1
|
Chen K, Wang G, Hu JC, Zhou YY, Ma HT. Clinical significance of Ki-67 in patients with lung adenocarcinoma in situ complicated by type 2 diabetes. World J Diabetes 2025; 16:98423. [DOI: 10.4239/wjd.v16.i2.98423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND The increasing number of type 2 diabetes mellitus (T2DM) patients leads to higher rates of morbidity and mortality related to lung cancer.
AIM To investigate the utility of the proliferating cell nuclear antigen Ki-67 in patients with lung adenocarcinoma in situ (AIS) complicated by T2DM.
METHODS One hundred patients with AIS and T2DM (group A), 100 patients with AIS alone (group B), and 60 patients with benign lung lesions (group C) admitted to the Department of Thoracic Surgery and Endocrinology of the First Affiliated Hospital of Soochow University from November 2021 to December 2022 were enrolled. Ki-67 expression was compared among the groups.
RESULTS Group A had significantly higher levels of fasting plasma glucose (FPG), total cholesterol (TC), total triglyceride, low-density lipoprotein cholesterol, glycosylated hemoglobin (HbA1c), and insulin than groups B and C (P < 0.01). Meanwhile, group B had higher insulin levels than group C (P < 0.01). Group A exhibited a significantly higher average Ki-67 positivity rate than group B (P < 0.01). The Ki-67 positivity rate in group A was 86.87%, while the positivity rate in group B was 77%. Ki-67 was positively correlated with FPG (P < 0.01) and HbA1c levels (P < 0.01). Ki-67, FBG, insulin, HbA1c, high-density lipoprotein cholesterol and TC were independent factors for patients with AIS complicated by T2DM.
CONCLUSION Ki-67 expression was higher in patients with AIS complicated by T2DM than in patients with AIS alone. Therefore, detecting the Ki-67 level might assist in the diagnosis of AIS in patients with T2DM.
Collapse
Affiliation(s)
- Ke Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Ge Wang
- Department of Endocrine, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Jing-Cheng Hu
- Department of Endocrine, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Ying-Yi Zhou
- Department of Endocrine, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Hai-Tao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| |
Collapse
|
2
|
Wan W, Zhu K, Ran Z, Zhu X, Wang D. Development of a Nomogram-Integrated Model Incorporating Intra-tumoral and Peri-tumoral Ultrasound Radiomics Alongside Clinical Parameters for the Prediction of Histological Grading in Invasive Breast Cancer. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:262-272. [PMID: 39477745 DOI: 10.1016/j.ultrasmedbio.2024.09.025] [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: 07/11/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE To develop a comprehensive nomogram to predict the histological grading of breast cancer and further examine its clinical significance by integrating both intra-tumoral and peri-tumoral ultrasound radiomics features. METHODS In a retrospective study 468 female breast cancer patients were analyzed from 2017 to 2020 at the Second Affiliated Hospital of Harbin Medical University. Patients were grouped into high-grade (n = 215) and low-grade (n = 253) categories based on pathological evaluation. Tumor regions of interest were defined and expanded automatically to peri-tumor regions of interest. Ultrasound radiomics features were extracted independently. To ensure rigor, cases were randomly divided into 80% training and 20% test sets. Optimal features were selected using statistical and machine learning methods. Intra-tumor, peri-tumor, and combined radiomics models were constructed. To determine the best predictors of breast cancer histological grading, we screened the features using single- and multi-factor logistic regression analyses. Finally, a nomogram was developed and evaluated for its predictive value in this context. RESULTS By applying logistic regression, we integrated ultrasound, clinicopathologic, and radiomics features to generate a nomogram. The combined model outperformed others, achieving areas under the curve of 0.934 and 0.812 in training and test sets. Calibration curves also showed high accuracy and reliability. CONCLUSION A nomogram constructed through the integration of combined intra-tumor-peri-tumor ultrasound radiomics features along with clinicopathologic characteristics exhibited remarkable performance in distinguishing the histologic grades of invasive breast cancer.
Collapse
Affiliation(s)
- Wenjia Wan
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Kai Zhu
- Radiology Department, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhicheng Ran
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xinyu Zhu
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Dongmo Wang
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
3
|
Emanuelle Pereira Santos V, Luiz de França Neto P, Eda de Oliveira Isídio B, Henrique Bezerra Fontes P, Andrêssa de Moura I, Isabel Santos Cruz B, Máyra Gois de Sousa M, Luana Dos Santos D, de França São Marcos B, Sousa de Pinho S, Mendonça Alves Bandeira B, Loureiro Leão S, de Almeida Lima T, da Conceição Viana Invenção M, Rosa Sales Leal L, Cristofer Flores Espinoza B, Silva de Macêdo L, do Nascimento Carvalho M, Jéssica Duarte Silva A, Carlos de Freitas A. An overview about biomarkers in breast cancer: Insights into the diagnostic and prognostic significance. Clin Chim Acta 2025; 567:120030. [PMID: 39515632 DOI: 10.1016/j.cca.2024.120030] [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: 04/30/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Breast cancer (BC) is one of the most significant neoplasms globally due to its high incidence and mortality, particularly among females. As a highly heterogeneous pathology, biomarkers are essential for characterizing specific tumors. Currently, several biological processes are well-described in the context of this neoplasm, such as alterations in BRCA1/2, HER, and pathways involving estrogen and progesterone hormone receptors. These studies have enabled the use of these findings as more precise methods for diagnosis, prognosis, and treatment. However, beyond patients who do not exhibit these classic markers, some individuals within the same risk group respond differently to treatment. Therefore, the search for biological markers that can improve diagnosis, aid in stratification, or serve as therapeutic targets is continuous and urgent. Genetic signatures have led to molecular tests currently used in clinical practice, though certain limitations persist. Understanding genetic and epigenetic mechanisms facilitates the identification of potential biomarkers. Biomarker targets must undergo experimental and clinical trials on samples of significant size before reaching clinical utility. In this review, we compile the classical markers and describe the potential use of other markers associated with the biological processes of this neoplasm.
Collapse
Affiliation(s)
- Vanessa Emanuelle Pereira Santos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Pedro Luiz de França Neto
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Beatriz Eda de Oliveira Isídio
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Pedro Henrique Bezerra Fontes
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Bruna Isabel Santos Cruz
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Mylenna Máyra Gois de Sousa
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Daffany Luana Dos Santos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Bianca de França São Marcos
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Samara Sousa de Pinho
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Beatriz Mendonça Alves Bandeira
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Stephanie Loureiro Leão
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Thainá de Almeida Lima
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Lígia Rosa Sales Leal
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Benigno Cristofer Flores Espinoza
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Matheus do Nascimento Carvalho
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Federal University of Pernambuco - Av. Prof. Moraes Rego, 1235. Cidade Universitária Recife, Pernambuco CEP: 50670-901, Brazil.
| |
Collapse
|
4
|
Yıldiz A, Bilici A, Acikgoz O, Hamdard J, Basim P, Cakir T, Cakir A, Olmez OF, Gezen C, Yildiz O. Prognostic implications of response to neoadjuvant chemotherapy in breast cancer subtypes. J Chemother 2025; 37:60-68. [PMID: 38351652 DOI: 10.1080/1120009x.2024.2314830] [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: 06/26/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 01/21/2025]
Abstract
The current study was designed to assess the response to treatment, as well as clinical and survival outcomes, across different breast cancer subtypes in patients who underwent neoadjuvant chemotherapy (NAC). From 2014 to 2019, a total of 139 patients who were histologically confirmed to have breast cancer, underwent NAC, and subsequently received breast and axillary surgery, were retrospectively included in this study. The rates of pathological complete response (pCR) to NAC were significantly higher for HER2-positive and triple-negative subtypes than for luminal A and HER2-negative subtypes (p = 0.013). Multivariate analysis for disease-free survival (DFS) revealed that tumour grade and the presence of pCR were independent prognostic factors. The presence or absence of a pCR with NAC was an independent prognostic indicator in the multivariate analysis for overall survival (OS). Lastly, achieving a pCR was independently predicted by 18F-FDG PET/CT findings, the HER2-positive subtype, and the triple-negative subtype. Despite the inherent methodological limitations, our findings underscore the significance of identifying predictive markers to tailor NAC plans, with the aim of improving survival outcomes.
Collapse
Affiliation(s)
- Anil Yıldiz
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Bilici
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Ozgur Acikgoz
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Jamshid Hamdard
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Pelin Basim
- Department of Breast Surgery, Istanbul Medipol University, Istanbul, Turkey
| | - Tansel Cakir
- Department of Nuclear Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Asli Cakir
- Department of Pathology, Istanbul Medipol University, Istanbul, Turkey
| | - Omer Fatih Olmez
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Cem Gezen
- Department of Breast Surgery, Istanbul Medipol University, Istanbul, Turkey
| | - Ozcan Yildiz
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
5
|
Sharma S, Kaur V, Duhan P, Singh R, Agnihotri N. Evaluation of Anticancer Activity of Novel and Tumor-Targeted Glutamine-Conjugated Organotin(IV) Compounds in Colorectal Cancer─An In Vitro and In Vivo Study. J Med Chem 2025. [PMID: 39834112 DOI: 10.1021/acs.jmedchem.4c01728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Over the years, numerous ligand-based organotin(IV) Schiff base compounds have shown remarkable cytotoxicity and anticancer activities, but their clinical use is restricted by systemic toxicity, prompting the search for targeted therapies. Targeted delivery can be enhanced by exploiting the inherent characteristics of cancer cells such as glutamine addiction, which is essential to support cellular biosynthesis and cell growth to sustain aberrant proliferation. Our previous study revealed glutamine-conjugated organotin(IV) compounds have strong DNA/protein affinities, favorable in silico ADME profiles, and significant antiproliferative activity. In this study, these compounds demonstrated significant cytotoxicity against human colon carcinoma and adenocarcinoma cell lines via the induction of cell cycle arrest and apoptosis. In DMH/DSS-induced experimental colon carcinogenesis, these compounds reduced tumor burden and volume and inhibited cell proliferation and induced apoptosis, with minimal toxicity. Tissue distribution studies revealed selective accumulation in the colon. These findings support their potential as chemotherapeutic candidates for colon cancer.
Collapse
Affiliation(s)
- Shagun Sharma
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Varinder Kaur
- Department of Chemistry, Panjab University, Chandigarh 160014, India
| | - Pratibha Duhan
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Raghubir Singh
- Department of Chemistry, DAV College, Sector 10, Chandigarh 160011, India
| | - Navneet Agnihotri
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| |
Collapse
|
6
|
Bruno PS, Arshad A, Gogu MR, Waterman N, Flack R, Dunn K, Darie CC, Neagu AN. Post-Translational Modifications of Proteins Orchestrate All Hallmarks of Cancer. Life (Basel) 2025; 15:126. [PMID: 39860065 PMCID: PMC11766951 DOI: 10.3390/life15010126] [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: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Post-translational modifications (PTMs) of proteins dynamically build the buffering and adapting interface between oncogenic mutations and environmental stressors, on the one hand, and cancer cell structure, functioning, and behavior. Aberrant PTMs can be considered as enabling characteristics of cancer as long as they orchestrate all malignant modifications and variability in the proteome of cancer cells, cancer-associated cells, and tumor microenvironment (TME). On the other hand, PTMs of proteins can enhance anticancer mechanisms in the tumoral ecosystem or sustain the beneficial effects of oncologic therapies through degradation or inactivation of carcinogenic proteins or/and activation of tumor-suppressor proteins. In this review, we summarized and analyzed a wide spectrum of PTMs of proteins involved in all regulatory mechanisms that drive tumorigenesis, genetic instability, epigenetic reprogramming, all events of the metastatic cascade, cytoskeleton and extracellular matrix (ECM) remodeling, angiogenesis, immune response, tumor-associated microbiome, and metabolism rewiring as the most important hallmarks of cancer. All cancer hallmarks develop due to PTMs of proteins, which modulate gene transcription, intracellular and extracellular signaling, protein size, activity, stability and localization, trafficking, secretion, intracellular protein degradation or half-life, and protein-protein interactions (PPIs). PTMs associated with cancer can be exploited to better understand the underlying molecular mechanisms of this heterogeneous and chameleonic disease, find new biomarkers of cancer progression and prognosis, personalize oncotherapies, and discover new targets for drug development.
Collapse
Affiliation(s)
- Pathea Shawnae Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Aneeta Arshad
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Maria-Raluca Gogu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania;
| | - Natalie Waterman
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Rylie Flack
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Kimberly Dunn
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
| |
Collapse
|
7
|
Nyqvist-Streng J, Helou M, Helou K, Chamalidou C, Kovács A, Parris TZ. The prognostic value of changes in Ki67 following neoadjuvant chemotherapy in residual triple-negative breast cancer: a Swedish nationwide registry-based study. Breast Cancer Res Treat 2025:10.1007/s10549-025-07610-z. [PMID: 39799529 DOI: 10.1007/s10549-025-07610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
PURPOSE To evaluate the prognostic significance of changes in pre- and post-neoadjuvant chemotherapy (NACT) Ki67 in patients with primary invasive triple-negative breast cancer (TNBC). METHODS Population-based registry data were retrieved for patients diagnosed with TNBC between 2007 and 2021 (n = 9262). Multivariable Cox regression analysis was performed for disease-specific survival (DSS) and overall survival (OS) adjusted for age and residual disease in the breast and nodes (RDBN). RESULTS Of the 1777 TNBC patients receiving NACT, 54 achieved pathologic complete response (pCR) and 755 had residual disease. Most patients were overweight with stage II disease (78%), grade 3 tumors (53%), and RDBN score 3 (42%). Compared to baseline, tumor size (30 vs. 15 mm; P < 0.0001) and Ki67 levels (63% vs. 48%; P < 2.2e - 16) generally decreased after NACT. Although only 5% of samples increased in size, Ki67 levels often remained unchanged (75%) or increased (0.9%) after treatment, respectively. However, 34% of patients discontinued treatment. Patients showing no changes in Ki67% had more unfavorable OS (P < 0.0001) and DSS (P = 0.00032), with significantly lower 5-year survival probabilities (OS: 66%; DSS: 78%) than those with decreased Ki67% (OS: 87%; DSS: 89%). All patients reaching pCR were alive 5 years after diagnosis. However, only the RDBN score was an independent predictor of survival in the multivariable analyses. CONCLUSION Ki67 often remained unchanged in TNBC patients treated with neoadjuvant chemotherapy, resulting in adverse clinical outcomes. These findings highlight the need for individualized treatment regimens and dynamic monitoring of TNBC patients with high Ki67 post-NACT.
