1
|
Ahmad GV, Nouri S, Mohammad Gholian A, Abdollahi E, Ghorbaninezhad F, Tahmasebi S, Eterafi M, Askari MR, Safarzadeh E. Breaking barriers: CAR-NK cell therapy breakthroughs in female-related cancers. Biomed Pharmacother 2025; 187:118071. [PMID: 40253831 DOI: 10.1016/j.biopha.2025.118071] [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: 01/16/2025] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025] Open
Abstract
Cancer stands as a leading cause of mortality globally. The main female-related malignancies are breast cancer, with 2.3 million new cases annually, and ovarian cancer, with 300,000 new cases per year worldwide. The current treatments like surgery, chemotherapy, and radiation therapy have presumably had deficiencies in sustaining long-term anti-tumor responses. Cellular immunotherapy, also referred to as adoptive cell therapy, has shown encouraging advances by employing genetically modified immune cells in fighting cancer by engineering chimeric antigen receptors (CARs) mainly on T cells and natural killer (NK) cells. Studies in NK cell therapies involve unmodified NK cells and CAR-NK cell therapies, targeting cancer cells while limiting the destruction of normal cells. CAR-NK cells represent the next generation of therapeutic immune cells that have been shown to eliminate malignancies through CAR-dependent and CAR-independent mechanisms. They also represent possible candidates for "off-the-shelf" therapies due to their advantages, including the ability to target cancer cells independently of the major histocompatibility complex, reduced risk of alloreactivity, and fewer severe toxicities compared to CAR-T cells. To date, there have been no comprehensive review studies examining the therapeutic potential of CAR-NK cell therapy specifically for female-related malignancies, such as breast and ovarian cancers. This review offers a thorough exploration of CAR-NK cell therapy in relation to these cancers and their responses to treatment.
Collapse
Affiliation(s)
- Ghorbani Vanan Ahmad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Samaneh Nouri
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Eileen Abdollahi
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farid Ghorbaninezhad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Safa Tahmasebi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Askari
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
2
|
Kheraldine H, Hassan AF, Saeed S, Merhi M, Mateo JM, Ulamec M, Peric-Balja M, Vranic S, Al-Thawadi H, Moustafa AEA. Neratinib and metformin: A novel therapeutic approach against HER2-Positive Breast Cancer. Biomed Pharmacother 2025; 187:118034. [PMID: 40252335 DOI: 10.1016/j.biopha.2025.118034] [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: 02/12/2025] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND HER2-positive breast cancer (BC) is highly aggressive with a poor prognosis. It is driven by HER2 oncoprotein activation/crosstalk with other receptors like EGFR/(HER1), HER3, and HER4, in addition to IGF-1R, making these receptors ideal therapeutic targets as they are expressed/overexpressed in this subtype. We postulated that targeting HER2 and IGF-1R together is a promising therapy for HER2-positive BC. Thus, we explored the outcome of a novel combination treatment using neratinib, a pan-HER inhibitor, and metformin, an IGF-1R inhibitor, on HER2-positive BC cells. METHODS In this investigation, we used cellular and molecular biology techniques in addition to an angiogenesis model and tissue microarray analysis. RESULTS Our data revealed that this combination therapy significantly reduced cell viability compared to individual treatments and exhibited a synergistic effect in HER2-positive BC cells. Moreover, the combination disrupted cell cycle progression and inhibited colony formation, and invasion of HER2-positive BC cells; this is accompanied by the deregulation of HER1-3 and IGF-1R expression patterns, in addition to Caspase-3, BCL2, Fascin, and Vimentin. Moreover, key regulator molecular pathways, including, ERK1/2, AKT, p38 MAPK, and mTOR, were significantly downregulated upon treatment with neratinib and metformin combination. Additionally, our data pointed out that neratinib and metformin combination inhibited angiogenesis, in-ovo, an important biological event in cancer progression. Finally, using a cohort of 55 HER2-positive BC samples, we revealed that HER2 and IGF-1R are co-expressed in most of the cases. CONCLUSIONS These findings suggest that neratinib and metformin combination can present a promising strategy for targeting multiple pathways in HER2-positive BC.
Collapse
Affiliation(s)
- Hadeel Kheraldine
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Arij Fouzat Hassan
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Sumayyah Saeed
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Jericha Miles Mateo
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Monika Ulamec
- Clinical Department of Pathology and Cytology Ljudevit Jurak, Sister of Charity University Hospital Center, Zagreb, Croatia; Department of Pathology and Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Melita Peric-Balja
- Oncological Pathology Department, Ljudevit Jurak Clinical Department of Pathology and Cytology, Sister of Charity University Hospital Center, Zagreb, Croatia
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hamda Al-Thawadi
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar; Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, Canada; ABS Research Review & Consultation, Montreal, QC, Canada.
| |
Collapse
|
3
|
Li QY, Liang Y, Zhang L, Li JH, Wang BJ, Wang CF. MRI-based habitat analysis for Intratumoral heterogeneity quantification combined with deep learning for HER2 status prediction in breast cancer. Magn Reson Imaging 2025:110429. [PMID: 40414575 DOI: 10.1016/j.mri.2025.110429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) is a crucial determinant of breast cancer prognosis and treatment options. The study aimed to establish an MRI-based habitat model to quantify intratumoral heterogeneity (ITH) and evaluate its potential in predicting HER2 expression status. METHODS Data from 340 patients with pathologically confirmed invasive breast cancer were retrospectively analyzed. Two tasks were designed for this study: Task 1 distinguished between HER2-positive and HER2-negative breast cancer. Task 2 distinguished between HER2-low and HER2-zero breast cancer. We developed the ITH, deep learning (DL), and radiomics signatures based on the features extracted from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Clinical independent predictors were determined by multivariable logistic regression. Finally, a combined model was constructed by integrating the clinical independent predictors, ITH signature, and DL signature. The area under the receiver operating characteristic curve (AUC) served as the standard for assessing the performance of models. RESULTS In task 1, the ITH signature performed well in the training set (AUC = 0.855) and the validation set (AUC = 0.842). In task 2, the AUCs of the ITH signature were 0.844 and 0.840, respectively, which still showed good prediction performance. In the validation sets of both tasks, the combined model exhibited the best prediction performance, with AUCs of 0.912 and 0.917 respectively, making it the optimal model. CONCLUSION A combined model integrating clinical independent predictors, ITH signature, and DL signature can predict HER2 expression status preoperatively and noninvasively.
