need to avoid overtreatment. Improved diagnostics procedures, such as VAE, could help distinguish non-invasive from potentially invasive lesions, reduce biopsy underestimation, enable personalised management and optimise treatment strategies. This study hypothesises that VAE could be a viable alternative to surgery, capable of removing pathology during the biopsy procedure. Clinical trial registration Clinicaltrials.gov, identifier NCT05932758.
Collapse
Affiliation(s)
- Luca Nicosia
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Breast Imaging Division, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Luciano Mariano
- Breast Imaging Division, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Antuono Latronico
- Breast Imaging Division, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Anna Carla Bozzini
- Breast Imaging Division, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Federica Bellerba
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Aurora Gaeta
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Department of Statistics and Quantitative Methods University of Milano-Bicocca, Milan, Italy
| | - Filippo Pesapane
- Breast Imaging Division, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giovanni Mazzarol
- Division of Pathology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giovanni Corso
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Breast Surgery, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Claudia Sangalli
- Clinical Trial Office, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Cristian Gialain
- Clinical Trial Office, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Matteo Lazzeroni
- Division of Cancer Prevention and Genetics, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Enrico Cassano
- Breast Imaging Division, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| |
Collapse
|
18
|
Haidar M, Rizkallah J, El Sardouk O, El Ghawi N, Omran N, Hammoud Z, Saliba N, Tfayli A, Moukadem H, Berjawi G, Nassar L, Marafi F, Choudhary P, Dadgar H, Sadeq A, Abi-Ghanem AS. Radiotracer Innovations in Breast Cancer Imaging: A Review of Recent Progress. Diagnostics (Basel) 2024; 14:1943. [PMID: 39272726 PMCID: PMC11394464 DOI: 10.3390/diagnostics14171943] [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: 07/31/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
This review focuses on the pivotal role of radiotracers in breast cancer imaging, emphasizing their importance in accurate detection, staging, and treatment monitoring. Radiotracers, labeled with radioactive isotopes, are integral to various nuclear imaging techniques, including positron emission tomography (PET) and positron emission mammography (PEM). The most widely used radiotracer in breast cancer imaging is 18F-fluorodeoxyglucose (18F-FDG), which highlights areas of increased glucose metabolism, a hallmark of many cancer cells. This allows for the identification of primary tumors and metastatic sites and the assessment of tumor response to therapy. In addition to 18F-FDG, this review will explore newer radiotracers targeting specific receptors, such as estrogen receptors or HER2, which offer more personalized imaging options. These tracers provide valuable insights into the molecular characteristics of tumors, aiding in tailored treatment strategies. By integrating radiotracers into breast cancer management, clinicians can enhance early disease detection, monitor therapeutic efficacy, and guide interventions, ultimately improving patient outcomes. Ongoing research aimed at developing more specific and sensitive tracers will also be highlighted, underscoring their potential to advance precision medicine in breast cancer care.
Collapse
Affiliation(s)
- Mohamad Haidar
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Joe Rizkallah
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Omar El Sardouk
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nour El Ghawi
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nadine Omran
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Zeinab Hammoud
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Nina Saliba
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Arafat Tfayli
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hiba Moukadem
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Ghina Berjawi
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Lara Nassar
- Department of Diagnostic Radiology, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Fahad Marafi
- Jaber Al-Ahmad Centre for Molecular Imaging, Kuwait City 70031, Kuwait
| | - Partha Choudhary
- Department of Nuclear Medicine, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi 110085, India
| | - Habibollah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad 9198613636, Iran
| | - Alyaa Sadeq
- Jaber Al-Ahmad Centre for Molecular Imaging, Kuwait City 70031, Kuwait
| | - Alain S Abi-Ghanem
- Department of Diagnostic Radiology, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| |
Collapse
|
19
|
Rajdeo P, Aronow B, Surya Prasath VB. Deep learning-based multimodal spatial transcriptomics analysis for cancer. Adv Cancer Res 2024; 163:1-38. [PMID: 39271260 PMCID: PMC11431148 DOI: 10.1016/bs.acr.2024.08.001] [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] [Indexed: 09/15/2024]
Abstract
The advent of deep learning (DL) and multimodal spatial transcriptomics (ST) has revolutionized cancer research, offering unprecedented insights into tumor biology. This book chapter explores the integration of DL with ST to advance cancer diagnostics, treatment planning, and precision medicine. DL, a subset of artificial intelligence, employs neural networks to model complex patterns in vast datasets, significantly enhancing diagnostic and treatment applications. In oncology, convolutional neural networks excel in image classification, segmentation, and tumor volume analysis, essential for identifying tumors and optimizing radiotherapy. The chapter also delves into multimodal data analysis, which integrates genomic, proteomic, imaging, and clinical data to offer a holistic understanding of cancer biology. Leveraging diverse data sources, researchers can uncover intricate details of tumor heterogeneity, microenvironment interactions, and treatment responses. Examples include integrating MRI data with genomic profiles for accurate glioma grading and combining proteomic and clinical data to uncover drug resistance mechanisms. DL's integration with multimodal data enables comprehensive and actionable insights for cancer diagnosis and treatment. The synergy between DL models and multimodal data analysis enhances diagnostic accuracy, personalized treatment planning, and prognostic modeling. Notable applications include ST, which maps gene expression patterns within tissue contexts, providing critical insights into tumor heterogeneity and potential therapeutic targets. In summary, the integration of DL and multimodal ST represents a paradigm shift towards more precise and personalized oncology. This chapter elucidates the methodologies and applications of these advanced technologies, highlighting their transformative potential in cancer research and clinical practice.