Collapse
Affiliation(s)
- Jenny Nyqvist-Streng
- Department of Surgery, Region Västra Götaland, Skaraborg Hospital, Skövde, Sweden.
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Mikael Helou
- Department of Psychiatry, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chaido Chamalidou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Region Västra Götaland, Skaraborg Hospital, Skövde, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Błaszczak E, Miziak P, Odrzywolski A, Baran M, Gumbarewicz E, Stepulak A. Triple-Negative Breast Cancer Progression and Drug Resistance in the Context of Epithelial-Mesenchymal Transition. Cancers (Basel) 2025; 17:228. [PMID: 39858010 PMCID: PMC11764116 DOI: 10.3390/cancers17020228] [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: 12/06/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most difficult subtypes of breast cancer to treat due to its distinct clinical and molecular characteristics. Patients with TNBC face a high recurrence rate, an increased risk of metastasis, and lower overall survival compared to other breast cancer subtypes. Despite advancements in targeted therapies, traditional chemotherapy (primarily using platinum compounds and taxanes) continues to be the standard treatment for TNBC, often with limited long-term efficacy. TNBC tumors are heterogeneous, displaying a diverse mutation profile and considerable chromosomal instability, which complicates therapeutic interventions. The development of chemoresistance in TNBC is frequently associated with the process of epithelial-mesenchymal transition (EMT), during which epithelial tumor cells acquire a mesenchymal-like phenotype. This shift enhances metastatic potential, while simultaneously reducing the effectiveness of standard chemotherapeutics. It has also been suggested that EMT plays a central role in the development of cancer stem cells. Hence, there is growing interest in exploring small-molecule inhibitors that target the EMT process as a future strategy for overcoming resistance and improving outcomes for patients with TNBC. This review focuses on the progression and drug resistance of TNBC with an emphasis on the role of EMT in these processes. We present TNBC-specific and EMT-related molecular features, key EMT protein markers, and various signaling pathways involved. We also discuss other important mechanisms and factors related to chemoresistance in TNBC within the context of EMT, highlighting treatment advancements to improve patients' outcomes.
Collapse
Affiliation(s)
- Ewa Błaszczak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland
| | | | | | | | | | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodzki Street, 20-093 Lublin, Poland
| |
Collapse
|
9
|
Drowne T, Armgardt E, Svoboda A. Real-world experience of abemaciclib for adjuvant and metastatic breast cancer. J Oncol Pharm Pract 2025; 31:141-146. [PMID: 39196656 DOI: 10.1177/10781552241279189] [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: 08/30/2024]
Abstract
OBJECTIVE Hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer is the most common subtype. Abemaciclib, an inhibitor of cyclin-dependent kinases 4 and 6, was approved to reduce risk of recurrence in high-risk, HR+, HER2-, early breast cancer based on the monarchE trial. The most common adverse events reported in monarchE were diarrhea, neutropenia, and fatigue. Real-world tolerability data and incidence of adverse events with abemaciclib in the adjuvant setting versus the metastatic setting is lacking. DATA SOURCES This is a retrospective analysis of HR+, HER2- breast cancer patients on abemaciclib from March 2018 to September 2021 at Robert H. Lurie Comprehensive Cancer Center in Chicago, Illinois. Incidence, grade of adverse events, dose reductions, and discontinuations were evaluated in patients taking abemaciclib in the adjuvant setting and the metastatic setting. DATA SUMMARY Of the 30 patients included in this analysis, 100% experienced an adverse event of any grade. During treatment, 12.5% treated in the adjuvant setting and 35.7% in the metastatic setting experienced grade ≥3 adverse events. Adverse events leading to discontinuation of abemaciclib occurred in 18.8% of patients in the adjuvant setting and 57.1% in the metastatic setting. CONCLUSIONS This data suggests abemaciclib is better tolerated in high-risk, HR+, HER2-, node-positive, early breast cancer treated in the adjuvant setting compared to the metastatic setting. Management of adverse events is crucial to help patients stay on therapy to improve clinical outcomes. Real-world tolerability of abemaciclib in both the adjuvant and metastatic settings is of importance.
Collapse
Affiliation(s)
- Taylor Drowne
- Department of Pharmacy, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Emily Armgardt
- Department of Pharmacy, Robert H. Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL, USA
| | - Alison Svoboda
- Department of Pharmacy, Robert H. Lurie Comprehensive Cancer Center of Northwestern, Chicago, IL, USA
| |
Collapse
|
10
|
Alam MR, Seo KJ, Yim K, Liang P, Yeh J, Chang C, Chong Y. Comparative analysis of Ki-67 labeling index morphometry using deep learning, conventional image analysis, and manual counting. Transl Oncol 2025; 51:102159. [PMID: 39489091 PMCID: PMC11567953 DOI: 10.1016/j.tranon.2024.102159] [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: 12/20/2023] [Revised: 08/25/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
The Ki-67 labeling index is essential for predicting the prognosis of breast cancer and for diagnosing neuroendocrine and gastrointestinal stromal tumors. However, current manual counting and digital image analysis (DIA)-based methods are limited in terms of accurate estimation. This study aimed to assess and compare the capabilities of different DIA systems for Ki-67 counting using the conventional manual counting method. A total of 239 tissue microarray cores from patients with stomach cancer were immunohistochemically stained for Ki-67 and digitally scanned. For the analysis, we employed three different annotation methods: whole TMA core, box selection of the epithelium, and hand-free selection of the epithelium. We used DIA system of 3DHistech, Roche, aetherAI, and manual counting by the pathologists. The annotation methods showed different Ki-67 positivity but were lower than the pathologist manual counting. The results demonstrate that the Roche system is the preferred method for analyzing the entire TMA, whereas aetherAI outperforms the box selection method. Furthermore, 3DHistech is the most accurate method for hands-free selection of the epithelium. The manual counting results showed good agreement among pathologists, with an average intraclass correlation coefficient of 0.93. These results emphasize the importance of carefully selecting annotation methods to determine Ki-67 positivity. To determine the most suitable method for individual laboratories, multiple approaches should be assessed before implementing a DIA system in routine practice.
Collapse
Affiliation(s)
- Mohammad Rizwan Alam
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kyung Jin Seo
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kwangil Yim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | | | - Joe Yeh
- aetherAI Co., Ltd, Taipei, Taiwan
| | | | - Yosep Chong
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
11
|
Nath P, Alfarsi LH, El-Ansari R, Masisi BK, Erkan B, Fakroun A, Ellis IO, Rakha EA, Green AR. The amino acid transporter SLC7A11 expression in breast cancer. Cancer Biol Ther 2024; 25:2291855. [PMID: 38073087 PMCID: PMC10761065 DOI: 10.1080/15384047.2023.2291855] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC), characterized by its diverse molecular profiles and clinical outcomes, presents a significant challenge in the development of effective therapeutic strategies. Metabolic reprogramming, a defining characteristic of cancer, has emerged as a promising target for novel therapies. SLC7A11, an amino acid transporter that facilitates cysteine uptake in exchange for glutamate, plays a crucial role in sustaining the altered metabolism of cancer cells. This study delves into the comprehensive analysis of SLC7A11 at the genomic, transcriptomic, and protein levels in extensive BC datasets to elucidate its potential role in different BC subtypes. SLC7A11 gene copy number and mRNA expression were evaluated using the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort (n = 1,980) and Breast Cancer Gene Expression Miner (n = 4,712). SLC7A11 protein was assessed using immunohistochemistry in a large BC cohort (n = 1,981). Additionally, The Cancer Genome Atlas (TCGA) dataset was used to explore SLC7A11 DNA methylation patterns using MethSurv (n = 782) and association of SLC7A11 mRNA expression with immune infiltrates using TIMER (n = 1,100). High SLC7A11 mRNA and SLC7A11 protein expression were significantly associated with high tumor grade (p ≤ .02), indicating a potential role in cancer progression. Interestingly, SLC7A11 copy number gain was observed in HER2+ tumors (p = .01), suggesting a subtype-specific association. In contrast, SLC7A11 mRNA expression was higher in the basal-like/triple-negative (TN; p < .001) and luminal B tumors (p = .02), highlighting its differential expression across BC subtypes. Notably, high SLC7A11 protein expression was predominantly observed in Estrogen Receptor (ER)-negative and Triple Negative (TN) BC, suggesting a role in these aggressive subtypes. Further analysis revealed that SLC7A11 was positively correlated with other amino acid transporters and enzymes associated with glutamine metabolism, implying a coordinated role in metabolic regulation. Additionally, SLC7A11 gene expression was positively associated with neutrophil and macrophage infiltration, suggesting a potential link between SLC7A11 and tumor immunity. Our findings suggest that SLC7A11 plays a significant role in BC metabolism, demonstrating differential expression across subtypes and associations with poor patient outcomes. Further functional studies are warranted to elucidate the precise mechanisms by which SLC7A11 contributes to BC progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Preyanka Nath
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Lutfi H. Alfarsi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Rokaya El-Ansari
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Brendah K. Masisi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Busra Erkan
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ali Fakroun
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ian O. Ellis
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A. Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R. Green
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
12
|
Korpinen K, Autere TA, Tuominen J, Löyttyniemi E, Eigeliene N, Talvinen K, Kronqvist P. Personalized multifactorial risk assessment in neoadjuvant-treated breast carcinoma. Breast Cancer Res Treat 2024:10.1007/s10549-024-07584-4. [PMID: 39739270 DOI: 10.1007/s10549-024-07584-4] [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: 10/28/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
PURPOSE Due to biological heterogeneity of breast carcinoma, predicting the individual response to neoadjuvant treatment (NAT) is complex. Consequently, there are no comprehensive, generally accepted practices to guide post-treatment follow-up. We present clinical and histopathological criteria to advance the prediction of disease outcome in NA-treated breast cancer. METHODS A retrospective consecutive cohort of 257 NA-treated Finnish breast cancer patients with up to 13-year follow-up and the corresponding tissue samples of pre- and post-NAT breast and metastatic specimen were evaluated for prognostic impacts. All relevant clinical and biomarker characteristics potentially correlated with tumor response to NAT, course of disease, or outcome of breast cancer were included in the statistical analyses. RESULTS The results highlight the intensified characterization of distinguished prognostic factors and previously overlooked histological features, e.g., mitotic and apoptotic activity. Particularly, decreased PR indicated 3.8-fold (CI 1.9-7.4, p = 0.0001) mortality risk, and a > 10.5-year shorter survival for the majority, > 75% of patients (Q1). Clinically applicable prognostic factors both preceding and following NAT were identified and compiled into heat maps to quantify mortality and recurrence risks. Combinations of risk factors for aggressive disease were exemplified as an interactive tool (bcnatreccalc.utu.fi) to illustrate the spectrum of disease outcomes. CONCLUSION The results emphasize the value of comprehensive evaluation of conventional patient and biomarker characteristics, especially concerning re-assessment of biomarkers, risk-adapted surveillance, and personalized treatment strategies. Future personalized NA-treatment strategies might benefit from models combining risk-adapted surveillance data and post-NAT re-assessed biomarkers.
Collapse
Affiliation(s)
- K Korpinen
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10/MedD5A, 20500, Turku, Finland.
| | - T A Autere
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10/MedD5A, 20500, Turku, Finland
| | - J Tuominen
- Department of Pathology, Turku University Hospital, Turku, Finland
| | - E Löyttyniemi
- Department of Biostatistics, University of Turku, Turku, Finland
| | - N Eigeliene
- Department of Oncology, Vaasa Central Hospital, Vaasa, Finland
| | - K Talvinen
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10/MedD5A, 20500, Turku, Finland
| | - P Kronqvist
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10/MedD5A, 20500, Turku, Finland
- Department of Pathology, Turku University Hospital, Turku, Finland
| |
Collapse
|
13
|
Nethi SK, Kothadiya S, White BM, Rachagani S, Bardhan R, Mallapragada SK. Polyanhydride Copolymer-Based Niclosamide Nanoparticles for Inhibiting Triple-Negative Breast Cancer: Metabolic Responses and Synergism with Paclitaxel. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70362-70377. [PMID: 39666980 DOI: 10.1021/acsami.4c17961] [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: 12/14/2024]
Abstract
The heterogeneity of tumors and the lack of effective therapies have resulted in triple-negative breast cancer (TNBC) exhibiting the least favorable outcomes among breast cancer subtypes. TNBC is characterized by its aggressive nature, often leading to high rates of relapse, metastasis, and mortality. Niclosamide (Nic), an Food and Drug Administration-approved anthelmintic drug, has been repurposed for cancer treatment; however, its application for TNBC is hindered by significant challenges, including strong hydrophobicity, poor aqueous solubility, and low bioavailability. This study aimed to develop Nic nanoparticles (Nic NPs) using biodegradable and biocompatible polyanhydride copolymers to enhance Nic's bioavailability and therapeutic efficacy. Nic NPs effectively inhibited migration, proliferation, and clonogenicity in both murine and human TNBC cells, inducing apoptosis and suppressing STAT3 signaling. For the first time, we utilized Raman spectroscopy and Seahorse extracellular flux assays to demonstrate the metabolic responses of TNBC cells to Nic NPs, revealing significant metabolic alterations, including the inhibition of mitochondrial respiration and glycolysis. Additionally, this study is the first to explore the combination therapy of repurposed Nic with the approved chemotherapeutic agent paclitaxel in the 4T1 TNBC immunocompetent mouse model. The combination of Nic NPs and paclitaxel significantly reduced tumor growth without adversely affecting the body weight of tumor-bearing mice. In summary, these findings suggest that Nic NPs could serve as a promising component in combination therapies for the effective treatment of TNBC.