Collapse
Affiliation(s)
- Qing-Yu Li
- Background: Huai-he Hospital of Henan University, Kaifeng, China
| | - Yue Liang
- Background: Huai-he Hospital of Henan University, Kaifeng, China
| | - Lan Zhang
- Background: Huai-he Hospital of Henan University, Kaifeng, China
| | - Jia-Hao Li
- Background: Huai-he Hospital of Henan University, Kaifeng, China
| | - Bin-Jie Wang
- Background: Huai-he Hospital of Henan University, Kaifeng, China
| | - Chang-Fu Wang
- Background: Huai-he Hospital of Henan University, Kaifeng, China.
| |
Collapse
|
4
|
Nicolò E, Gianni C, Tarantino P. Redefining Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer in the Era of Novel Antibody-Drug Conjugates. JCO Oncol Pract 2025:OP2500129. [PMID: 40359470 DOI: 10.1200/op-25-00129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
The advent of next-generation antibody-drug conjugates (ADCs), particularly trastuzumab deruxtecan (T-DXd), has transformed our understanding of human epidermal growth factor receptor 2 (HER2) targetability for breast cancer (BC) treatment. Historically categorized as HER2-positive or HER2-negative on the basis of trastuzumab eligibility, this classification has evolved significantly over the past 5 years. The DESTINY-Breast04 trial marked the entry of anti-HER2 therapies for patients with HER2-low BC, while DESTINY-Breast06 demonstrated the potential for earlier and broader use of T-DXd. The latter trial revealed that even minimal HER2 expression in tumors previously classified as HER2-0 might be clinically relevant and targetable with T-DXd. This has led to further refinement of HER2 classification, introducing the concepts of HER2-ultralow (HER2-0 with staining) and HER2-null BC (HER2-0 without staining). With these findings, most patients with metastatic BC are currently considered eligible for T-DXd. Accurately identifying candidates for these therapies has highlighted the limitations of current HER2 diagnostic practices, on the basis of immunohistochemistry (IHC)/in situ hybridization assessment. IHC assay, optimized to detect high levels of HER2 protein, faces limitations in discriminating finer variations at the lower end of the HER2 expression spectrum. This is further complicated by the heterogeneity of HER2 expression. To overcome these barriers, new approaches may be required. Quantitative methods for HER2 membrane assessment, genomic and transcriptomic evaluations of HER2, and the integration of artificial intelligence into tissue analysis hold promise and are currently under investigation. Additionally, noninvasive strategies, such as analysis of circulating tumor DNA or circulating tumor cells, may enable real-time HER2 status assessment and better patient selection for ADC. However, these techniques require rigorous validation to ensure their clinical utility. This evolving landscape underscores the need for improvement of diagnostic approaches to support the expanding role of ADCs in BC treatment.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Division of Hematology-Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Caterina Gianni
- Breast & GYN Unit, Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paolo Tarantino
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
5
|
Chai S, Cui J, Sun Y, Wang X, Cai C. Exploring Novel Therapeutic Targets in Breast Cancer via Comprehensive Omics Profiling and Experimental Verification. BIOLOGY 2025; 14:405. [PMID: 40282270 PMCID: PMC12025194 DOI: 10.3390/biology14040405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Breast cancer is the leading cause of cancer-related deaths among women worldwide. Deciphering the molecular mechanisms of breast cancer is crucial for developing targeted therapeutic approaches. METHODS This study analyzed gene expression profiles from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) in breast cancer. Mendelian randomization (MR) analysis was then employed using publicly available eQTL databases to evaluate potential causal relationships between these DEGs and breast cancer. Enrichment analyses were further conducted to explore their functional significance. Furthermore, external validation of co-expressed genes was conducted using The Cancer Genome Atlas (TCGA) database. In vitro functional assays and drug sensitivity analyses were performed on selected target genes to validate their roles in breast cancer pathogenesis and treatment. RESULTS A total of 1052 upregulated and 1380 downregulated genes were identified in breast cancer. Additionally, MR analysis revealed 12 significant co-expressed genes potentially contributing to breast cancer pathogenesis. These genes were primarily enriched in lipid metabolism and immune responses via regulating microRNA functions and AMPK signaling. Validation through the TCGA database confirmed differential expression of these genes in breast cancer tissues. Strikingly, functional assays of the less-reported genes DNASE2 and ATOH8 demonstrated their involvement in breast cancer pathogenesis through modulating proliferation, migration, and invasion of cancer cells. Notably, several commonly used clinical drugs for breast cancer management, such as 5-Fluorouracil, exhibited dramatically increased sensitivity to DNASE2 and ATOH8 expression. CONCLUSIONS Our study provides novel insights into the molecular basis of breast cancer pathogenesis and identifies promising therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Shengjun Chai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Jiayong Cui
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
| | - Yinuo Sun
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
- Department of Economics, School of Finance and Economics, Qinghai University, Xining 810016, China
| | - Xiaowu Wang
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Chunmei Cai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
| |
Collapse
|
6
|
Jin W, Zhang Y, Wang B, Kang Z, Li H, Song J, Chen Y, Xiong H, Chen J. Structural optimization and characterization of highly potent and selective STAT3 inhibitors for the treatment of triple negative breast cancer. Eur J Med Chem 2025; 287:117332. [PMID: 39938409 DOI: 10.1016/j.ejmech.2025.117332] [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: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/14/2025]
Abstract
Effective targeted treatments for triple-negative breast cancer (TNBC), which has the worst prognosis among various types of breast cancer, are lacking owing to its clinical heterogeneity and malignant nature. STAT3, a key transcription factor, regulates multiple physiological functions. Aberrant activation of STAT3 plays a pivotal role in the initiation and progression of TNBC and is closely associated with a poor prognosis. Therefore, targeting STAT3 is a promising potential therapeutic approach for TNBC. In this study, we further optimized the core structure of 6f, which our research group previously identified as a STAT3 inhibitor and treatment for osteosarcoma, to identify additional potential STAT3 inhibitors for TNBC treatment. We identified WR-S-462 as a high-binding affinity inhibitor of STAT3 that effectively suppresses its phosphorylation and biological functions in vitro. Notably, WR-S-462 significantly inhibits TNBC growth and metastasis in a dose-dependent manner, providing robust evidence for its potential as a clinical intervention for TNBC.