Collapse
Affiliation(s)
- Pankaj Rajdeo
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - V B Surya Prasath
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States; Department of Biomedical Informatics, College of Medicine, University of Cincinnati, Cincinnati, OH, United States; Department of Computer Science, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
20
|
Zhang M, Zheng Z, Wang S, Liu R, Zhang M, Guo Z, Wang H, Tan W. The role of circRNAs and miRNAs in drug resistance and targeted therapy responses in breast cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:30. [PMID: 39267922 PMCID: PMC11391347 DOI: 10.20517/cdr.2024.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/20/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs comprising 19-24 nucleotides that indirectly control gene expression. In contrast to other non-coding RNAs (ncRNAs), circular RNAs (circRNAs) are defined by their covalently closed loops, forming covalent bonds between the 3' and 5' ends. circRNAs regulate gene expression by interacting with miRNAs at transcriptional or post-transcriptional levels. Accordingly, circRNAs and miRNAs control many biological events related to cancer, including cell proliferation, metabolism, cell cycle, and apoptosis. Both circRNAs and miRNAs are involved in the pathogenesis of diseases, such as breast cancer. This review focuses on the latest discoveries on dysregulated circRNAs and miRNAs related to breast cancer, highlighting their potential as biomarkers for clinical diagnosis, prognosis, and chemotherapy response.
Collapse
Affiliation(s)
- Meilan Zhang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, Guangdong, China
| | - Zhaokuan Zheng
- Department of Orthopedics, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of HuaduDistrict), Guangzhou 510810, Guangdong, China
| | - Shouliang Wang
- Department of Breast Surgery, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China
| | - Ruihan Liu
- Department of Breast Surgery, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China
| | - Mengli Zhang
- Department of Breast Surgery, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China
| | - Zhiyun Guo
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, Guangdong, China
| | - Hao Wang
- The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, Guangdong, China
| | - Weige Tan
- Department of Breast Surgery, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China
| |
Collapse
|
21
|
Xu AY, Hanna M. Detection of Contralateral Malignancies on Breast MRI. Cureus 2024; 16:e66510. [PMID: 39246927 PMCID: PMC11380806 DOI: 10.7759/cureus.66510] [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: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024] Open
Abstract
INTRODUCTION Women with unilateral breast cancer are at increased risk for having simultaneous cancer of the contralateral breast. Overall, earlier detection of contralateral breast cancer prevents the burden of additional surgery or chemotherapy rounds and is associated with higher overall survival. However, MRI screening for the contralateral breast is seldom done following an initial unilateral breast cancer diagnosis. The purpose of this study is to retrospectively evaluate patients with known, biopsy-proven malignancy who went on to obtain a breast MRI and were later found to have cancer of the contralateral breast. Methods: This was a retrospective study that reviewed the charts of women aged over 18 years who were determined to have synchronous bilateral breast cancer from January 2017 to January 2022 at the University of Florida, Gainesville, FL. The study extracted data from this institution's cancer registry database, which provided information on patients with breast cancer diagnoses. The study conducted a review of mammography (MAM) and MRI imaging reports to ascertain the presence or absence of contralateral breast cancer identified by each respective imaging modality. Surgical pathology reports from the biopsy of the contralateral breast were reviewed to obtain information on the histological type of cancer and TNM (tumor, node, metastasis) staging. RESULTS Of the 17 cases in which MAM missed contralateral cancer, follow-up MRI detected contralateral malignancy in 12 cases (70.59%) and subsequently changed management, resulting in additional imaging, biopsy, and eventual diagnosis and treatment of contralateral breast cancer. Examining the number of contralateral breast cancers detected by patients who had undergone MAM followed by MRI and those who had only undergone MAM, the study found that the detection rate of contralateral breast cancer from MAM was 45.45% (15/33). The tumor stages of the missed cancers were all T1 or Tis stage with one T1mi, and there was no nodal involvement. Conclusion: In addition to its utility in staging breast cancers, MRI also has the superior ability to detect otherwise undetected contralateral breast malignancy. This retrospective study found that MRI imaging led to a considerable increase in the detection of contralateral cancer. The study found that these undetected contralateral breast cancers by MAM were often of lower staging with no nodal involvement, highlighting the opportunity for MRI to assist in early cancer detection while the patient's prognosis is still good. Its high cost should be balanced with staging and occult malignancy detection utility in future practice.