Collapse
Affiliation(s)
- Susheel Kumar Nethi
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa50011, United States
| | - Siddhant Kothadiya
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
| | - Brianna M White
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri 65211, United States
- Roy Blunt NextGen Precision Health, University of Missouri, Columbia, Missouri 65211, United States
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa50011, United States
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa50011, United States
| |
Collapse
|
14
|
Mokhtar A, Mohamed T, Eigza AO, El-Khouly ME. Combining Water-Soluble Porphyrin and Phthalocyanine Photosensitizers With Doxorubicin Improves the Efficacy of Chemo-Photodynamic Therapy Against DMBA-Induced Breast Carcinoma. Chem Biodivers 2024:e202402782. [PMID: 39714972 DOI: 10.1002/cbdv.202402782] [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: 10/30/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024]
Abstract
Breast cancer ranks as the second most widespread form of cancer globally. Currently, combination therapy is being actively employed in clinical practice to augment the efficiency of anticancer treatment. Hence, the objective of this study was to assess the therapeutic efficacy of a combination of femtosecond laser-based photodynamic therapy (PDT) utilizing two distinct photosensitizers (PSs), zinc phthalocyanine tetrasulfonate (ZnPcS4) and α,β,χ,δ porphyrin-Tetrakis (1-methylpyridinium-4-yl) p-Toluenesulfonate porphyrin (TMPyP) in conjunction with doxorubicin chemotherapeutic agent, on mammary carcinomas experimentally induced in female mice using 7,12-dimethylbenz[a] anthracene (DMBA). Our results showed the efficiency of the combined therapy for promoting tissue apoptosis and necrosis as evidenced by histopathological observations and the noticeable reduction of Bcl-2 and Ki-67 expression. Moreover, there was a reduction in serum levels of the carcinoma antigen CA15-3 and transforming growth factor beta (TGF-β). Co-treatment of doxorubicin with ZnPcS4-PDT or TMPyP-PDT or a combination of both resulted in a decrease in the expression of epidermal growth factor receptor (EGFR) and its downstream oncogenes NRAS, NF-κB, mTERT, and c-Myc, and an increase in the expression of the caspase-3 apoptotic gene. These results validate the therapeutic potential of combining doxorubicin with PDT, highlighting the potential of this co-treatment strategy as a promising alternative for enhancing existing anticancer approaches.
Collapse
Affiliation(s)
- Aya Mokhtar
- Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El Arab, Alexandria, Egypt
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Tarek Mohamed
- Laser Institute for Research and Applications (LIRA), Beni-Suef University, Beni Suef, Egypt
| | - Ahmed O Eigza
- Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El Arab, Alexandria, Egypt
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohamed E El-Khouly
- Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El Arab, Alexandria, Egypt
| |
Collapse
|
15
|
Toàn NM. Novel Molecular Classification of Breast Cancer with PET Imaging. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2099. [PMID: 39768978 PMCID: PMC11678748 DOI: 10.3390/medicina60122099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
Breast cancer is a heterogeneous disease characterized by a wide range of biomarker expressions, resulting in varied progression, behavior, and prognosis. While traditional biopsy-based molecular classification is the gold standard, it is invasive and limited in capturing tumor heterogeneity, especially in deep or metastatic lesions. Molecular imaging, particularly positron emission tomography (PET) imaging, offering a non-invasive alternative, potentially plays a crucial role in the classification and management of breast cancer by providing detailed information about tumor location, heterogeneity, and progression. This narrative review, which focuses on both clinical patients and preclinical studies, explores the latest advancements in PET imaging for breast cancer, emphasizing the development of new tracers targeting hormone receptors such as the estrogen alpha receptor, progesterone receptor, androgen receptor, estrogen beta receptor, as well as the ErbB family of receptors, VEGF/VEGFR, PARP1, PD-L1, and markers for indirectly assessing Ki-67. These innovative radiopharmaceuticals have the potential to guide personalized treatment approaches based on the unique tumor profiles of individual patients. Additionally, they may improve the assessment of treatment efficacy, ultimately leading to better outcomes for those diagnosed with breast cancer.
Collapse
Affiliation(s)
- Ngô Minh Toàn
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
- Medical Imaging Clinic, Clinical Centre, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
16
|
Liao T, Yang Y, Lin X, Ouyang R, Deng Y, Ma J. High-risk breast lesions: a combined intratumoral and peritumoral radiomics nomogram model to predict pathologic upgrade and reduce unnecessary surgical excision. Front Oncol 2024; 14:1479565. [PMID: 39744004 PMCID: PMC11688276 DOI: 10.3389/fonc.2024.1479565] [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: 08/12/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Objective This study aimed to develop a nomogram that combines intratumoral and peritumoral radiomics based on multi-parametric MRI for predicting the postoperative pathological upgrade of high-risk breast lesions and sparing unnecessary surgeries. Methods In this retrospective study, 138 patients with high-risk breast lesions (January 1, 2019, to January 1, 2023) were randomly divided into a training set (n=96) and a validation set (n=42) at a 7:3 ratio. The best-performing MRI sequence for intratumoral radiomics was selected to develop individual and combined radiomics scores (Rad-Scores). The best Rad-Score was integrated with independent clinical and radiological risk factors by a nomogram. The diagnostic performance of the nomogram was evaluated using the area under the curve (AUC) of the receiver operating characteristic curve, along with accuracy, specificity, and sensitivity analysis. Results The nomogram based on the combined intratumoral and peritumoral Rad-Score of the dynamic contrast-enhanced MRI and clinical-radiological features achieved superior diagnostic efficacy in the training (AUC=0.914) and validation set (AUC=0.867) compared to other models. It also achieved a specificity and accuracy of 85.1% and 82.3% during training and 66.7% and 76.2% during validation. Conclusion The nomogram encapsulating the combined intratumoral and peritumoral radiomics demonstrated superior diagnostic efficacy in postoperative pathological upgrades of high-risk breast lesions, enabling clinicians to make more informed decisions about interventions and follow-up strategies.
Collapse
Affiliation(s)
- Tingting Liao
- Department of Radiology, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Yuting Yang
- Department of Radiology, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Xiaohui Lin
- Department of Radiology, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Rushan Ouyang
- Department of Radiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yaohong Deng
- Department of Research & Development, Yizhun Medical AI Co. Ltd., Beijing, China
| | - Jie Ma
- Department of Radiology, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
17
|
Ekholm A, Wang Y, Vallon-Christersson J, Boissin C, Rantalainen M. Prediction of gene expression-based breast cancer proliferation scores from histopathology whole slide images using deep learning. BMC Cancer 2024; 24:1510. [PMID: 39663527 PMCID: PMC11633006 DOI: 10.1186/s12885-024-13248-9] [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: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND In breast cancer, several gene expression assays have been developed to provide a more personalised treatment. This study focuses on the prediction of two molecular proliferation signatures: an 11-gene proliferation score and the MKI67 proliferation marker gene. The aim was to assess whether these could be predicted from digital whole slide images (WSIs) using deep learning models. METHODS WSIs and RNA-sequencing data from 819 invasive breast cancer patients were included for training, and models were evaluated on an internal test set of 172 cases as well as on 997 cases from a fully independent external test set. Two deep Convolutional Neural Network (CNN) models were optimised using WSIs and gene expression readouts from RNA-sequencing data of either the proliferation signature or the proliferation marker, and assessed using Spearman correlation (r). Prognostic performance was assessed through Cox proportional hazard modelling, estimating hazard ratios (HR). RESULTS Optimised CNNs successfully predicted the proliferation score and proliferation marker on the unseen internal test set (ρ = 0.691(p < 0.001) with R2 = 0.438, and ρ = 0.564 (p < 0.001) with R2 = 0.251 respectively) and on the external test set (ρ = 0.502 (p < 0.001) with R2 = 0.319, and ρ = 0.403 (p < 0.001) with R2 = 0.222 respectively). Patients with a high proliferation score or marker were significantly associated with a higher risk of recurrence or death in the external test set (HR = 1.65 (95% CI: 1.05-2.61) and HR = 1.84 (95% CI: 1.17-2.89), respectively). CONCLUSIONS The results from this study suggest that gene expression levels of proliferation scores can be predicted directly from breast cancer morphology in WSIs using CNNs and that the predictions provide prognostic information that could be used in research as well as in the clinical setting.
Collapse
Affiliation(s)
- Andreas Ekholm
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, Stockholm, 171 77, Sweden
| | - Yinxi Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, Stockholm, 171 77, Sweden
| | | | - Constance Boissin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, Stockholm, 171 77, Sweden
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, Stockholm, 171 77, Sweden.
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
18
|
Aljohani AI. Prognostic Significance of DSCC1, a Biomarker Associated with Aggressive Features of Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1929. [PMID: 39768811 PMCID: PMC11677291 DOI: 10.3390/medicina60121929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Invasive breast cancer (BC) was traditionally investigated visually, and no technique could identify the key molecular drivers of patient survival. However, essential molecular drivers of invasive BC have now been discovered using innovative genomic, transcriptomic, and proteomic methodologies. Nevertheless, few evaluations of the prognostic factors of BC in Saudi Arabia have been performed. Evaluating the biomarkers associated with the development of early-stage BC could help determine the risk of metastasis and guide treatment decisions. In a previous study, using large BC cohorts and artificial neural network techniques, DNA replication and sister chromatid cohesion 1 (DSCC1) was found to be one of the principal genes in invasive BC samples. To date, no studies have addressed the prognostic significance of DSCC1 in invasive BC and its association with aggressive tumor behavior. This research aimed to address this gap. Materials and Methods: The association of clinicopathological features and patient outcomes with DSCC1 expression at the mRNA level was assessed using the Molecular Taxonomy Breast Cancer International Consortium (METABRIC; n = 1980) and The Cancer Genome Atlas (TCGA; n = 854) cohorts. DSCC1 was also evaluated at the protein level using immunohistochemistry on samples from invasive BC patients (n = 100) presenting to King Abdul Aziz Specialist Hospital in Saudi Arabia. The association of clinicopathological parameters (including patient age, tumor grade, tumor size, and patient outcome) with protein level was also evaluated. Results: In both METABRIC and TCGA cohorts, high expression of DSCC1 was significantly associated with high histological grade, large tumor size, lymphovascular invasion positivity, and hormone receptor negativity (all p < 0.001). A high DSCC1 mRNA level was associated with poor outcomes (p < 0.001 for METABRIC, p = 0.23 for TCGA). At the protein level, high DSCC1 expression was associated with high histological grade (p = 0.001), lymph node presence (p = 0.008), hormone receptor negativity (p = 0.005), high Ki67 expression (p = 0.036), and shorter survival (p = 0.008). Conclusions: This study confirmed the prognostic significance of DSCC1 in invasive BC patients. DSCC1 could be a therapeutic target in BC cases with poor outcomes.
Collapse
Affiliation(s)
- Abrar I Aljohani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| |
Collapse
|
19
|
El-Masry TA, El-Nagar MMF, Oriquat GA, Alotaibi BS, Saad HM, El Zahaby EI, Ibrahim HA. Therapeutic efficiency of Tamoxifen/Orlistat nanocrystals against solid ehrlich carcinoma via targeting TXNIP/HIF1-α/MMP-9/P27 and BAX/Bcl2/P53 signaling pathways. Biomed Pharmacother 2024; 180:117429. [PMID: 39293373 DOI: 10.1016/j.biopha.2024.117429] [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: 06/27/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Orlistat (Orli) is an anti-obesity medication that has been approved by the US Food and Drug Administration. It has relatively limited oral bioavailability with promising inhibitory effects on cell proliferation as well as reducing the growth of tumors. AIMS This investigation was done to evaluate the potential protective effect of Tamoxifen/Orlistat nanocrystals alone or in combination against Solid Ehrlich Carcinoma (SEC) and to clarify the possible underlying influences. MATERIALS AND METHODS The liquid antisolvent precipitation technique (bottom-up technology) was utilized to manufacture Orlistat Nanocrystals. To explore potential causes for the anti-tumor action, female Swiss Albino mice bearing SEC were randomly assigned into five equal groups (n = 6). Group 1: Tumor control group, group 2: Tam group: tamoxifen (0.01 g/kg, IP), group 3: Free-Orli group: orlistat (0.24 g/kg, IP), group 4: Nano-Orli: orlistat nanocrystals (0.24 g/kg, IP), group 5: Tam-Nano-Orli: Both doses of Tam and Nano-Orli. All treatments were administered for 16 days. KEY FINDINGS The untreated mice showed development in the tumor volume and weight. As well as histopathology results from these mice revealed many tumor large cells as well as solid sheets of malignant cells. Also, untreated mice showed raised VEGF and TGF-1beta content. Moreover, results of gene expression in the SEC-bearing mice noted upregulation in HIF-1α, MMP-9, Bcl-2, and P27 gene expression and downregulation of TXNIP, BAX, and P53 gene expression. On the other hand, administrated TAM, Free-Orli, Nano-Orli, and a combination of Tam-Nano-Orli distinctly suppressed the tumor effects on estimated parameters with special reference to Tam-Nano-Orli. SIGNIFICANCE The developed Tamoxifen/Orlistat nanocrystals combination could be considered a promising approach to augment antitumor effects.
Collapse
Affiliation(s)
- Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Maysa M F El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ghaleb Ali Oriquat
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Cairo 51511, Egypt.
| | - Enas I El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt.
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
20
|
Huang Z, Wang M, Kong Y, Li G, Tian H, Wu H, Zheng J, Mo S, Xu J, Dong F. Photoacoustic-Based Intra- and Peritumoral Radiomics Nomogram for the Preoperative Prediction of Expression of Ki-67 in Breast Malignancy. Acad Radiol 2024. [DOI: 10.1016/j.acra.2024.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
21
|
Fujisawa S, Takagi K, Yamaguchi-Tanaka M, Sato A, Miki Y, Miyashita M, Tada H, Ishida T, Suzuki T. Receptor for Hyaluronan Mediated Motility (RHAMM)/Hyaluronan Axis in Breast Cancer Chemoresistance. Cancers (Basel) 2024; 16:3600. [PMID: 39518040 PMCID: PMC11545538 DOI: 10.3390/cancers16213600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Receptor for hyaluronan-mediated motility (RHAMM) is a hyaluronan (HA) receptor, which exerts diverse biological functions in not only physiological but also pathological conditions in human malignancies, including breast cancer. Although chemoresistance is a significant clinical challenge in breast cancer, a possible contribution of RHAMM and hyaluronan to breast cancer chemoresistance has remained unclear. Methods: We immunolocalized RHAMM and HA in breast carcinoma tissues. Also, we utilized epirubicin-sensitive (parental) and rpirubicin-resistant (EPIR) breast cancer cell lines to explore the role of RHAMMM in breast cancer progression. Results: We found out that RHAMM and HA were cooperatively correlated with breast cancer aggressiveness and recurrence after chemotherapy. In vitro studies demonstrated that RHAMM was overexpressed in EPIR cells compared to parental cells. In addition, the knockdown of RHAMM significantly suppressed proliferation and migration of both parental and EPIR cells. On the other hand, the expression level of cancer stem cell marker CD44, which was overexpressed in M-EPIR (epirubicin-resistant MCF-7 subline) compared to MCF-7, was significantly suppressed by knockdown of RHAMM. In addition, the knockdown of RHAMM significantly altered the expression of N-cadherin and E-cadherin, leading to an epithelial phenotype. Conclusions: Aberrant RHAMM signaling were considered to cause chemoresistance related to cancer stemness and epithelial to mesenchymal transition, and increased cell proliferation and migration of both chemo-sensitive and chemo-resistant breast cancer cells.