Collapse
Affiliation(s)
- Wangrui Jin
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China; Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Yuzhu Zhang
- Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, China
| | - Baozhen Wang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, 750004, China; Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Ningxia, 750004, China
| | - Zhaoyong Kang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Huachao Li
- Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, China
| | - Jingfeng Song
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China
| | - Yihua Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, China.
| | - Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, Ningxia, 750004, China; Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Ningxia, 750004, China.
| |
Collapse
|
7
|
Uematsu M, Nakajima H, Miyake H, Wakabayashi M, Funasaka C, Kondoh C, Harano K, Matsubara N, Hosono A, Naito Y, Sakamoto N, Kojima M, Onishi T, Ishii G, Mukohara T. Digitally quantified area of residual tumor after neoadjuvant chemotherapy in HER2-positive breast cancer. Breast Cancer 2025:10.1007/s12282-025-01694-7. [PMID: 40172786 DOI: 10.1007/s12282-025-01694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The area of residual tumor (ART) is a quantitative method for assessing tumors after neoadjuvant chemotherapy (NAC). This study evaluated whether ART can identify a favorable prognosis group in patients with HER2-positive surgically resected breast cancer and residual tumors post-NAC. METHODS We retrospectively reviewed patients with HER2-positive who underwent surgery after NAC, including trastuzumab, from 2005 to 2022 at our institution. ART was assessed at the maximum cut surface of the residual primary tumor using digital pathology images. Receiver operating characteristic curve analysis determined ART-Low and ART-High cutoffs, excluding ART-0 (0 mm2) patients. RESULTS Of the 219 patients, 82 had ART greater than 0 mm2. The median follow-up was 90.2 months. The number of patients in the ART-0, ART-Low (0 < ART ≤ 4.0 mm2), and ART-High (> 4.0 mm2) groups were 137, 39, and 43, respectively. The ART-Low group showed significantly shorter event-free survival compared to the ART-0 group (HR 3.50, 95% CI 1.52-8.06), and the ART-High group also tended toward poorer prognosis (HR 2.31, 95% CI 0.89-5.97). However, there was no significant difference in prognosis between the ART-Low and ART-High groups. CONCLUSIONS The current study suggests that even minimal residual tumor cells in the primary site can significantly impact on prognosis in HER2-positive early breast cancer.
Collapse
Affiliation(s)
- Mao Uematsu
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
| | - Hiromichi Nakajima
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
- Department of General Internal Medicine, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan.
| | - Hirohiko Miyake
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Masashi Wakabayashi
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Chuo-Ku, Japan
| | - Chikako Funasaka
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Chihiro Kondoh
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Kenichi Harano
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Ako Hosono
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Pediatric Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Yoichi Naito
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Department of General Internal Medicine, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Exploratory Oncology Research and Clinical Trial Center, Division of Pathology, Chuo-Ku, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Tatsuya Onishi
- Department of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Genichiro Ishii
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Bunkyō, Japan
| |
Collapse
|
8
|
Lian J, Yao R, Pang S, Ren X, Pan B, Zhou Y. Comprehensive analysis of HER2 Low Breast Cancer Response to Neoadjuvant Chemotherapy, a Retrospective Cohort Study. Clin Breast Cancer 2025:S1526-8209(25)00081-3. [PMID: 40254501 DOI: 10.1016/j.clbc.2025.03.013] [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: 11/20/2024] [Revised: 02/08/2025] [Accepted: 03/20/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND This study aimed to identify the response and survival outcomes of HER2-low patients following neoadjuvant chemotherapy (NAC). METHOD A retrospective cohort of patients who received NAC in the Department of Breast Surgery at Peking Union Medical College Hospital (PUMCH) from September 27, 2016, to January 23, 2024, was reviewed. Multivariate logistic regression and Cox proportional hazards models were used to identify factors associated with pathological complete response (pCR) and survival outcomes. The Kaplan-Meier method was applied to compare survival outcomes between HER2-zero and HER2-low patients. RESULT Four hundred and twenty-one patients meeting the inclusion criteria. The pCR rate of HER2-zero patients was significantly higher than that of HER2-low patients after adjusting for confounders (OR = 2.83, 95% CI: 1.44-5.65, adjusted P = .003). HER2-low patients demonstrated the most stable status after NAC within the hormone receptor (HR)-positive subset (80.82%) and the most unstable status within the HR-negative subset (57.69%). HER2-zero status was associated with worse disease-free survival (DFS) (HR = 1.94, 95% CI: 1.13-3.32, adjusted P = .02), but not with overall survival (OS) (HR = 1.18, 95% CI: 0.59-2.37, P = .65). HER2-zero patients had significantly worse DFS than HER2-low patients in the entire cohort (P = .014) and the HR-positive subset (P < .001). pCR could serve as a surrogate endpoint of favorable survival outcomes for HER2-zero patients, but not for HER2-low patients. CONCLUSION HER2-low patients might exhibit distinct characteristics compared with HER2-zero patients following NAC. Further multicenter, prospective studies are warranted to validate the conclusions of this exploratory research and assess whether HER2-low expression could serve as a new clinical subtype for evaluating NAC treatment outcomes.