Collapse
Affiliation(s)
- Alan Y Xu
- Radiology, University of Florida College of Medicine, Gainesville, USA
| | - Mariam Hanna
- Radiology, University of Florida College of Medicine, Gainesville, USA
| |
Collapse
|
22
|
Nava-Tapia DA, Román-Justo NY, Cuenca-Rojo A, Guerrero-Rivera LG, Patrón-Guerrero A, Poblete-Cruz RI, Zacapala-Gómez AE, Sotelo-Leyva C, Navarro-Tito N, Mendoza-Catalán MA. Exploring the potential of tocopherols: mechanisms of action and perspectives in the prevention and treatment of breast cancer. Med Oncol 2024; 41:208. [PMID: 39060448 DOI: 10.1007/s12032-024-02454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Currently, breast cancer is the most common cause of mortality caused by neoplasia in women worldwide. The unmet challenges of conventional cancer therapy are chemoresistance and lack of selectivity, which can lead to serious side effects in patients; therefore, new treatments based on natural compounds that serve as adjuvants in breast cancer therapy are urgently needed. Tocopherols are naturally occurring antioxidant compounds that have shown antitumor activity against several types of cancer, including breast cancer. This review summarizes the antitumoral activity of tocopherols, such as the antiproliferative, apoptotic, anti-invasive, and antioxidant effects of tocopherols, through different molecular mechanisms. According to the studies described, α-T, δ-T and γ-T are the most studied in breast tumor cells; however, α-T and γ-T show a more critical antitumor activity and significant potential as a complements to chemotherapeutic drugs against breast cancer, enhancing toxicity against tumor cells and preventing cytotoxicity in nontumor cells. However, the possible relationship between tocopherol intake, related to concentration, and the promotion of cancer in particular cases should not be ruled out, so additional studies are required to determine the correct dose to obtain the desired antitumor effect. Moreover, nanomicelles of D-α-tocopherol have promising potential as pharmaceutical excipients for drug delivery to improve the cytotoxicity and selectivity of first-line chemotherapeutics against breast cancer.
Collapse
Affiliation(s)
- Dania A Nava-Tapia
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Norely Y Román-Justo
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Antonio Cuenca-Rojo
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Lizeth G Guerrero-Rivera
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Annet Patrón-Guerrero
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Ruth I Poblete-Cruz
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Ana E Zacapala-Gómez
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - César Sotelo-Leyva
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Napoleón Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico.
| | - Miguel A Mendoza-Catalán
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
23
|
Kocaman N, Onat E, Balta H, Üçer Ö. Are Meteorin-Like Peptide and Asprosin Important in the Diagnosis of Breast Tumors? Cureus 2024; 16:e62979. [PMID: 39044875 PMCID: PMC11265791 DOI: 10.7759/cureus.62979] [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] [Accepted: 06/22/2024] [Indexed: 07/25/2024] Open
Abstract
INTRODUCTION Breast cancer (BC) is one of the most common and leading causes of death in women. Therefore, early and accurate diagnosis is vital. In this study, meteorin-like (METRNL) peptide and asprosin levels were examined in breast tissue in invasive ductal carcinoma (IDC) of the breast, and the roles of these molecules in the diagnosis of BC were investigated. METHODS In this retrospective study, tissues from patients with BC in the Pathology Department Laboratory of Fırat University Faculty of Medicine, Elazığ, Turkey, were used. Samples from 30 patients were used. The control group consisted of healthy breast tissues from the same patients. The pathology group consisted of breast tissues with IDC from the same patients. Breast tissue samples from both groups were evaluated immunohistochemically for METRNL and asprosin. RESULTS Statistically significant differences were observed between both groups in terms of METRNL and asprosin. It was observed that METRNL and asprosin immunoreactivities were higher in breast tissues with IDC than in healthy breast tissues (p<0.001). CONCLUSION When the study results were evaluated, it was seen that there was a significant relationship between healthy breast tissues and the ones with IDC in terms of METRNL and asprosin. It is thought that both METRNL and asprosin may be really important in the future for the early diagnosis and treatment of BC.
Collapse
Affiliation(s)
- Nevin Kocaman
- Department of Histology and Embryology, Firat University Faculty of Medicine, Elazığ, TUR
| | - Elif Onat
- Department of Medical Pharmacology, Adıyaman University Faculty of Medicine, Adıyaman, TUR
| | - Hilal Balta
- Department of Pathology, Fırat University Faculty of Medicine, Elazığ, TUR
| | - Özlem Üçer
- Department of Pathology, Fırat University Faculty of Medicine, Elazığ, TUR
| |
Collapse
|