Collapse
Affiliation(s)
- Shiori Fujisawa
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (S.F.); (M.M.); (H.T.)
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.Y.-T.); (A.S.); (T.S.)
| | - Kiyoshi Takagi
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.Y.-T.); (A.S.); (T.S.)
| | - Mio Yamaguchi-Tanaka
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.Y.-T.); (A.S.); (T.S.)
- Personalized Medicine Center, Tohoku University Hospital, Sendai 980-8574, Miyagi, Japan
| | - Ai Sato
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.Y.-T.); (A.S.); (T.S.)
| | - Yasuhiro Miki
- Department of Anatomic Pathology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Miyagi, Japan;
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (S.F.); (M.M.); (H.T.)
| | - Hiroshi Tada
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (S.F.); (M.M.); (H.T.)
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (S.F.); (M.M.); (H.T.)
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (M.Y.-T.); (A.S.); (T.S.)
- Department of Anatomic Pathology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Miyagi, Japan;
- Department of Pathology, Tohoku University Hospital, Sendai 980-8574, Miyagi, Japan
| |
Collapse
|
22
|
Heikal LA, Ashour AA, Aboushanab AR, El-Kamel AH, Zaki II, El-Moslemany RM. Microneedles integrated with atorvastatin-loaded pumpkisomes for breast cancer therapy: A localized delivery approach. J Control Release 2024; 376:354-368. [PMID: 39413849 DOI: 10.1016/j.jconrel.2024.10.013] [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: 05/31/2024] [Revised: 09/20/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Breast cancer is the most common invasive cancer in women worldwide, having a significant impact on women's well-being. Early diagnosis of breast cancer followed by appropriate treatment is considered the best survival factor. Microneedles (MN) have been utilized for non-invasive localized breast cancer treatment. The combination of nano-carriers with MN technology represents an appealing strategy for improving drug delivery efficacy. It is worth noting that atorvastatin (ATV) has received substantial interest as a drug with potential anticancer activity. Our study aimed to formulate an ATV-loaded bioactive pumpkin seed oil vesicular nanocarrier; pumpkisomes (PUMP) for enhanced localized delivery to breast cancer using MN. The selected PUMP formulation had a particle size of 151.8 ± 2.7 nm, zeta potential of -54.1 mV, and % entrapment efficiency of 73 %. PUMP showed a sustained ATV release, potent selective cytotoxic effect (IC50 of 2.82 ± 0.02 μg/mL), enhanced internalization (2.8-fold increase compared to the free drug), and potent anti-migratory effect on MDA-MB-231 cells (21.15 ± 3.6 % wound closure compared to 80.81 ± 4.1 % for free drug). Moreover, integrating ATV-PUMP in dissolving microneedles (ATV-PUMP@dMN) showed a quick dissolution rate and appropriate mechanical strength with high piercing efficiency. ATV permeation across the skin from ATV-PUMP@dMN was also improved (1.8-fold increase compared to ATV-PUMP@gel). ATV-PUMP@dMN demonstrated an efficient anticancer effect when applied in an Ehrlich ascites mammary tumor model attaining significant improvement in ATV antiproliferative (PTEN and Ki-67), antiangiogenic (VEGF) and apoptotic (Bcl2, Bax and caspase3) effects restoring tumor biomarkers to levels comparable to the negative control group. Thus, our study presents PUMP as a novel and promising bioactive vesicular nanosystem with potential synergistic effect with ATV or other antitumor drugs. PUMP-integrated MN could be considered a promising platform for future applications in localized breast cancer therapy.
Collapse
Affiliation(s)
- Lamia A Heikal
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Asmaa A Ashour
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Alaa R Aboushanab
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amal H El-Kamel
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Inass I Zaki
- Department of Pathology, Faculty of Medicine Alexandria University, Alexandria, Egypt
| | - Riham M El-Moslemany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
23
|
Chang H, Chen J, Wang D, Li H, Ming L, Li Y, Yu D, Yang YX, Kong P, Jia W, Yan Q, Liu X, Zeng Q. Multimodal apparent diffusion MRI model in noninvasive evaluation of breast cancer and Ki-67 expression. Cancer Imaging 2024; 24:137. [PMID: 39394171 PMCID: PMC11470582 DOI: 10.1186/s40644-024-00780-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND To assess the capability of multimodal apparent diffusion (MAD) weighted magnetic resonance imaging (MRI) to distinguish between malignant and benign breast lesions, and to predict Ki-67 expression level in breast cancer. METHODS This retrospective study was conducted with 93 patients who had postoperative pathology-confirmed breast cancer or benign breast lesions. MAD images were acquired using a 3.0 T MRI scanner with 16 b values. The MAD parameters, as flow (fF, DF), unimpeded (fluid) (fUI), hindered (fH, DH, and αH), and restricted (fR, DR), were calculated. The differences of the parameters were compared by Mann-Whitney U test between the benign/malignant lesions and high/low Ki-67 expression level. The diagnostic performance was assessed by the area under the receiver operating characteristic curve (AUC). RESULTS The fR in the malignant lesions was significantly higher than in the benign lesions (P = 0.001), whereas the fUI and DH were found to be significantly lower (P = 0.007 and P < 0.001, respectively). Compared with individual parameter in differentiating malignant from benign breast lesions, the combination parameters of MAD (fR, DH, and fUI) provided the highest AUC (0.851). Of the 73 malignant lesions, 42 (57.5%) were assessed as Ki-67 low expression and 31 (42.5%) were Ki-67 high expression. The Ki-67 high status showed lower DH, higher DF and higher αH (P < 0.05). The combination parameters of DH, DF, and αH provided the highest AUC (0.691) for evaluating Ki-67 expression level. CONCLUSIONS MAD weighted MRI is a useful method for the breast lesions diagnostics and the preoperative prediction of Ki-67 expression level.
Collapse
Affiliation(s)
- Huan Chang
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan, Shandong, China
| | - Jinming Chen
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan, Shandong, China
| | - Dawei Wang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Hongxia Li
- Department of Radiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Ming
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yuting Li
- Department of Radiology, The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dan Yu
- United Imaging Research Institute of Intelligent Imaging, Beijing, People's Republic of China
| | - Yu Xin Yang
- United Imaging Research Institute of Intelligent Imaging, Beijing, People's Republic of China
| | - Peng Kong
- Department of Breast Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Wenjing Jia
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical Universityand, Shandong Academy of Medical Sciences , Jinan, Shandong, China
| | - Qingqing Yan
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical Universityand, Shandong Academy of Medical Sciences , Jinan, Shandong, China
| | - Xinhui Liu
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical Universityand, Shandong Academy of Medical Sciences , Jinan, Shandong, China
| | - Qingshi Zeng
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan, Shandong, China.
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China.
| |
Collapse
|
24
|
Hong SP, Lee SM, Yoo ID, Lee JE, Han SW, Kim SY, Lee JW. Clinical value of SUVpeak-to-tumor centroid distance on FDG PET/CT for predicting neoadjuvant chemotherapy response in patients with breast cancer. Cancer Imaging 2024; 24:136. [PMID: 39394156 PMCID: PMC11468257 DOI: 10.1186/s40644-024-00787-4] [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: 07/09/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Since it has been found that the maximum metabolic activity of a cancer lesion shifts toward the lesion edge during cancer progression, normalized distances from the hot spot of radiotracer uptake to tumor centroid (NHOC) and tumor perimeter (NHOP) have been suggested as novel F-18 fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) parameters that can reflect cancer aggressiveness. This study aimed to investigate whether NHOC and NHOP parameters could predict pathological response to neoadjuvant chemotherapy (NAC) and progression-free survival (PFS) in breast cancer patients. METHODS This study retrospectively enrolled 135 female patients with breast cancer who underwent pretreatment FDG PET/CT and received NAC and subsequent surgical resection. From PET/CT images, normalized distances of maximum SUV and peak SUV-to-tumor centroid (NHOCmax and NHOCpeak) and -to-tumor perimeter (NHOPmax and NHOPpeak) were measured, in addition to conventional PET/CT parameters. RESULTS Of 135 patients, 32 (23.7%) achieved pathological complete response (pCR), and 34 (25.2%) had events during follow-up. In the receiver operating characteristic (ROC) curve analysis, NHOCmax showed the highest area under the ROC curve value (0.710) for predicting pCR, followed by NHOCpeak (0.694). In the multivariate logistic regression analysis, NHOCmax, NHOCpeak, and NHOPmax were independent predictors for pCR (p < 0.05). In the multivariate survival analysis, NHOCpeak (p = 0.026) was an independent predictor for PFS along with metabolic tumor volume, with patients having higher NHOCpeak showing worse PFS. CONCLUSION NHOCpeak on pretreatment FDG PET/CT could be a potential imaging parameter for predicting NAC response and survival in patients with breast cancer.
Collapse
Affiliation(s)
- Sun-Pyo Hong
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam-gu, Cheonan, 31151, Republic of Korea
| | - Sang Mi Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam-gu, Cheonan, 31151, Republic of Korea
| | - Ik Dong Yoo
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam-gu, Cheonan, 31151, Republic of Korea
| | - Jong Eun Lee
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Sun Wook Han
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Sung Yong Kim
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Jeong Won Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam-gu, Cheonan, 31151, Republic of Korea.
| |
Collapse
|
25
|
Obeagu EI, Obeagu GU. Predictive models and biomarkers for survival in stage III breast cancer: a review of clinical applications and future directions. Ann Med Surg (Lond) 2024; 86:5980-5987. [PMID: 39359789 PMCID: PMC11444610 DOI: 10.1097/ms9.0000000000002517] [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: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Stage III breast cancer, characterized by locally advanced tumors and potential regional lymph node involvement, presents a formidable challenge to both patients and healthcare professionals. Accurate prediction of survival outcomes is crucial for guiding treatment decisions and optimizing patient care. This publication explores the potential clinical utility of predictive tools, encompassing genetic markers, imaging techniques, and clinical parameters, to improve survival outcome predictions in stage III breast cancer. Multimodal approaches, integrating these tools, hold the promise of delivering more precise and personalized predictions. Despite the inherent challenges, such as data standardization and genetic heterogeneity, the future offers opportunities for refinement, driven by precision medicine, artificial intelligence, and global collaboration. The goal is to empower healthcare providers to make informed treatment decisions, ultimately leading to improved survival outcomes and a brighter horizon for individuals facing this challenging disease.
Collapse
|
26
|
Duygulu ME, Aşık E, Tükenmez MA, Teoman G, Yıldırım A, Fidan E. Effect of Ki-67 proliferation index on survival in large cell neuroendocrine carcinoma of the lung. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240398. [PMID: 39292084 PMCID: PMC11404992 DOI: 10.1590/1806-9282.20240398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE Large cell neuroendocrine carcinoma of the lung is a rare type of lung cancer. There is a limited number of studies on clinical and histopathological characteristics that are effective in survival. The aim of this study was to investigate the relationship between histopathological and clinical characteristics, mainly Ki-67 proliferation index, and survival in patients diagnosed with large cell neuroendocrine carcinoma of the lung. METHODS The data of 38 patients followed up with the diagnosis of large cell neuroendocrine carcinoma of the lung were evaluated. The mean Ki-67 value was determined to be 65.8% (±20.8). The patients' clinical characteristics and survival times were compared according to the cut-off value determined for Ki-67 index. RESULTS When median overall survival times were compared, it was seen that overall survival was numerically lower in patients aged 65 years and over, in tumors located on the right side, in cases who were in the metastatic stage at diagnosis, whose Ki-67 index was 65% and above, who did not receive chemotherapy, who did not undergo curative surgery, and in patients with chronic diseases (p>0.05). In the Kaplan-Meier analysis, the median overall survival was determined to be 22.2 months (95%CI 21.7-22.7) in the patients with Ki-67<65%, while it was found to be 20.3 months (95%CI 4.5-36.2) in the patients with Ki-67≥65% (p=0.351). CONCLUSION Our study identified subgroups with decreased survival in large cell neuroendocrine carcinoma of lung patients. Studies including a larger number of patients are needed to identify the prognostic importance of these clinical and histopathological characteristics.
Collapse
Affiliation(s)
- Mustafa Emre Duygulu
- Karadeniz Technical University, Farabi Hospital, Department of Medical Oncology - Trabzon, Turkey
| | - Esra Aşık
- Karadeniz Technical University, Farabi Hospital, Department of Medical Oncology - Trabzon, Turkey
| | - Mehmet Akif Tükenmez
- Karadeniz Technical University, Farabi Hospital, Department of Medical Oncology - Trabzon, Turkey
| | - Gizem Teoman
- Karadeniz Technical University, Farabi Hospital, Department of Medical Pathology - Trabzon, Turkey
| | - Atila Yıldırım
- Kahramanmaraş Necip Fazıl City Hospital, Department of Medical Oncology - Kahramanmaraş, Turkey
| | - Evren Fidan
- Karadeniz Technical University, Farabi Hospital, Department of Medical Oncology - Trabzon, Turkey
| |
Collapse
|
27
|
Cai B, Cai T, Feng Z, Zhu H. The possible anti-tumor actions and mechanisms of active metabolites from Cortex Fraxini. Front Pharmacol 2024; 15:1404172. [PMID: 39346560 PMCID: PMC11427270 DOI: 10.3389/fphar.2024.1404172] [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: 03/20/2024] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Cortex Fraxini is a traditional Chinese herb that is widely available, inexpensive, and has low toxicity. Modern pharmacological studies have demonstrated that the active metabolites in Cortex Fraxini, including esculin, esculetin, and fraxetin, exert anti-tumor activities by regulating genes and proteins involved in cancer cell proliferation, apoptosis, invasion, and migration. Additionally, these metabolites play a pivotal role in the regulation of several tumor-associated signaling pathways, including the PI3K/Akt, MAPK/ERK, JAK/STAT3, and Wnt/β-catenin pathways. Due to their pro-apoptotic and anti-proliferative properties in vitro and in vivo, Cortex Fraxini and its active metabolites may be considered as potential candidates for the treatment of tumor. The aim of this review is to highlight the anti-tumor biological activities and underlying mechanisms of action of the active metabolites of Cortex Fraxini, with a view to providing a reference for their further development and application in the treatment of tumors.