Collapse
Affiliation(s)
- Jie Lian
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ru Yao
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siyuan Pang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinyu Ren
- Department of Pathology and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bo Pan
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
9
|
Shen Y, Zhang X, Zheng J, Wang S, Ding J, Sun S, Bai Q, Fu C, Wang J, Gong J, You C, Gu Y. Distinguishing Low Expression Levels of Human Epidermal Growth Factor Receptor 2 in Breast Cancer: Insights from Qualitative and Quantitative Magnetic Resonance Imaging Analysis. Tomography 2025; 11:31. [PMID: 40137571 PMCID: PMC11945706 DOI: 10.3390/tomography11030031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/14/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The discovery of novel antibody-drug conjugates for low-expression human epidermal growth factor receptor 2 (HER2-low) breast cancer highlights the inadequacy of the conventional binary classification of HER2 status as either negative or positive. Identification of HER2-low breast cancer is crucial for selecting patients who may benefit from targeted therapies. This study aims to determine whether qualitative and quantitative magnetic resonance imaging (MRI) features can effectively reflect low-HER2-expression breast cancer. METHODS Pre-treatment breast MRI images from 232 patients with pathologically confirmed breast cancer were retrospectively analyzed. Both clinicopathologic and MRI features were recorded. Qualitative MRI features included Breast Imaging Reporting and Data System (BI-RADS) descriptors from dynamic contrast-enhanced MRI (DCE-MRI), as well as intratumoral T2 hyperintensity and peritumoral edema observed in T2-weighted imaging (T2WI). Quantitative features were derived from diffusion kurtosis imaging (DKI) using multiple b-values and included statistics such as mean, median, 5th and 95th percentiles, skewness, kurtosis, and entropy from apparent diffusion coefficient (ADC), Dapp, and Kapp histograms. Differences in clinicopathologic, qualitative, and quantitative MRI features were compared across groups, with multivariable logistic regression used to identify significant independent predictors of HER2-low breast cancer. The discriminative power of MRI features was assessed using receiver operating characteristic (ROC) curves. RESULTS HER2 status was categorized as HER2-zero (n = 60), HER2-low (n = 91), and HER2-overexpressed (n = 81). Clinically, estrogen receptor (ER), progesterone receptor (PR), hormone receptor (HR), and Ki-67 levels significantly differed between the HER2-low group and others (all p < 0.001). In MRI analyses, intratumoral T2 hyperintensity was more prevalent in HER2-low cases (p = 0.009, p = 0.008). Mass lesions were more common in the HER2-zero group than in the HER2-low group (p = 0.038), and mass shape (p < 0.001) and margin (p < 0.001) significantly varied between the HER2 groups, with mass shape emerging as an independent predictive factor (HER2-low vs. HER2-zero: p = 0.010, HER2-low vs. HER2-over: p = 0.012). Qualitative MRI features demonstrated an area under the curve (AUC) of 0.763 (95% confidence interval [CI]: 0.667-0.859) for distinguishing HER2-low from HER2-zero status. Quantitative features showed distinct differences between HER2-low and HER2-overexpression groups, particularly in non-mass enhancement (NME) lesions. Combined variables achieved the highest predictive accuracy for HER2-low status, with an AUC of 0.802 (95% CI: 0.701-0.903). CONCLUSIONS Qualitative and quantitative MRI features offer valuable insights into low-HER2-expression breast cancer. While qualitative features are more effective for mass lesions, quantitative features are more suitable for NME lesions. These findings provide a more accessible and cost-effective approach to noninvasively identifying patients who may benefit from targeted therapy.
Collapse
Affiliation(s)
- Yiyuan Shen
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Xu Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Jinlong Zheng
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Jie Ding
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Shiyun Sun
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Qianming Bai
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China
| | - Caixia Fu
- MR Application Development, Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen 518057, China
| | - Junlong Wang
- Department of Nuclear Medicine, Insel Hospital, University of Bern, 3010 Bern, Switzerland;
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China (J.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Shanghai 200032, China;
| |
Collapse
|
10
|
Zhou Y, Gong J, Deng X, Shen L, Ge A, Fan H, Ling J, Wu S, Liu L. A comprehensive exploration of adverse reactions to lapatinib: a disproportionate analysis based on the FAERS database. Expert Opin Drug Saf 2025:1-10. [PMID: 39985750 DOI: 10.1080/14740338.2025.2471515] [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: 10/04/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Lapatinib, an FDA-approved tyrosine kinase inhibitor, treats HER2+ advanced/metastatic breast cancer. This study comprehensively analyzed its adverse reaction profile using FDA Adverse Event Reporting System (FAERS) to guide clinical use. RESEARCH DESIGN AND METHODS Adverse event (AE) reports for lapatinib from the second quarter of 2007 to the second quarter of 2024 in FAERS were analyzed using Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Multi-item Gamma Poisson Shrinkage (MGPS) and Bayesian Confidence Propagation Neural Network (BCPNN) to identify AE signals. RESULTS Among 8300 AE reports, females (91.47%) and ages 40-59.9 (33.71%) were predominant. 20 system organ classifications (SOCs) were affected, with gastrointestinal disorders (ROR = 3.46) and skin disorders (ROR = 2.47) most significant. Based on the PT level, a total of 111 PTs were analyzed that met the four algorithms, including typical AEs such as diarrhea (n = 3410), vomiting (n = 856), and rash (n = 856), as well as some rare AEs that were not prompted by the drug inserts, such as neutropenia (n = 252), pericardial effusion (n = 43), lymphedema (n = 20). The majority of lapatinib-associated AEs had onset within 30 days (51%). CONCLUSIONS Lapatinib has a generally favorable safety profile, but gastrointestinal toxicity and dermatotoxicity require close monitoring to prevent serious AEs.
Collapse
Affiliation(s)
- Yao Zhou
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Gong
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xianguang Deng
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lele Shen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Anqi Ge
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hongqiao Fan
- Department of Aesthetic Plastic Surgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Ling
- Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| | - Shiting Wu
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lifang Liu
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
11
|
Turova P, Kushnarev V, Baranov O, Butusova A, Menshikova S, Yong ST, Nadiryan A, Antysheva Z, Khorkova S, Guryleva MV, Bagaev A, Lennerz JK, Chernyshov K, Kotlov N. The Breast Cancer Classifier refines molecular breast cancer classification to delineate the HER2-low subtype. NPJ Breast Cancer 2025; 11:19. [PMID: 39979291 PMCID: PMC11842814 DOI: 10.1038/s41523-025-00723-0] [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: 04/19/2024] [Accepted: 01/19/2025] [Indexed: 02/22/2025] Open
Abstract
Current breast cancer classification methods, particularly immunohistochemistry and PAM50, face challenges in accurately characterizing the HER2-low subtype, a therapeutically relevant entity with distinct biological features. This notable gap can lead to misclassification, resulting in inappropriate treatment decisions and suboptimal patient outcomes. Leveraging RNA-seq and machine-learning algorithms, we developed the Breast Cancer Classifier (BCC), a unique transcriptomic classifier for more precise breast cancer subtyping, specifically by delineating and incorporating HER2-low as a distinct subtype. BCC also redefined the PAM50 Normal subtype into other subtypes, disputing its classification as a unique molecular group. Our statistical analysis not only confirmed the reproducibility and accuracy of BCC, but also revealed similarities in prognostic characteristics between the HER2-low and Basal subtypes. Addressing this gap in breast cancer classification is clinically significant because it not only improves treatment stratification, but also uncovers novel molecular and immunohistochemical features associated with the HER2-low and HER2-high subtypes, thereby advancing our understanding of breast cancer heterogeneity and providing guidance in precision oncology.
Collapse
|
12
|
Wu Y, Liu Y, Wu H, Tong M, Du L, Ren S, Che Y. Advances in Ultrasound-Targeted Microbubble Destruction (UTMD) for Breast Cancer Therapy. Int J Nanomedicine 2025; 20:1425-1442. [PMID: 39925678 PMCID: PMC11804227 DOI: 10.2147/ijn.s504363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
Breast cancer is one of the most common types of cancer in women worldwide and is a leading cause of cancer deaths among women. As a result, various treatments have been developed to combat this disease. Breast cancer treatment varies based on its stage and type of pathology. Among the therapeutic options, ultrasound has been employed to assist in the treatment of breast cancer, including radiation therapy, chemotherapy, targeted immunotherapy, hormonal therapy, and, more recently, radiofrequency ablation for early-stage and inoperable patients. One notable advancement is ultrasound-targeted microbubble destruction (UTMD), which is gradually becoming a highly effective and non-invasive anti-tumor modality. This technique can enhance chemical, genetic, immune, and anti-vascular therapies through its physical and biological effects. Specifically, UTMD improves drug transfer efficiency and destroys tumor neovascularization while reducing toxic side effects on the body during tumor treatment. Given these developments, the application of ultrasound-assisted therapy to breast cancer has gained significant attention from research scholars. In this review, we will discuss the development of various therapeutic modalities for breast cancer and, importantly, highlight the application of ultrasound microbubble-targeted disruption techniques in breast cancer treatment.
Collapse
Affiliation(s)
- Yunfeng Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Yuxi Liu
- Department of Ultrasound, Shandong Second Medical University Affiliated Hospital, Shan Dong, Weifang, People’s Republic of China
| | - Han Wu
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Mengying Tong
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Linyao Du
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Shuangsong Ren
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| | - Ying Che
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, People’s Republic of China
| |
Collapse
|
13
|
Scalambra L, Ruzzi F, Pittino OM, Semprini MS, Cappello C, Angelicola S, Palladini A, Nanni P, Goksøyr L, Fougeroux C, Penichet ML, Sander AF, Lollini PL. Targeting PCSK9, through an innovative cVLP-based vaccine, enhanced the therapeutic activity of a cVLP-HER2 vaccine in a preclinical model of HER2-positive mammary carcinoma. J Transl Med 2025; 23:136. [PMID: 39885551 PMCID: PMC11784117 DOI: 10.1186/s12967-025-06126-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] [Received: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND HER2-targeted therapies have revolutionized the treatment of HER2-positive breast cancer patients, leading to significant improvements in tumor response rates and survival. However, resistance and incomplete response remain considerable challenges. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition is a novel therapeutic strategy for the management of dyslipidemia by enhancing the clearance of low-density lipoprotein cholesterol receptors, however recent evidence also shows links between PCSK9 and cancer cells. We present an innovative immunization approach combining capsid virus-like particle (cVLP)-based vaccines against HER2 and PCSK9. METHODS The therapeutic activity of the combined vaccine was evaluated in female mice challenged with HER2-positive mammary carcinoma cells. Controls included untreated mice and mice treated with cVLP-PCSK9 and cVLP-HER2 as standalone therapies. Antibodies elicited by vaccinations were detected through ELISA immunoassay. The functional activity of the antibodies was tested in 3D-soft agar assay on human HER2 + + + trastuzumab sensitive and resistant cells. RESULTS Mice vaccinated with cVLP-HER2 + cVLP-PCSK9 displayed tumor regression from the 40th day after cell challenge in 100% of mice remaining tumor-free even 4 months later. In contrast, 83% of mice treated with cVLP-HER2 vaccine alone experienced an initial tumor regression, followed by tumor relapse in 60% of subjects. Untreated mice and mice treated with the cVLP-PCSK9 vaccine alone developed progressive tumors within 1-2 months after cell injection. The combined vaccine approach elicited strong anti-human HER2 antibody responses (reaching 1-2 mg/ml range) comprising multiple immunoglobulins isotypes. cVLP-PCSK9 vaccine elicited anti-PCSK9 antibody responses, resulting in a marked reduction in PCSK9 serum levels. Although the anti-PCSK9 response was reduced when co-administered with cVLP-HER2, it remained significant. Moreover, both cVLP-HER2 + cVLP-PCSK9 and cVLP-HER2 alone induced anti-HER2 antibodies able to inhibit the 3D growth of human HER2 + + + BT-474 and trastuzumab-resistant BT-474 C5 cells. Strikingly, antibodies elicited by the combined vaccination were more effective than those elicited by the cVLP-HER2 vaccine alone in the inhibition of trastuzumab-resistant C5 cells. CONCLUSIONS The results indicate that cVLP-PCSK9 vaccination shows adjuvant activity when combined with cVLP-HER2 vaccine, enhancing its therapeutic efficacy against HER2-positive breast cancer and holding promise in overcoming the challenges posed by resistance and incomplete responses to HER2-targeted therapy.
Collapse
Affiliation(s)
- Laura Scalambra
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Ruzzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
| | - Olga Maria Pittino
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Sofia Semprini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Chiara Cappello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Stefania Angelicola
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Policlinico Di Sant'Orsola University Hospital, Bologna, Italy
| | - Arianna Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Medical Oncology Division, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Patrizia Nanni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | | | | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery and Department of Microbiology, Immunology and Molecular GeneticsThe Molecular Biology InstituteJonsson Comprehensive Cancer Centre, University of California, Los Angeles (UCLA), CA, USA
| | - Adam Frederik Sander
- AdaptVac Aps, Copenhagen, Denmark
- Centre for Translational Medicine and Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pier-Luigi Lollini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
- IRCCS Policlinico Di Sant'Orsola University Hospital, Bologna, Italy.