Collapse
Affiliation(s)
- Bin Cai
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Ting Cai
- Department of Nephrology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Wuxi, China
| | - Zeyu Feng
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Huanhuan Zhu
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
28
|
Zhang C, Wang S, Lu X, Zhong W, Tang Y, Huang W, Wu F, Wang X, Wei W, Tang H. POP1 Facilitates Proliferation in Triple-Negative Breast Cancer via m6A-Dependent Degradation of CDKN1A mRNA. RESEARCH (WASHINGTON, D.C.) 2024; 7:0472. [PMID: 39268503 PMCID: PMC11391272 DOI: 10.34133/research.0472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
Triple-negative breast cancer (TNBC) is currently the worst prognostic subtype of breast cancer, and there is no effective treatment other than chemotherapy. Processing of precursors 1 (POP1) is the most substantially up-regulated RNA-binding protein (RBP) in TNBC. However, the role of POP1 in TNBC remains clarified. A series of molecular biological experiments in vitro and in vivo and clinical correlation analyses were conducted to clarify the biological function and regulatory mechanism of POP1 in TNBC. Here, we identified that POP1 is significantly up-regulated in TNBC and associated with poor prognosis. We further demonstrate that POP1 promotes the cell cycle and proliferation of TNBC in vitro and vivo. Mechanistically, POP1 directly binds to the coding sequence (CDS) region of CDKN1A mRNA and degrades it. The degradation process depends on the N6-methyladenosine (m6A) modification at the 497th site of CDKN1A and the recognition of this modification by YTH N6-methyladenosine RNA binding protein 2 (YTHDF2). Moreover, the m6A inhibitor STM2457 potently impaired the proliferation of POP1-overexpressed TNBC cells and improved the sensitivity to paclitaxel. In summary, our findings reveal the pivotal role of POP1 in promoting TNBC proliferation by degrading the mRNA of CDKN1A and that inhibition of m6A with STM2457 is a promising therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Chao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sifen Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuqing Lu
- Department of Breast Surgery, Zhongshan City People's Hospital, ZhongShan, China
| | - Wenjing Zhong
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Yunyun Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangzhou Kangda Vocational Technical College, Guangzhou 510700, China
| | - Weiling Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fengjia Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiumei Wang
- Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010020, Inner Mongolia, China
| | - Weidong Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
29
|
Hu B, Xu Y, Gong H, Tang L, Li H. Radiomics Analysis of Intratumoral and Various Peritumoral Regions From Automated Breast Volume Scanning for Accurate Ki-67 Prediction in Breast Cancer Using Machine Learning. Acad Radiol 2024:S1076-6332(24)00603-2. [PMID: 39256084 DOI: 10.1016/j.acra.2024.08.040] [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: 07/13/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
RATIONALE AND OBJECTIVES Current radiomics research primarily focuses on intratumoral regions and fixed peritumoral areas, lacking optimization for accurate Ki-67 prediction. This study aimed to develop machine learning (ML) models to analyze radiomic features from Automated Breast Volume Scanning (ABVS) images of different peritumoral region sizes to identify the optimal size for accurate preoperative Ki-67 prediction. MATERIALS AND METHODS A total of 668 breast cancer patients were enrolled and divided into training (486) and testing (182) cohorts. In the training cohort, ML models were developed for intratumoral and peritumoral regions (2, 4, 6, 8, and 10 mm). Relevant Ki-67 features for each ROI were identified, and different models were compared to determine the optimal one. These models were validated using a testing cohort to find the most accurate peritumoral region for Ki-67 prediction. SHAP (Shapley Additive Explanations) analysis was performed to identify key radiomic features from the optimal model. RESULTS The Extreme Gradient Boosting (XGBoost) model for the intratumoral region combined with the 6 mm peritumoral region achieved the highest predictive accuracy, with an AUC of 0.957 in the training cohort and 0.920 in the testing cohort. Calibration curves confirmed reliability, and decision curve analysis demonstrated the highest net benefit. SHAP indicated that 6 mm peritumoral radiomic features are more significant than intratumoral features. CONCLUSION The XGBoost model using ABVS-derived radiomic features from both the intratumoral and 6 mm peritumoral regions provides the most accurate preoperative Ki-67 prediction.
Collapse
Affiliation(s)
- Bin Hu
- Department of Ultrasound, Minhang Hospital, Fudan University, Shanghai, China (B.H., H.G., L.T.).
| | - Yanjun Xu
- Department of Ultrasonography, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.X.); Shanghai Institute of Ultrasound in Medicine, Shanghai, China (Y.X.)
| | - Huiling Gong
- Department of Ultrasound, Minhang Hospital, Fudan University, Shanghai, China (B.H., H.G., L.T.)
| | - Lang Tang
- Department of Ultrasound, Minhang Hospital, Fudan University, Shanghai, China (B.H., H.G., L.T.)
| | - Hongchang Li
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China (H.L.)
| |
Collapse
|
30
|
Banerjee R, Maitra I, Bhattacharya T, Banerjee M, Ramanathan G, Rayala SK, Venkatraman G, Rajeswari D. Next-generation biomarkers for prognostic and potential therapeutic enhancement in Triple negative breast cancer. Crit Rev Oncol Hematol 2024; 201:104417. [PMID: 38901639 DOI: 10.1016/j.critrevonc.2024.104417] [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: 05/01/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Triple-negative breast carcinoma (TNBC) is one of the most challenging subtypes of breast carcinoma and it has very limited therapeutic options as it is highly aggressive. The prognostic biomarkers are crucial for early diagnosis of the tumor, it also helps in anticipating the trajectory of the illness and optimizing the therapy options. Several therapeutic biomarkers are being used. Among them, the next-generation biomarkers that include Circulating tumor (ct) DNA, glycogen, lipid, and exosome biomarkers provide intriguing opportunities for enhancing the prognosis of TNBC. Lipid and glycogen biomarkers serve as essential details on the development of the tumor along with the efficacy of the treatment, as it exhibits metabolic alteration linked to TNBC. Several types of biomarkers have predictive abilities in TNBC. Elevated levels are associated with worse outcomes. ctDNA being a noninvasive biomarker reveals the genetic composition of the tumor, as well as helps to monitor the progression of the disease. Traditional therapies are ineffective in TNBC due to a lack of receptors, targeted drug delivery provides a tailored approach to overcome drug resistance and site-specific action by minimizing the side effects in TNBC treatment. This enhances therapeutic outcomes against the aggressive nature of breast cancer. This paper includes all the recent biomarkers which has been researched so far in TNBC and the state of art for TNBC which is explored.
Collapse
Affiliation(s)
- Risav Banerjee
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Indrajit Maitra
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Trisha Bhattacharya
- Department of Biotechnology, Indian Academy Degree College, Autonomous, Hennur cross, Kalyan Nagar, Bengaluru, Karnataka 560043, India
| | - Manosi Banerjee
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology, Madras, Tamil Nadu 600036, India
| | - Ganesh Venkatraman
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Devi Rajeswari
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
31
|
Zhang L, Shen M, Zhang D, He X, Du Q, Liu N, Huang X. Radiomics Nomogram Based on Dual-Sequence MRI for Assessing Ki-67 Expression in Breast Cancer. J Magn Reson Imaging 2024; 60:1203-1212. [PMID: 38088478 DOI: 10.1002/jmri.29149] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Radiomics has been extensively applied in predicting Ki-67 in breast cancer (BC). However, this is often confined to the exploration of a single sequence, without considering the varying sensitivity and specificity among different sequences. PURPOSE To develop a nomogram based on dual-sequence MRI derived radiomic features combined with clinical characteristics for assessing Ki-67 expression in BC. STUDY TYPE Retrospective. POPULATION 227 females (average age, 51 years) with 233 lesions and pathologically confirmed BC, which were divided into the training set (n = 163) and test set (n = 70). FIELD STRENGTH/SEQUENCE 3.0-T, T1-weighted dynamic contrast-enhanced MRI (DCE-MRI) and apparent diffusion coefficient (ADC) maps from diffusion-weighted MRI (EPI sequence). ASSESSMENT The regions of interest were manually delineated on ADC and DCE-MRI sequences. Three radiomics models of ADC, DCE-MRI, and dsMRI (combined ADC and DCE-MRI sequences) were constructed by logistic regression and the radiomics score (Radscore) of the best model was calculated. The correlation between Ki-67 expression and clinical characteristics such as receptor status, axillary lymph node (ALN) metastasis status, ADC value, and time signal intensity curve was analyzed, and the clinical model was established. The Radscore was combined with clinical predictors to construct a nomogram. STATISTICAL TESTS The independent sample t-test, Mann-Whitney U test, Chi-squared test, Interclass correlation coefficients (ICCs), single factor analysis, least absolute shrinkage and selection operator (LASSO), logistic regression, receiver operating characteristics, Delong test, Hosmer_Lemeshow test, calibration curve, decision curve. A P-value <0.05 was considered statistically significant. RESULTS In the test set, the prediction efficiency of the dsMRI model (AUC = 0.862) was higher than ADC model (AUC = 0.797) and DCE-MRI model (AUC = 0.755). With the inclusion of estrogen receptor (ER) and ALN metastasis, the nomogram displayed quality improvement (AUC = 0.876), which was superior to the clinical model (AUC = 0.787) and radiomics model. DATA CONCLUSION The nomogram based on dsMRI radiomic features and clinical characteristics may be able to assess Ki-67 expression in BC. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Li Zhang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Mengyi Shen
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Dingyi Zhang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xin He
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qinglin Du
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Nian Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaohua Huang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
32
|
Ferriere F, Percevault F, Plu N, Le Page Y, Pham TH, Lecomte S, Costet N, Surel C, Efstathiou T, Pakdel F. Natural glyceollin soybean extracts elicited with Aspergillus sojae reduce estrogen-dependent breast cancer growth in orally fed mice. J Food Sci 2024; 89:5951-5966. [PMID: 39150682 DOI: 10.1111/1750-3841.17293] [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: 04/02/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Previous studies have demonstrated antiestrogenic and antiproliferative effects of these molecules in breast cancer cells. Notably, we have reported that pure synthetic glyceollins I and II act through various pathways, including ERα, FOXM1, AhR, and HIF pathways to inhibit cell proliferation and migration. In this study, the potential antitumor activity of glyceollins enriched in crude soybean extracts, obtained by solid fermentation with Aspergillus sojae, was investigated in vivo on MCF-7 breast cancer cells implanted in the chorioallantoic membrane of the chick egg and on ovariectomized nude mice. The first trial showed a substantial reduction in the migration of MCF-7 cells treated with the natural extracts. However, the natural extracts significantly reduced the estrogen-dependent growth of transplanted tumors in orally fed nude mice. Our results showed that natural soybean extracts slightly but significantly reduced estrogen-dependent growth of the transplanted tumors in orally fed nude mice. These results were confirmed by immunohistochemistry of Ki-67 and histone H3S10 phosphorylation (H3S10P), revealing lower expression of these proliferation markers in the transplanted tumors from mice fed with the fermented extracts. Additionally, compared to the control animals, we observed a lower expression of angiogenesis markers such as CD31 and CD34. Surprisingly, transcriptomic analysis of RNA from transplanted MCF-7 cells revealed no differential gene expression. These results may suggest that orally consumed natural glyceollins exert biological effects throughout the body, acting indirectly to reduce tumor angiogenesis and consequently tumor volume. Overall, our results indicate that glyceollins, elicited components of the soy origin, hold potential therapeutic applications for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- François Ferriere
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Frederic Percevault
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Nicolas Plu
- Olga, Pôle Développement, Noyal-sur-Vilaine, France
| | - Yann Le Page
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Tu-Ha Pham
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Sylvain Lecomte
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Nathalie Costet
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Claire Surel
- Olga, Pôle Développement, Noyal-sur-Vilaine, France
| | | | - Farzad Pakdel
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| |
Collapse
|
33
|
Sato N, Tsujimoto M, Nakatsuji M, Tsuji H, Sugama Y, Shimazu K, Shimoda M, Ishihara H. Flow cytometric analysis for Ki67 assessment in formalin-fixed paraffin-embedded breast cancer tissue. BMC Biol 2024; 22:181. [PMID: 39183273 PMCID: PMC11346000 DOI: 10.1186/s12915-024-01980-4] [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: 08/20/2023] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Pathologists commonly employ the Ki67 immunohistochemistry labelling index (LI) when deciding appropriate therapeutic strategies for patients with breast cancer. However, despite several attempts at standardizing the Ki67 LI, inter-observer and inter-laboratory bias remain problematic. We developed a flow cytometric assay that employed tissue dissociation, enzymatic treatment and a gating process to analyse Ki67 in formalin-fixed paraffin-embedded (FFPE) breast cancer tissue. RESULTS We demonstrated that mechanical homogenizations combined with thrombin treatment can be used to recover efficiently intact single-cell nuclei from FFPE breast cancer tissue. Ki67 in the recovered cell nuclei retained reactivity against the MIB-1 antibody, which has been widely used in clinical settings. Additionally, since the method did not alter the nucleoskeletal structure of tissues, the nuclei of cancer cells can be enriched in data analysis based on differences in size and complexity of nuclei of lymphocytes and normal mammary cells. In a clinical study using the developed protocol, Ki67 positivity was correlated with the Ki67 LI obtained by hot spot analysis by a pathologist in Japan (rho = 0.756, P < 0.0001). The number of cancer cell nuclei subjected to the analysis in our assay was more than twice the number routinely checked by pathologists in clinical settings. CONCLUSIONS The findings of this study showed the application of this new flow cytometry method could potentially be used to standardize Ki67 assessments in breast cancer.
Collapse
Affiliation(s)
- Natsuki Sato
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
| | - Masahiko Tsujimoto
- Department of Diagnostic Pathology, Daini Osaka Police Hospital, 2-6-40 Karasugatsuji, Tennoji-Ku, Osaka, 543-8922, Japan
- Present Address: Osaka Pathology and Cytology Laboratory, 2-2-26 Kunijima, Higashiyodogawa-Ku, Osaka, 533-0024, Japan
| | - Masatoshi Nakatsuji
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
- Department of Pathobiochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Hiromi Tsuji
- Department of Diagnostic Pathology, Osaka Police Hospital, 10-31 Kitayamacho, Tennoji-Ku, Osaka, Japan
| | - Yuji Sugama
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hideki Ishihara
- Nitto Boseki Co., Ltd, 2-4-1, Kojimachi, Chiyoda-ku, Tokyo, 102-8489, Japan.