| |
Collapse
|
14
|
Veeraraghavan J, De Angelis C, Gutierrez C, Liao FT, Sabotta C, Rimawi MF, Osborne CK, Schiff R. HER2-Positive Breast Cancer Treatment and Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:495-525. [PMID: 39821040 DOI: 10.1007/978-3-031-70875-6_24] [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: 01/19/2025]
Abstract
HER2-positive (+) breast cancer is an aggressive disease with poor prognosis, a narrative that changed drastically with the advent and approval of trastuzumab, the first humanized monoclonal antibody targeting HER2. In addition to another monoclonal antibody, more classes of HER2-targeted agents, including tyrosine kinase inhibitors, and antibody-drug conjugates were developed in the years that followed. While these potent therapies have substantially improved the outcome of patients with HER2+ breast cancer, resistance has prevailed as a clinical challenge ever since the arrival of targeted agents. Efforts to develop new treatment regimens to treat/overcome resistance is futile without a primary understanding of the mechanistic underpinnings of resistance. Resistance could be attributed to mechanisms that are either specific to the tumor epithelial cells or those that emerge through changes in the tumor microenvironment. Reactivation of the HER receptor layer due to incomplete blockade of the HER receptor layer or due to alterations in the HER receptors is one of the major mechanisms. In other instances, resistance may occur due to deregulations in key downstream signaling such as the PI3K/AKT or RAS/MEK/ERK pathways or due to the emergence of compensatory pathways such as ER, other RTKs, or metabolic pathways. Potent new targeted agents and approaches to target key actionable drivers of resistance have already been identified, many of which are in early clinical development or under preclinical evaluation. Ongoing and future translational research will continue to uncover additional therapeutic vulnerabilities, as well as new targeted agents and approaches to treat and/or overcome anti-HER2 treatment resistance.
Collapse
Affiliation(s)
- Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Carmine De Angelis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Carolina Gutierrez
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Fu-Tien Liao
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Caroline Sabotta
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mothaffar F Rimawi
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - C Kent Osborne
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
15
|
Pallathadka H, Jabir M, Rasool KH, Hanumanthaiah M, Sharma N, Pramanik A, Rab SO, Jawad SF, Oghenemaro EF, Mustafa YF. siRNA-based therapy for overcoming drug resistance in human solid tumours; molecular and immunological approaches. Hum Immunol 2025; 86:111221. [PMID: 39700968 DOI: 10.1016/j.humimm.2024.111221] [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: 08/10/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
RNA interference (RNAi) is a primordial biological process that protects against external intrusion. SiRNA has the potential to selectively silence disease-related genes in a sequence-specific way, thus offering a promising therapeutic approach. The efficacy of siRNA-based therapies in cancer treatment has gained significant recognition due to multiple studies demonstrating its ability to effectively suppress cancer cells' growth and multiplication. Moreover, siRNA-based medicines have shown considerable promise in enhancing the sensitivity of cancer cells to chemotherapy and other treatment methods by suppressing genes that play a role in the development of drug resistance. Exploring and identifying functional genes linked to cancer cell characteristics and drug resistance is crucial for developing effective siRNAs for cancer treatment and advancing targeted and personalized therapeutics. Targeting and silencing genes in charge of resistance mechanisms, such as those involved in drug efflux, cell survival, or DNA repair, is possible with siRNA therapy in the context of drug resistance, especially cancer. Through inhibiting these genes, siRNA therapy can prevent resistance and restore the efficacy of traditional medications. This review addresses the potential of siRNAs in addressing drug resistance in human tumours, opening up new possibilities in cancer therapy. This review article offers a non-systematic summary of how different siRNA types contribute to cancer cells' treatment resistance. Using pertinent keywords, sources were chosen from reliable databases, including PubMed, Scopus, and Google Scholar. The review covered essential papers in this area and those that mainly addressed the function of siRNA in drug resistance. The articles examined in connection with the title of this review were primarily published from 2020 onward and are based on in vitro studies. Furthermore, this article examines the potential barriers and prospective perspectives of siRNA therapies.
Collapse
Affiliation(s)
| | - Majid Jabir
- Department of Applied Sciences, University of Technology, Iraq
| | | | - Malathi Hanumanthaiah
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri - 140307, Mohali, Punjab, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001 Babil, Iraq.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Delta State University, Faculty of Pharmacy, PMB 1 Abraka, Delta State, Nigeria
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
16
|
Abdel-Aziz AAM, El-Azab AS, Brogi S, Ayyad RR, Al-Suwaidan IA, Hefnawy M. Antitumor Activity and Multi-Target Mechanism of Phenolic Schiff Bases Bearing Methanesulfonamide Fragments: Cell Cycle Analysis and a Molecular Modeling Study. Int J Mol Sci 2024; 25:13621. [PMID: 39769383 PMCID: PMC11728000 DOI: 10.3390/ijms252413621] [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: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/30/2025] Open
Abstract
Five phenolic Schiff bases (7-11) incorporating a fragment of methanesulfonamide were synthesized and evaluated for their efficacy as antitumor agents. Compounds 7 and 8 demonstrated the most potent antitumor action, with a positive cytotoxic effect (PCE) of 54/59 and 59/59 and a mean growth percentage (MG%) of 67.3% and 19.5%, respectively, compared with imatinib (PCE = 20/59 and MG% = 92.6%). The PCE values for derivatives 9-11 were 3/59, 4/59, and 4/59, respectively, indicating poor antitumor effect. Compound 8 exhibited the most significant efficacy, suppressing cell proliferation by an average of 50% at a dosage of 0.501 µM, in comparison with the reference drugs sorafenib (2.33 µM), gefitinib (2.10 µM), erlotinib (7.68 µM), and celecoxib (17.5 µM). Compounds 7 and 8 had substantial inhibitory effects on the human epidermal growth factor receptor 2 (HER2), with IC50 values of 0.183 μM and 0.464 μM, respectively. Furthermore, they exhibited significant inhibition of the epidermal growth factor receptor (EGFR), with IC50 values of 0.752 μM and 0.166 μM, respectively. Compound 8 exhibited the highest COX-2 inhibition (IC50 = 12.76 μM). We performed molecular docking dynamic experiments to examine the precise interaction and structural prerequisites for the anticancer activity of derivatives 7 and 8 by targeting EGFR and HER2.
Collapse
Affiliation(s)
- Alaa A.-M. Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.S.E.-A.); (I.A.A.-S.); (M.H.)
| | - Adel S. El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.S.E.-A.); (I.A.A.-S.); (M.H.)