- Department of Research Support, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka, 567-0085, Japan.
| |
Collapse
|
34
|
de Moraes FCA, Souza MEC, Sano VKT, Moraes RA, Melo AC. Association of tumor-infiltrating lymphocytes with clinical outcomes in patients with triple-negative breast cancer receiving neoadjuvant chemotherapy: a systematic review and meta-analysis. Clin Transl Oncol 2024:10.1007/s12094-024-03661-8. [PMID: 39154313 DOI: 10.1007/s12094-024-03661-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVE Triple-negative breast cancer (TNBC) presents a clinical challenge as an aggressive tumor, correlated with unfavorable prognosis. Tumor-infiltrating lymphocytes (TILs) have garnered interest as a potential prognostic biomarker. However, the disparity in outcomes between varying TILs rates remains inadequately explored. METHODS PubMed, Scopus, Web of Science, and Cochrane databases were searched for studies about the prognostic value of TILs in patients with TNBC receiving neoadjuvant chemotherapy. The hazard ratios (HRs) or odds ratios (ORs) were computed for binary endpoints, with 95% confidence intervals (CIs). RESULTS Twenty-nine studies were included, involving a population of six thousand one hundred sixty-one (80.41%) with TNBC. The cut-off TILs value ranged from 10 to 60%, with 50% being the most related value. Compared with the low-TIL expression group, the disease-free survival (DFS) (HR 0.71; 95% CI 0.61-0.82; p < 0.00001) and overall survival (OS) (HR 0.76; 95% CI 0.63-0.90; p = 0.002) rates showed significant improvement with higher TIL infiltrations. In the subgroup analyses of the lymphocyte subtypes CD4 + and CD8 + , there was statistical significance favoring higher TILs rates in both subtypes, each associated with improved DFS (HR 0.48; 95% CI 0.33-0.71; p = 0.0002) and OS (HR 0.53; 95% CI 0.36-0.78; p = 0.001), regardless of which cell subtype was predominantly infiltrated. The complete pathological response analysis showed better rates for the higher TIL group than the control for both the TIL (OR 1.29; 95% CI 1.13-1.48; p = 0.0003) and Ki-67 (OR 2.74; 95% CI 2.01-3.73; p < 0.00001) analyses. CONCLUSION Higher expressions of TILs in patients with TNBC were associated with improved significantly DFS, OS, and pCR outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Ana C Melo
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
35
|
Burciu OM, Sas I, Popoiu TA, Merce AG, Moleriu L, Cobec IM. Correlations of Imaging and Therapy in Breast Cancer Based on Molecular Patterns: An Important Issue in the Diagnosis of Breast Cancer. Int J Mol Sci 2024; 25:8506. [PMID: 39126074 PMCID: PMC11312504 DOI: 10.3390/ijms25158506] [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: 06/08/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Breast cancer is a global health issue affecting countries worldwide, imposing a significant economic burden due to expensive treatments and medical procedures, given the increasing incidence. In this review, our focus is on exploring the distinct imaging features of known molecular subtypes of breast cancer, underlining correlations observed in clinical practice and reported in recent studies. The imaging investigations used for assessment include screening modalities such as mammography and ultrasonography, as well as more complex investigations like MRI, which offers high sensitivity for loco-regional evaluation, and PET, which determines tumor metabolic activity using radioactive tracers. The purpose of this review is to provide a better understanding as well as a revision of the imaging differences exhibited by the molecular subtypes and histopathological types of breast cancer.
Collapse
Affiliation(s)
- Oana Maria Burciu
- Doctoral School, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
- Department of Functional Sciences, Medical Informatics and Biostatistics Discipline, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ioan Sas
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Tudor-Alexandru Popoiu
- Department of Functional Sciences, Medical Informatics and Biostatistics Discipline, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adrian-Grigore Merce
- Department of Cardiology, Institute of Cardiovascular Diseases, 300310 Timisoara, Romania
| | - Lavinia Moleriu
- Department of Functional Sciences, Medical Informatics and Biostatistics Discipline, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ionut Marcel Cobec
- Clinic of Obstetrics and Gynecology, Klinikum Freudenstadt, 72250 Freudenstadt, Germany
| |
Collapse
|
36
|
Zhao Y. A novel mutation in PTEN in anaplastic thyroid carcinoma: A case report. Biomed Rep 2024; 21:127. [PMID: 39006510 PMCID: PMC11240280 DOI: 10.3892/br.2024.1815] [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: 02/25/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Anaplastic thyroid cancer (ATC) is a rare disease with a poor prognosis and accounts for a high proportion of thyroid cancer deaths. The present study reported on a 56-year-old male patient with ATC and examined the clinical manifestations, pathological features, differential diagnosis and genetic mutations. Immunohistochemical analysis showed positivity for vimentin, Ki-67 and cytokeratin in the tumor specimen. In addition, pathological mitotic figures of tumor cells and intra-lymph node metastasis were observed. Genetic analysis revealed the presence of a novel mutation (c.388C>T, p.R130X) in exon 5 of the phosphatase and tensin homolog (PTEN) gene, which was first detected in ATC. Gene conservation analysis showed that R130 is a highly conserved amino acid. Protein structure model predictions implied that p.R130X mutation results in a severe defect of the C2 domain and the TAD domain of PTEN, which may be a reason for the high malignancy of the tumor. The present case report highlights a novel mutation of PTEN in ATC, which expands the molecular spectrum of PTEN and further underlines the importance of PTEN.
Collapse
Affiliation(s)
- Yanli Zhao
- Department of Pathology, Linfen Central Hospital, Linfen, Shanxi 041099, P.R. China
| |
Collapse
|
37
|
Moraru L, Mitranovici MI, Moraru R, Voidazan S, Munteanu M, Georgescu R, Costachescu D, Turdean SG. Combining Molecular and Traditional Prognostic Factors: A Holistic Approach to Breast Cancer Prognostication. Diagnostics (Basel) 2024; 14:1449. [PMID: 39001339 PMCID: PMC11241232 DOI: 10.3390/diagnostics14131449] [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: 05/21/2024] [Revised: 06/20/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Breast cancer is a heterogeneous disease with various morphologies and molecular features, and it is the second leading cause of cancer death in women in developed countries. According to the literature, we currently lack both prognostic biomarkers and therapeutic targets. The most important prognostic factors are disease stage and Nottingham grade. We conducted a retrospective analysis involving 273 patients with BC who underwent neoadjuvant therapy before proceeding to curative surgical treatment between 1 January 2014 and 31 December 2023. Pathological procedures were conducted at the Department of Pathology, Emergency County Hospital of Targu Mureș, Romania. A statistical analysis was performed. Regarding the relationship between Nottingham grade and Ki67, grade I was associated with a Ki67 of less than 14. The relationship between tumor grade and luminal was similar (p = 0.0001): Grade I was associated with luminal A. Regarding TNM stage, it was statistically significantly correlated with TILs (p = 0.01) and RCB (p = 0.0001). Stages III and IV were associated with a high RCB and poor prognosis. Regarding the prognostic value, Nottingham grade 3 and TNM stages III and IV were correlated with low overall survival and disease-free survival, with poor prognosis, and, among the molecular variables, RCB played the most important prognostic role.
Collapse
Affiliation(s)
- Liviu Moraru
- Department of Anatomy, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Melinda Ildiko Mitranovici
- Department of Obstetrics and Gynecology, Emergency County Hospital Hunedoara, 14 Victoriei Street, 331057 Hunedoara, Romania
| | - Raluca Moraru
- Faculty of Medicine, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Septimiu Voidazan
- Department of Epidemiology, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Mihai Munteanu
- Faculty of Electrical Engineering, Technical University, George Baritiu Street, 400394 Cluj Napoca, Romania
| | - Rares Georgescu
- Department of Surgery, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Dan Costachescu
- Department of Orthopedisc-Traumatology, Urology, Radiology and Medical Imaging, University of Medicine and Pharmacy Victor Babes, Square Eftimie Murgu, 300041 Timisoara, Romania
| | - Sabin Gligore Turdean
- Department of Pathology, County Clinical Hospital of Targu Mures, 540072 Targu Mures, Romania
| |
Collapse
|
38
|
Rais G, Mokfi R, Boutaggount F, Maskrout M, Bennour S, Senoussi C, Rais F. Assessment of the Predictive Role of Ki-67 in Breast Cancer Patients' Responses to Neoadjuvant Chemotherapy. Eur J Breast Health 2024; 20:199-206. [PMID: 39257012 PMCID: PMC11589294 DOI: 10.4274/ejbh.galenos.2024.2024-3-8] [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: 03/26/2024] [Accepted: 06/02/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE Neoadjuvant chemotherapy (NAC) in breast cancer (BC) is being considered for a broader range of cases, including locally advanced tumors and situations where downstaging could reduce extensive surgery. Several trials have explored predictive markers of pathological complete response (pCR). The role of Ki-67 as a predictor of pCR has been demonstrated in studies. However, the cut-off remains vague, given the lack of standardization of measurement methods. The aim of our study was to evaluate the predictive value of Ki-67 in response to NAC and to identify the cut-off values that exhibit the strongest correlation with best response. MATERIALS AND METHODS This retrospective study included 187 patients who had undergone surgery following NAC for BC at the CHU Souss Massa of Agadir between January 2020 and January 2023. Logistic regression was used to assess the correlation between Ki-67 and patients' characteristics. Optimal Ki-67 cutoff was identified by receiver operating characteristic curve. Kaplan-Meier curves were used to assess disease-free survival (DFS), and survival comparisons were assessed with the log-rank test. RESULTS The median age was 51.8±10.7 years and 51.4% of tumors were smaller than 5 cm. Node invasion was found in 55.4%. Luminal B subtype was found in 49.7%, followed by human epidermal growth factor receptor-2 (HER-2)-positive in 27.4%, triple-negative in 14.3% and Luminal A in 8.6%. pCR occurred in 40% of patients overall. Subgroup analysis revealed a significant association between pCR and tumor size (p<0.001), lymph node involvement (p<0.001), grade 2 (p<0.001), vascular invasion (p<0.001), and positive HER-2 status (p = 0.022). In statistical analysis, pathological responses were improved in patients with Ki-67 >35% (p<0.001). DFS was 98.8% at 12 months. No statistical difference was found in DFS according to Ki-67 values and pCR status. CONCLUSION Our results indicate that Ki-67 is a predictive marker for response in the neoadjuvant setting in BC patients. Our study showed that a Ki-67 cut-off >35% predicts a better pCR rate in response to NAC. However, this cutoff value remains controversial due to the absence of a standard method of measurement, with inter- and intra-observer variability. It would be necessary to validate this cutoff in other studies.
Collapse
Affiliation(s)
- Ghizlane Rais
- Department of Medical Oncology, CHU Souss Massa, Biomed Laboratory, University Ibn Zohr Agadir Faculty of Medicine and Pharmacy of Agadir, Agadir, Morocco
| | - Rania Mokfi
- Department of Medical Oncology, CHU Souss Massa, University Ibn Zohr Agadir Faculty of Medicine and Pharmacy of Agadir, Agadir, Morocco
| | - Farah Boutaggount
- Department of Medical Oncology, CHU Souss Massa, University Ibn Zohr Agadir Faculty of Medicine and Pharmacy of Agadir, Agadir, Morocco
| | - Meryem Maskrout
- Department of Medical Oncology, CHU Souss Massa, University Ibn Zohr Agadir Faculty of Medicine and Pharmacy of Agadir, Agadir, Morocco
| | - Soundouss Bennour
- Department of Medical Oncology, CHU Souss Massa, University Ibn Zohr Agadir Faculty of Medicine and Pharmacy of Agadir, Agadir, Morocco
| | - Chaymae Senoussi
- Department of Medical Oncology, CHU Souss Massa, University Ibn Zohr Agadir Faculty of Medicine and Pharmacy of Agadir, Agadir, Morocco
| | - Fadoua Rais
- Department of Radiation Therapy, University Hospital Center of Montreal, Montreal, Canada
| |
Collapse
|
39
|
Li F, Zhu TW, Lin M, Zhang XT, Zhang YL, Zhou AL, Huang DY. Enhancing Ki-67 Prediction in Breast Cancer: Integrating Intratumoral and Peritumoral Radiomics From Automated Breast Ultrasound via Machine Learning. Acad Radiol 2024; 31:2663-2673. [PMID: 38182442 DOI: 10.1016/j.acra.2023.12.036] [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: 11/16/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/07/2024]
Abstract
RATIONALE AND OBJECTIVES Traditional Ki-67 evaluation in breast cancer (BC) via core needle biopsy is limited by repeatability and heterogeneity. The automated breast ultrasound system (ABUS) offers reproducibility but is constrained to morphological and echoic assessments. Radiomics and machine learning (ML) offer solutions, but their integration for improving Ki-67 predictive accuracy in BC remains unexplored. This study aims to enhance ABUS by integrating ML-assisted radiomics for Ki-67 prediction in BC, with a focus on both intratumoral and peritumoral regions. MATERIALS AND METHODS A retrospective analysis was conducted on 936 BC patients, split into training (n = 655) and testing (n = 281) cohorts. Radiomics features were extracted from intra- and peritumoral regions via ABUS. Feature selection involved Z-score normalization, intraclass correlation, Wilcoxon rank sum tests, minimum redundancy maximum relevance, and least absolute shrinkage and selection operator logistic regression. ML classifiers were trained and optimized for enhanced predictive accuracy. The interpretability of the optimized model was further augmented by employing Shapley additive explanations (SHAP). RESULTS Of the 2632 radiomics features in each patient, 15 were significantly associated with Ki-67 levels. The support vector machine (SVM) was identified as the optimal classifier, with area under the receiver operating characteristic curve values of 0.868 (training) and 0.822 (testing). SHAP analysis indicated that five peritumoral and two intratumoral features, along with age and lymph node status, were key determinants in the predictive model. CONCLUSION Integrating ML with ABUS-based radiomics effectively enhances Ki-67 prediction in BC, demonstrating the SVM model's strong performance with both radiomics and clinical factors.
Collapse
Affiliation(s)
- Fang Li
- Department of Ultrasound, The People's Hospital of Yuhuan, No. 18, Changle Rd, Yuhuan 317600, Zhejiang, China (F.L., X.Z., Y.Z., A.Z., D.H.)
| | - Tong-Wei Zhu
- Department of Ultrasound, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang, China (T.Z.)
| | - Miao Lin
- Second Department of General Surgery, The People's Hospital of Yuhuan, Yuhuan, Zhejiang, China (M.L.)
| | - Xiao-Ting Zhang
- Department of Ultrasound, The People's Hospital of Yuhuan, No. 18, Changle Rd, Yuhuan 317600, Zhejiang, China (F.L., X.Z., Y.Z., A.Z., D.H.)
| | - Ya-Li Zhang
- Department of Ultrasound, The People's Hospital of Yuhuan, No. 18, Changle Rd, Yuhuan 317600, Zhejiang, China (F.L., X.Z., Y.Z., A.Z., D.H.)
| | - Ai-Li Zhou
- Department of Ultrasound, The People's Hospital of Yuhuan, No. 18, Changle Rd, Yuhuan 317600, Zhejiang, China (F.L., X.Z., Y.Z., A.Z., D.H.)
| | - De-Yi Huang
- Department of Ultrasound, The People's Hospital of Yuhuan, No. 18, Changle Rd, Yuhuan 317600, Zhejiang, China (F.L., X.Z., Y.Z., A.Z., D.H.).
| |
Collapse
|
40
|
Lin JY, Ye JY, Chen JG, Lin ST, Lin S, Cai SQ. Prediction of Receptor Status in Radiomics: Recent Advances in Breast Cancer Research. Acad Radiol 2024; 31:3004-3014. [PMID: 38151383 DOI: 10.1016/j.acra.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 12/29/2023]
Abstract
Breast cancer is a multifactorial heterogeneous disease and the leading cause of cancer-related deaths in women; its diagnosis and treatment require clinical sensitivity and a comprehensive disciplinary research approach. The expression of different receptors on tumor cells not only provides the basis for molecular typing of breast cancer but also has a decisive role in the diagnosis, treatment, and prognosis of breast cancer. To date, immunohistochemistry (IHC), which uses invasive histological sampling, has been extensively used in clinical practice to analyze the status of receptors and to make an accurate diagnosis of breast cancer. As an invasive assay, IHC can provide important biological information on tumors at a single point in time, but cannot predict future changes (due to treatment or tumor mutations) without additional invasive procedures. These issues highlight the need to develop a non-invasive method for predicting receptor status. The emerging field of radiomics may offer a non-invasive approach to identification of receptor status without requiring biopsy. In this paper, we present a review of the latest research results in radiomics for predicting the status of breast cancer receptors, with potential important clinical applications.
Collapse
Affiliation(s)
- Jun-Yuan Lin
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Jia-Yi Ye
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Jin-Guo Chen
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Shu-Ting Lin
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.)
| | - Shu Lin
- Center of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.Y., J.G.C., S.T.L., S.L.); Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia (S.L.)
| | - Si-Qing Cai
- Department of Radiology, the Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian Province, China (J.Y.L., S.Q.C.).
| |
Collapse
|
41
|
Joung H, Liu H. 2‑D08 mediates notable anticancer effects through multiple cellular pathways in uterine leiomyosarcoma cells. Oncol Rep 2024; 52:97. [PMID: 38874019 PMCID: PMC11200159 DOI: 10.3892/or.2024.8756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
2',3',4'‑trihydroxyflavone (2‑D08), a SUMO E2 inhibitor, has several biological functions, including anticancer activity, but its effects on uterine leiomyosarcoma (Ut‑LMS) are unknown. The anticancer activity of 2‑D08 was explored in an in vitro model using SK‑LMS‑1 and SK‑UT‑1B cells (human Ut‑LMS cells). Treatment with 2‑D08 inhibited cell viability in a dose‑ and time‑dependent manner and significantly inhibited the colony‑forming ability of Ut‑LMS cells. In SK‑UT‑1B cells treated with 2‑D08, flow cytometric analysis revealed a slight increase in apoptotic rates, while cell cycle progression remained unaffected. Western blotting revealed elevated levels of RIP1, indicating induction of necrosis, but LC3B levels remained unchanged, suggesting no effect on autophagy. A lactate dehydrogenase (LDH) assay confirmed increased LDH release, further supporting the induction of apoptosis and necrosis by 2‑D08 in SK‑UT‑1B cells. 2‑D08‑induced production of reactive oxygen species and apoptosis progression were observed in SK‑LMS‑1 cells. Using Ki67 staining and bromodeoxyuridine assays, it was found that 2‑D08 suppressed proliferation in SK‑LMS‑1 cells, while treatment for 48 h led to cell‑cycle arrest. 2‑D08 upregulated p21 protein expression in SK‑LMS‑1 cells and promoted apoptosis through caspase‑3. Evaluation of α‑SM‑actin, calponin 1 and TAGLN expression indicated that 2‑D08 did not directly initiate smooth muscle phenotypic switching in SK‑LMS‑1 cells. Transcriptome analysis on 2‑D08‑treated SK‑LMS‑1 cells identified significant differences in gene expression and suggested that 2‑D08 modulates cell‑cycle‑ and apoptosis‑related pathways. The analysis identified several differentially expressed genes and significant enrichment for biological processes related to DNA replication and molecular functions associated with the apoptotic process. It was concluded that 2‑D08 exerts antitumor effects in Ut‑LMS cells by modulating multiple signaling pathways and that 2‑D08 may be a promising candidate for the treatment of human Ut‑LMS. The present study expanded and developed knowledge regarding Ut‑LMS management and indicated that 2‑D08 represents a notable finding in the exploration of fresh treatment options for such cancerous tumors.
Collapse
Affiliation(s)
- Hosouk Joung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Hwasun, Jeonnam 58128, Republic of Korea
| | - Hyunju Liu
- Department of Obstetrics and Gynecology, Chosun University College of Medicine, Gwangju 61452, Republic of Korea
- Department of Obstetrics and Gynecology, Chosun University Hospital, Gwangju 61453, Republic of Korea
| |
Collapse
|
42
|
Martín-Sanz R, Rodrigues-Françoso A, García-Mesa Y, García-Alonso FJ, Gómez-Muñoz MA, Malmierca-González S, Salazar-Blázquez R, García-Suárez O, Feito J. Prognostic Evaluation of Piezo2 Channels in Mammary Gland Carcinoma. Cancers (Basel) 2024; 16:2413. [PMID: 39001475 PMCID: PMC11240440 DOI: 10.3390/cancers16132413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
In the last decade, a group of Ca2+ channels called Piezo were discovered, demonstrating a decisive role in the cellular response to mechanical stimuli and being essential in the biological behavior of cells regarding the extracellular compartment. Several investigations have suggested a potential role in carcinogenesis, with a tumor suppressor role in some cases but increased expression in several high-grade neoplasms. Regarding Piezo2 expression in mammary gland neoplasms, a protective role for Piezo2 was initially suggested, but a subsequent study demonstrated a relationship between Piezo2 expression and the highly aggressive triple-negative phenotype of breast carcinoma. A cohort of 125 patients with clinical follow-up was chosen to study Piezo2 expression and clarify its clinical implications using the same immunohistochemical evaluation performed for other breast carcinoma parameters. Fisher's exact test was chosen to identify potential relationships between the different variables. A significant association was found with the Ki67 proliferation index, but not with mitoses. The tendency of most proliferative tumors was to have an increased score for Piezo2. A similar association was found between Piezo2 expression and perineural invasion.
Collapse
Affiliation(s)
- Raquel Martín-Sanz
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (R.M.-S.); (S.M.-G.)
- Servicio de Oftalmología, Complejo Asistencial de Zamora, 49022 Zamora, Spain
| | | | - Yolanda García-Mesa
- Grupo SINPOS, Department of Cell Biology and Morphology, University of Oviedo, 33003 Oviedo, Spain; (Y.G.-M.); (O.G.-S.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | | | - María Asunción Gómez-Muñoz
- Servicio de Anatomía Patológica, Complejo Asistencial Universitario de Salamanca, 37007 Salamanca, Spain; (M.A.G.-M.); (R.S.-B.)
| | - Sandra Malmierca-González
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (R.M.-S.); (S.M.-G.)
- Servicio de Anatomía Patológica, Complejo Asistencial Universitario de Salamanca, 37007 Salamanca, Spain; (M.A.G.-M.); (R.S.-B.)
| | - Rocío Salazar-Blázquez
- Servicio de Anatomía Patológica, Complejo Asistencial Universitario de Salamanca, 37007 Salamanca, Spain; (M.A.G.-M.); (R.S.-B.)
| | - Olivia García-Suárez
- Grupo SINPOS, Department of Cell Biology and Morphology, University of Oviedo, 33003 Oviedo, Spain; (Y.G.-M.); (O.G.-S.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jorge Feito
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (R.M.-S.); (S.M.-G.)
- Servicio de Anatomía Patológica, Complejo Asistencial Universitario de Salamanca, 37007 Salamanca, Spain; (M.A.G.-M.); (R.S.-B.)
| |
Collapse
|
43
|
Martins SA, Costa RR, Brito A, Reis RL, Alves NM, Pashkuleva I, Soares da Costa D. Multifunctional calcium-based nanocarriers for synergistic treatment of triple-negative breast cancer. J Colloid Interface Sci 2024; 674:500-512. [PMID: 38943911 DOI: 10.1016/j.jcis.2024.06.159] [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: 04/08/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Targeted breast cancer therapies hold the potential to improve the efficiency of drug delivery to the pathology site without impacting the viability and function of healthy cells. Herein, we developed multifunctional nanocarriers that target simultaneously several downstream signaling processes in triple negative breast cancer cells. The system comprises pH sensitive CaCO3 nanoparticles (NPs) as carriers of the anticancer drug doxorubicin (DOX). The NPs were coated in a layer-by-layer (LbL) fashion using poly-l-lysine and hyaluronic acid to target receptors overexpressed in breast cancer (e.g. CD44, RHAMM). Spheroids of the triple-negative Hs578T cell line were used as a 3D model to assess the therapeutic potential of this system. Our results showed that the NPs act via a synergistic mechanism that combines Ca2+ overload causing cell calcification and DNA damage by DOX. The LbL coating was crucial for the protection of the healthy cells, i.e. it provides NPs with targeting capacity. The overall data suggests that the LbL-coated NPs loaded with DOX hold great potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Sara A Martins
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Rui R Costa
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Alexandra Brito
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Natália M Alves
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
44
|
Smith AA, Vesey A, Helfrich C, Pasternak JA. Late gestation fetal hypothyroidism alters cell cycle regulation across multiple organ systems. BMC Vet Res 2024; 20:268. [PMID: 38902754 PMCID: PMC11188211 DOI: 10.1186/s12917-024-04102-y] [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: 12/21/2023] [Accepted: 05/29/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Hypothyroidism is a common endocrine disruption observed in utero that adversely affects fetal growth and maturation leading to long-term impacts on health; however, the exact molecular mechanisms by which these deleterious effects occur are unknown. We hypothesize that fetal hypothyroidism during late gestation will disrupt cell cycle regulation in a tissue-specific manner. To evaluate this, eight pregnant gilts were dosed with either methimazole or an equivalent negative control during days 85-106 out of 114 days of gestation (n = 4/group). Following treatment, the gilts were humanely euthanized, and tissue samples of fetal heart, ileum, kidney, lung, liver, muscle, spleen, and thymus taken from two male and two female fetuses (n = 32) from each gilt. RESULTS The relative expression of three cell cycle promoters (CDK1, CDK2, and CDK4), and one cell cycle inhibitor (CDKN1A) was compared in each tissue to determine the effect of hypothyroidism on the developing fetus. All of the eight tissues examined experienced at least one significant up- or downregulation in the expression of the aforementioned genes as a result of treatment with methimazole. Substantial changes were observed in the liver and muscle, with the latter experiencing significant downregulations of CDK1, CDK2, and CDK4 as a result of treatment. In addition, all tissues were examined for changes in protein content, which further elucidated the impact of hypothyroidism on the fetal liver by the observation of a marked increase in protein content in the methimazole-treated group. Finally, the heart and liver were histologically examined for evidence of cellular hyperplasia and hypertrophy by measuring average nuclei density and size in each tissue, with the results showing a significant decrease in average nuclei size in the liver of hypothyroid fetuses. CONCLUSIONS Collectively, these findings indicate the occurrence of organ-specific disruptions in cell cycle progression as a result of in utero hypothyroidism, which may explain the long term and widespread effects of hypothyroidism on fetal development.
Collapse
Affiliation(s)
- Alyssa A Smith
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47906, USA
| | - Alexa Vesey
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47906, USA
| | - Caden Helfrich
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47906, USA
| | - J Alex Pasternak
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47906, USA.
| |
Collapse
|
45
|
Saatci O, Alam R, Huynh-Dam KT, Isik A, Uner M, Belder N, Ersan PG, Tokat UM, Ulukan B, Cetin M, Calisir K, Gedik ME, Bal H, Sener Sahin O, Riazalhosseini Y, Thieffry D, Gautheret D, Ogretmen B, Aksoy S, Uner A, Akyol A, Sahin O. Targeting LINC00152 activates cAMP/Ca 2+/ferroptosis axis and overcomes tamoxifen resistance in ER+ breast cancer. Cell Death Dis 2024; 15:418. [PMID: 38879508 PMCID: PMC11180193 DOI: 10.1038/s41419-024-06814-3] [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: 11/21/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/19/2024]
Abstract
Tamoxifen has been the mainstay therapy to treat early, locally advanced, and metastatic estrogen receptor-positive (ER + ) breast cancer, constituting around 75% of all cases. However, the emergence of resistance is common, necessitating the identification of novel therapeutic targets. Here, we demonstrated that long-noncoding RNA LINC00152 confers tamoxifen resistance by blocking tamoxifen-induced ferroptosis, an iron-mediated cell death. Mechanistically, inhibiting LINC00152 reduces the mRNA stability of phosphodiesterase 4D (PDE4D), leading to activation of the cAMP/PKA/CREB axis and increased expression of the TRPC1 Ca2+ channel. This causes cytosolic Ca2+ overload and generation of reactive oxygen species (ROS) that is, on the one hand, accompanied by downregulation of FTH1, a member of the iron sequestration unit, thus increasing intracellular Fe2+ levels; and on the other hand, inhibition of the peroxidase activity upon reduced GPX4 and xCT levels, in part by cAMP/CREB. These ultimately restore tamoxifen-dependent lipid peroxidation and ferroptotic cell death which are reversed upon chelating Ca2+ or overexpressing GPX4 or xCT. Overexpressing PDE4D reverses LINC00152 inhibition-mediated tamoxifen sensitization by de-activating the cAMP/Ca2+/ferroptosis axis. Importantly, high LINC00152 expression is significantly correlated with high PDE4D/low ferroptosis and worse survival in multiple cohorts of tamoxifen- or tamoxifen-containing endocrine therapy-treated ER+ breast cancer patients. Overall, we identified LINC00152 inhibition as a novel mechanism of tamoxifen sensitization via restoring tamoxifen-dependent ferroptosis upon destabilizing PDE4D, increasing cAMP and Ca2+ levels, thus leading to ROS generation and lipid peroxidation. Our findings reveal LINC00152 and its effectors as actionable therapeutic targets to improve clinical outcome in refractory ER+ breast cancer.