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Rezk R. Ayyad
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hilla, Babylon 6202, Iraq;
| | - Ibrahim A. Al-Suwaidan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.S.E.-A.); (I.A.A.-S.); (M.H.)
| | - Mohamed Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (A.S.E.-A.); (I.A.A.-S.); (M.H.)
| |
Collapse
|
17
|
Suha H, Tasnim SA, Rahman S, Alodhayb A, Albrithen H, Poirier RA, Uddin KM. Evaluating the Anticancer Properties of Novel Piscidinol A Derivatives: Insights from DFT, Molecular Docking, and Molecular Dynamics Studies. ACS OMEGA 2024; 9:49639-49661. [PMID: 39713673 PMCID: PMC11656217 DOI: 10.1021/acsomega.4c07808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024]
Abstract
Cancer is characterized by uncontrolled cell growth and spreading throughout the body. This study employed computational approaches to investigate 18 naturally derived anticancer piscidinol A derivatives (1-18) as potential therapeutics. By examining their interactions with 15 essential target proteins (HIF-1α, RanGAP, FOXM1, PARP2, HER2, ERα, NGF, FAS, GRP78, PRDX2, SCF complex, EGFR, Bcl-xL, ERG, and HSP70) and comparing them with established drugs such as camptothecin, docetaxel, etoposide, irinotecan, paclitaxel, and teniposide, compound 10 emerged as noteworthy. In molecular dynamics simulations, the protein with the strongest binding to the crucial 1A52 protein exceeded druglikeness criteria and displayed extraordinary stability within the enzyme's pocket over varied temperatures (300-320 K). Additionally, density functional theory was used to calculate dipole moments and molecular orbital characteristics, as well as analyze the thermodynamic stability of the putative anticancer derivatives. This finding reveals a well-defined, potentially therapeutic relationship supported by theoretical analysis, which is in good agreement with subsequent assessments of their potential in vitro cytotoxic effects of piscidinol A derivatives (6-18) against various cancer cell lines. Future in vivo and clinical studies are required to validate these findings further. Compound 10 thus emerges as an intriguing contender in the fight against cancer.
Collapse
Affiliation(s)
- Humaera
Noor Suha
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Syed Ahmed Tasnim
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Shofiur Rahman
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah Alodhayb
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Department
of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hamad Albrithen
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Department
of Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Raymond A. Poirier
- Department
of Chemistry, Memorial University, St. John’s, Newfoundland
and Labrador A1C 5S7, Canada
| | - Kabir M. Uddin
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1229, Bangladesh
| |
Collapse
|
18
|
Tian H, Li G, Zheng J, Ding Z, Luo Y, Mai S, Hu J, Huang Z, Xu J, Wu H, Dong F. Comparing core needle biopsy and surgical excision in breast cancer diagnosis: implications for clinical practice from a retrospective cohort study. Quant Imaging Med Surg 2024; 14:8281-8293. [PMID: 39698620 PMCID: PMC11652020 DOI: 10.21037/qims-24-198] [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: 01/29/2024] [Accepted: 09/03/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Preoperative ultrasound-guided core needle biopsy (CNB) is currently the standard procedure for managing breast illnesses. However, the differences in outcomes between CNB and surgical excision (SE) have not been thoroughly assessed. This study aimed to explore the disparities in pathological outcomes between these two procedures, using a large sample dataset. METHODS This retrospective study consecutively included patients who underwent CNB and SE at Shenzhen People's Hospital from May 2016 to June 2023. Immunohistochemistry (IHC) was utilized to determine the status of estrogen receptor (ER), progesterone receptor (PgR), human epidermal growth factor receptor-2 (HER2), and Ki-67. Patients presenting with HER2 IHC 2+ underwent additional fluorescence in situ hybridization (FISH) examination. The cutoff value for high Ki-67 expression was established at 14%. Molecular subtypes were classified into four groups (Luminal A, Luminal B, Triple-negative, and HER2-positive) and five groups [Luminal A, Luminal B+ (HER2-positive), Luminal B- (HER2-negative), Triple-negative, and HER2-positive], based on different criteria. RESULTS A total of 4,209 patients were included in this study. Post-surgical confirmation revealed 2,410 cases as benign and 1,799 as malignant. Among the malignant cases, 334 were excluded due to either not having undergone direct surgery or having incomplete IHC results. The remaining 1,465 cases underwent IHC testing. CNB demonstrated a 97% concordance rate (CR) in diagnosing benign cases. The CRs for diagnosing invasive breast cancer (IBC) and carcinoma in situ (CIS) were 92% and 54%, respectively. ER, PgR, HER2, and Ki-67 exhibited CRs of 94%, 91%, 98%, and 84%, respectively. In the four-group classification, the overall diagnostic CR was 82%, with CRs for Luminal A, Luminal B, HER2-positive, and triple-negative breast cancer (TNBC) being 84%, 82%, 78%, and 85%, respectively. Under the five-group classification, the overall diagnostic CR was also 82%, with CRs for Luminal A, Luminal B+, Luminal B-, HER2-positive, and TNBC being 86%, 85%, 94%, 88%, and 92%, respectively. CONCLUSIONS This study demonstrates that CNB is highly accurate in differentiating benign from malignant breast lesions, particularly showing significant consistency in the diagnosis of molecular subtypes, providing a reliable reference for clinical diagnosis.
Collapse
Affiliation(s)
- Hongtian Tian
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Guoqiu Li
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Jing Zheng
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Zhimin Ding
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Yuwei Luo
- Department of Thyroid and Breast Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Simin Mai
- Department of Pathology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Jintao Hu
- Department of Pathology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Zhibin Huang
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Jinfeng Xu
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Huaiyu Wu
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Fajin Dong
- Department of Ultrasound, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| |
Collapse
|
19
|
Zhang XD, Kou M, Zou WY, Duan X, Di GX, Qin W, Bian LH, Ma ZP. HER-2 expression is correlated with multimodal imaging features in breast cancer: a pilot study. Ann Med 2024; 56:2434182. [PMID: 39618080 PMCID: PMC11613333 DOI: 10.1080/07853890.2024.2434182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/15/2023] [Accepted: 10/23/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVE A pilot study to evaluate the correlation between multimodal imaging features and the expression of the human epidermal growth factor receptor type 2 (HER-2) in breast cancer to provide a basis for clinical treatment and prognosis evaluation. METHODS We included a total of 62 patients with breast cancer admitted to the Affiliated Hospital of Hebei University between 2018 and 2022. All of them underwent the relevant investigations, including ultrasound, mammography, and enhanced magnetic resonance imaging (MRI), in the hospital within one month before surgery or biopsy. HER-2 expression level was divided into negative and positive by immunohistochemistry(IHC). Using SPSS 24.0 statistical software to analyze the differences in imaging features between the HER-2 positive and the HER-2 negative groups. RESULTS There was a statistically significant difference between the HER-2 positive and the HER-2 negative groups (p = 0.005) in the hyperechoic halo sign around the lesion detected by ultrasonography as well as in the apparent diffusion coefficient (ADC) on MRI (p = 0.047). The sensitivity and specificity of the hyperechoic halo sign in predicting HER-2 positivity was 48.3% and 84.8% respectively, and the area under the curve (AUC) for the ADC value to predict HER-2 expression was 0.533. When b was equal to 800 and the ADC value (cutoff value) was 0.000888 mm2/s, the sensitivity and specificity were 65.5% and 51.5%, respectively. CONCLUSION A combination of multimodal imaging features and HER-2 gene expression can provide more valuable information for clinical diagnosis and therapeutic schedule in breast cancer.