Collapse
Affiliation(s)
- Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Rashedul Alam
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Kim-Tuyen Huynh-Dam
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Aynur Isik
- Department of Pathology, Faculty of Medicine, Hacettepe University, 06100, Ankara, Turkey
| | - Meral Uner
- Department of Pathology, Faculty of Medicine, Hacettepe University, 06100, Ankara, Turkey
| | - Nevin Belder
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800, Turkey
| | - Pelin Gulizar Ersan
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800, Turkey
| | - Unal Metin Tokat
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800, Turkey
| | - Burge Ulukan
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Metin Cetin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Kubra Calisir
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Mustafa Emre Gedik
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hilal Bal
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800, Turkey
| | - Ozlem Sener Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
| | - Denis Thieffry
- Département de biologie de l'Ecole normale supérieure, PSL Université, 75005, Paris, France
- Bioinformatics and Computational Systems Biology of Cancer, U900 Institut Curie - INSERM - Mines ParisTech, PSL Université, 75005, Paris, France
| | - Daniel Gautheret
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CNRS, CEA, 91190, Gif-sur-Yvette, France
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100, Ankara, Turkey
| | - Aysegul Uner
- Department of Pathology, Faculty of Medicine, Hacettepe University, 06100, Ankara, Turkey
| | - Aytekin Akyol
- Department of Pathology, Faculty of Medicine, Hacettepe University, 06100, Ankara, Turkey
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
46
|
Wehbe N, Badran A, Baydoun S, Al-Sawalmih A, Maresca M, Baydoun E, Mesmar JE. The Antioxidant Potential and Anticancer Activity of Halodule uninervis Ethanolic Extract against Triple-Negative Breast Cancer Cells. Antioxidants (Basel) 2024; 13:726. [PMID: 38929164 PMCID: PMC11200955 DOI: 10.3390/antiox13060726] [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: 04/28/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Natural remedies have been indispensable to traditional medicine practices for generations, offering therapeutic solutions for various ailments. In modern times, these natural products continue to play a pivotal role in the discovery of new drugs, especially for cancer treatment. The marine ecosystem offers a wide range of plants with potential anticancer activities due to their distinct biochemical diversity and adaptation to extreme situations. The seagrass Halodule uninervis is rich in diverse bioactive metabolites that bestow the plant with various pharmacological properties. However, its anticancer activity against invasive triple-negative breast cancer (TNBC) is still poorly investigated. In the present study, the phytochemical composition of an ethanolic extract of H. uninervis (HUE) was screened, and its antioxidant potential was evaluated. Moreover, the anticancer potential of HUE against MDA-MB-231 cells was investigated along with the possible underlying mechanisms of action. Our results showed that HUE is rich in diverse phytochemicals that are known for their antioxidant and anticancer effects. In MDA-MB-231 cells, HUE targeted the hallmarks of cancer, including cell proliferation, adhesion, migration, invasion, and angiogenesis. The HUE-mediated anti-proliferative and anti-metastatic effects were associated with the downregulation of the proto-oncogenic STAT3 signaling pathway. Taken together, H. uninervis could serve as a valuable source for developing novel drugs targeting TNBC.
Collapse
Affiliation(s)
- Nadine Wehbe
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| | - Adnan Badran
- Department of Nutrition, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan;
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Ali Al-Sawalmih
- Marine Science Station, University of Jordan, Aqaba 11942, Jordan;
| | - Marc Maresca
- Aix-Marseille Univ, CNRS, Centrale Marseille, iSM2, 13013 Marseille, France
| | - Elias Baydoun
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| | - Joelle Edward Mesmar
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| |
Collapse
|
47
|
Razmi M, Tajik F, Hashemi F, Yazdanpanah A, Hashemi-Niasari F, Divsalar A. The Prognostic Importance of Ki-67 in Gastrointestinal Carcinomas: A Meta-analysis and Multi-omics Approach. J Gastrointest Cancer 2024; 55:599-624. [PMID: 38411875 DOI: 10.1007/s12029-024-01022-w] [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] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
PURPOSE This study aimed to determine if Ki-67, a commonly used marker to measure tumor proliferation, is a reliable prognostic factor in various types of gastrointestinal (GI) cancers based on current high-quality multivariable evidence. METHODS A comprehensive search was conducted in PubMed, Embase, Scopus, and ISI Web of Science databases to investigate the association between Ki-67 positivity and overall survival (OS) and disease/recurrence-free survival (DFS/RFS) in GI cancers. Heterogeneity was assessed using Chi-square-based Q and I2 analyses and publication bias using funnel plots and Egger's analysis. In addition, Ki-67 levels in different GI cancers were examined by different platforms. The prognostic capability of Ki-67, gene ontology (GO), and pathway enrichment analysis were obtained from GEPIA2 and STRING. RESULTS Totally, 61 studies, involving 13,034 patients, were deemed eligible for our evaluation. The combined hazard ratios (HRs) demonstrated the prediction ability of overexpressed Ki-67 for a worse OS (HR: 1.67, P < 0.001; HR: 1.37, P = 0.021) and DFS/RFS (HR: 2.06, P < 0.001) in hepatocellular and pancreatic malignancies, respectively, as confirmed by multi-omics databases. However, similar correlation was not found in esophageal, gastric, and colorectal cancers. Furthermore, most of the associations were identified to be robust based on different subcategories and publication bias assessment. Finally, enriched Ki-67-related genes were found to be involved in various important signaling pathways, such as cell cycle, P53 signaling network, and DNA damage responses. CONCLUSION This study supports that Ki-67 can serve as an independent prognostic biomarker for pancreatic and hepatocellular malignancies in clinical settings.
Collapse
Affiliation(s)
- Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Surgery, University of California, Irvine, CA, USA
| | - Farideh Hashemi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ayna Yazdanpanah
- Department of Tissue Engineering and Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Hashemi-Niasari
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Adeleh Divsalar
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
48
|
Mendoza-Rodríguez MG, Medina-Reyes D, Sánchez-Barrera CA, Fernández-Muñoz KV, García-Castillo V, Ledesma-Torres JL, González-González MI, Reyes JL, Pérez-Plascencia C, Rodríguez-Sosa M, Vaca-Paniagua F, Meraz MA, Terrazas LI. Helminth-derived molecules improve 5-fluorouracil treatment on experimental colon tumorigenesis. Biomed Pharmacother 2024; 175:116628. [PMID: 38663106 DOI: 10.1016/j.biopha.2024.116628] [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: 01/16/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 06/03/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent fatal neoplasias worldwide. Despite efforts to improve the early diagnosis of CRC, the mortality rate of patients is still nearly 50%. The primary treatment strategy for CRC is surgery, which may be accompanied by chemotherapy and radiotherapy. The conventional and first-line chemotherapeutic agent utilized is 5-fluorouracil (5FU). However, it has low efficiency. Combination treatment with leucovorin and oxaliplatin or irinotecan improves the effectiveness of 5FU therapy. Unfortunately, most patients develop drug resistance, leading to disease progression. Here, we evaluated the effect of a potential alternative adjuvant treatment for 5FU, helminth-derived Taenia crassiceps (TcES) molecules, on treating advanced colitis-associated colon cancer. The use of TcES enhanced the effects of 5FU on established colonic tumors by downregulating the expression of the immunoregulatory cytokines, Il-10 and Tgf-β, and proinflammatory cytokines, Tnf-α and Il-17a, and reducing the levels of molecular markers associated with malignancy, cyclin D1, and Ki67, both involved in apoptosis inhibition and the signaling pathway of β-catenin. TcES+5FU therapy promoted NK cell recruitment and the release of Granzyme B1 at the tumor site, consequently inducing tumor cell death. Additionally, it restored P53 activity which relates to decreased Mdm2 expression. In vitro assays with human colon cancer cell lines showed that therapy with TcES+5FU significantly reduced cell proliferation and migration by modulating the P53 and P21 signaling pathways. Our findings demonstrate, for the first time in vivo, that helminth-derived excreted/secreted products may potentiate the effect of 5FU on established colon tumors.
Collapse
Affiliation(s)
- Mónica G Mendoza-Rodríguez
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico.
| | - Daniela Medina-Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Cuauhtémoc A Sánchez-Barrera
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Karen V Fernández-Muñoz
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Avenida Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - Verónica García-Castillo
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Jorge L Ledesma-Torres
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Marisol I González-González
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - José L Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Carlos Pérez-Plascencia
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, Mexico
| | - Miriam Rodríguez-Sosa
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Felipe Vaca-Paniagua
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico
| | - Marco A Meraz
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Avenida Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - Luis I Terrazas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico; Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Mexico.
| |
Collapse
|
49
|
Zhou JY, Pan CG, Ye Y, Li ZW, Fu WD, Jiang BH. Development and Validation of a Prognostic Nomogram for HR+ HER- Breast Cancer. Cancer Manag Res 2024; 16:491-505. [PMID: 38800665 PMCID: PMC11127650 DOI: 10.2147/cmar.s459714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Purpose We aimed to develop a nomogram to predict prognosis of HR+ HER2- breast cancer patients and guide the application of postoperative adjuvant chemotherapy. Methods We identified 310 eligible HR+ HER- breast cancer patients and randomly divided the database into a training group and a validation group. The endpoint was disease free survival (DFS). Concordance index (C-index), area under the curve (AUC) and calibration curves were used to evaluate predictive accuracy and discriminative ability of the nomogram. We also compared the predictive accuracy and discriminative ability of our nomogram with the eighth AJCC staging system using overall data. Results According to the training group, platelet-to-lymphocyte ratio (PLR), tumor size, positive lymph nodes and Ki-67 index were used to construct the nomogram of DFS. The C-index of DFS was 0.708 (95% CI: 0.623-0.793) in the training group and 0.67 (95% CI: 0.544-0.796) in the validation group. The calibration curves revealed great consistencies in both groups. Conclusion We have developed and validated a novel and practical nomogram that can provide individual prediction of DFS for patients with HR+ HER- breast cancer. This nomogram may help clinicians in risk consulting and guiding the application of postoperative adjuvant chemotherapy.
Collapse
Affiliation(s)
- Jie-Yu Zhou
- Department of Thyroid and Breast Surgery, The Wenzhou Central Hospital, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Cheng-Geng Pan
- Department of Thyroid and Breast Surgery, The Wenzhou Central Hospital, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Yang Ye
- Department of Thyroid and Breast Surgery, The Wenzhou Central Hospital, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Zhi-Wei Li
- Department of Thyroid and Breast Surgery, The Wenzhou Central Hospital, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Wei-Da Fu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315000, People’s Republic of China
| | - Bin-Hao Jiang
- Department of Urinary Surgery, Yueqing People’s Hospital, Wenzhou, Zhejiang, 325000, People’s Republic of China
| |
Collapse
|
50
|
Nayeem N, Sauma S, Ahad A, Rameau R, Kebadze S, Bazett M, Park BJ, Casaccia P, Prabha S, Hubbard K, Contel M. Insights into Mechanisms and Promising Triple Negative Breast Cancer Therapeutic Potential for a Water-Soluble Ruthenium Compound. ACS Pharmacol Transl Sci 2024; 7:1364-1376. [PMID: 38751641 PMCID: PMC11092013 DOI: 10.1021/acsptsci.4c00020] [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: 01/17/2024] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 05/18/2024]
Abstract
Triple negative breast cancer (TNBC) represents a subtype of breast cancer that does not express the three major prognostic receptors of human epidermal growth factor receptor 2 (HER2), progesterone (PR), and estrogen (ER). This limits treatment options and results in a high rate of mortality. We have reported previously on the efficacy of a water-soluble, cationic organometallic compound (Ru-IM) in a TNBC mouse xenograft model with impressive tumor reduction and targeted tumor drug accumulation. Ru-IM inhibits cancer hallmarks such as migration, angiogenesis, and invasion in TNBC cells by a mechanism that generates apoptotic cell death. Ru-IM displays little interaction with DNA and appears to act by a P53-independent pathway. We report here on the mitochondrial alterations caused by Ru-IM treatment and detail the inhibitory properties of Ru-IM in the PI3K/AKT/mTOR pathway in MDA-MB-231 cells. Lastly, we describe the results of an efficacy study of the TNBC xenografted mouse model with Ru-IM and Olaparib monotherapy and combinatory treatments. We find 59% tumor shrinkage with Ru-IM and 65% with the combination. Histopathological analysis confirmed no test-article-related toxicity. Immunohistochemical analysis indicated an inhibition of the angiogenic marker CD31 and increased levels of apoptotic cleaved caspase 3 marker, along with a slight inhibition of p-mTOR. Taken together, the effects of Ru-IM in vitro show similar trends and translation in vivo. Our investigation underscores the therapeutic potential of Ru-IM in addressing the challenges posed by TNBC as evidenced by its robust efficacy in inhibiting key cancer hallmarks, substantial tumor reduction, and minimal systemic toxicity.
Collapse
Affiliation(s)
- Nazia Nayeem
- Department
of Chemistry, Brooklyn College, The City
University of New York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Program The Graduate Center, The City
University of New York, New York, New York 10016, United States
| | - Sami Sauma
- Biology
PhD Program The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Neuroscience
Initiative, Advanced Science Research Center, New York, New York 10065, United States
- Department
of Biology, City College, The City University
of New York, New York, New York 10031, United States
| | - Afruja Ahad
- Department
of Chemistry, Brooklyn College, The City
University of New York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Program The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10031, United States
| | - Rachele Rameau
- Biology
PhD Program The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Biology, City College, The City University
of New York, New York, New York 10031, United States
| | - Sophia Kebadze
- Department
of Chemistry, Brooklyn College, The City
University of New York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Mark Bazett
- Bold
Therapeutics Inc., Vancouver, British Columbia V6C 1E1, Canada
| | - Brian J. Park
- Bold
Therapeutics Inc., Vancouver, British Columbia V6C 1E1, Canada
| | - Patrizia Casaccia
- Neuroscience
Initiative, Advanced Science Research Center, New York, New York 10065, United States
| | - Swayam Prabha
- Fels
Cancer Institute for Personalized Medicine and Department of Cancer
and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19104, United States
- Cancer
Signaling and Tumor Microenvironment Program, Fox Chase Center, Temple University, Philadelphia, Pennsylvania 19111, United States
| | - Karen Hubbard
- Biology
PhD Program The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Biology, City College, The City University
of New York, New York, New York 10031, United States
| | - Maria Contel
- Department
of Chemistry, Brooklyn College, The City
University of New York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Program The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Chemistry
PhD Program, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Biochemistry
PhD Program, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| |
Collapse
|