Collapse
Affiliation(s)
- Xiao-Dan Zhang
- Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, China
| | - Min Kou
- Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, China
| | - Wei-Yan Zou
- Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, China
| | - Xu Duan
- Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, China
| | - Gui-Xin Di
- Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, China
| | - Wei Qin
- Department of Integrated Chinese and Western Medicine, Affiliated Hospital of Hebei University, Baoding, China
| | - Li-Hui Bian
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Ze-Peng Ma
- Department of Radiology, Affiliated Hospital of Hebei University/ Clinical Medical College, Hebei University, Baoding, China
| |
Collapse
|
20
|
Zhang L, Cui QX, Zhou LQ, Wang XY, Zhang HX, Zhu YM, Sang XQ, Kuai ZX. MRI-based vector radiomics for predicting breast cancer HER2 status and its changes after neoadjuvant therapy. Comput Med Imaging Graph 2024; 118:102443. [PMID: 39427545 DOI: 10.1016/j.compmedimag.2024.102443] [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: 04/25/2024] [Revised: 07/24/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
PURPOSE To develop a novel MRI-based vector radiomic approach to predict breast cancer (BC) human epidermal growth factor receptor 2 (HER2) status (zero, low, and positive; task 1) and its changes after neoadjuvant therapy (NAT) (positive-to-positive, positive-to-negative, and positive-to-pathologic complete response; task 2). MATERIALS AND METHODS Both dynamic contrast-enhanced (DCE) MRI data and multi-b-value (MBV) diffusion-weighted imaging (DWI) data were acquired in BC patients at two centers. Vector-radiomic and conventional-radiomic features were extracted from both DCE-MRI and MBV-DWI. After feature selection, the following models were built using the retained features and logistic regression: vector model, conventional model, and combined model that integrates the vector-radiomic and conventional-radiomic features. The models' performances were quantified by the area under the receiver-operating characteristic curve (AUC). RESULTS The training/external test set (center 1/2) included 483/361 women. For task 1, the vector model (AUCs=0.73∼0.86) was superior to (p<.05) the conventional model (AUCs=0.68∼0.81), and the addition of vector-radiomic features to conventional-radiomic features yielded an incremental predictive value (AUCs=0.80∼0.90, p<.05). For task 2, the combined MBV-DWI model (AUCs=0.85∼0.89) performed better than (p<.05) the conventional MBV-DWI model (AUCs=0.73∼0.82). In addition, for the combined DCE-MRI model and the combined MBV-DWI model, the former (AUCs=0.85∼0.90) outperformed (p<.05) the latter (AUCs=0.80∼0.85) in task 1, whereas the latter (AUCs=0.85∼0.89) outperformed (p<.05) the former (AUCs=0.76∼0.81) in task 2. The above results are true for the training and external test sets. CONCLUSIONS MRI-based vector radiomics may predict BC HER2 status and its changes after NAT and provide significant incremental prediction over and above conventional radiomics.
Collapse
Affiliation(s)
- Lan Zhang
- Imaging Center, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin, 150081, China
| | - Quan-Xiang Cui
- Imaging Center, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin, 150081, China
| | - Liang-Qin Zhou
- Imaging Center, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin, 150081, China
| | - Xin-Yi Wang
- Imaging Center, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin, 150081, China
| | - Hong-Xia Zhang
- Imaging Center, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin, 150081, China
| | - Yue-Min Zhu
- CREATIS, CNRS UMR 5220-INSERM U1206-University Lyon 1-INSA Lyon-University Jean Monnet Saint-Etienne, Lyon 69621, France
| | - Xi-Qiao Sang
- Division of Respiratory Disease, Fourth Affiliated Hospital of Harbin Medical University, Yiyuan Street No. 37, Nangang District, Harbin, 150001, China
| | - Zi-Xiang Kuai
- Imaging Center, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin, 150081, China.
| |
Collapse
|
21
|
Luo HJ, Ren JL, Mei Guo L, Liang Niu J, Song XL. MRI-based machine learning radiomics for prediction of HER2 expression status in breast invasive ductal carcinoma. Eur J Radiol Open 2024; 13:100592. [PMID: 39149534 PMCID: PMC11324846 DOI: 10.1016/j.ejro.2024.100592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/08/2024] [Accepted: 07/14/2024] [Indexed: 08/17/2024] Open
Abstract
Background Human epidermal growth factor receptor 2 (HER2) is a tumor biomarker with significant prognostic and therapeutic implications for invasive ductal breast carcinoma (IDC). Objective This study aimed to explore the effectiveness of a multisequence magnetic resonance imaging (MRI)-based machine learning radiomics model in classifying the expression status of HER2, including HER2-positive, HER2-low, and HER2 completely negative (HER2-zero), among patients with IDC. Methods A total of 402 female patients with IDC confirmed through surgical pathology were enrolled and subsequently divided into a training group (n = 250, center I) and a validation group (n = 152, center II). Radiomics features were extracted from the preoperative MRI. A simulated annealing algorithm was used for key feature selection. Two classification tasks were performed: task 1, the classification of HER2-positive vs. HER2-negative (HER2-low and HER2-zero), and task 2, the classification of HER2-low vs. HER2-zero. Logistic regression, random forest (RF), and support vector machine were conducted to establish radiomics models. The performance of the models was evaluated using the area under the curve (AUC) of the operating characteristics (ROC). Results In total, 4506 radiomics features were extracted from multisequence MRI. A radiomics model for prediction of expression state of HER2 was successfully developed. Among the three classification algorithms, RF achieved the highest performance in classifying HER2-positive from HER2-negative and HER2-low from HER2-zero, with AUC values of 0.777 and 0.731, respectively. Conclusions Machine learning-based MRI radiomics may aid in the non-invasive prediction of the different expression status of HER2 in IDC.
Collapse
Affiliation(s)
- Hong-Jian Luo
- Department of Radiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zuiyi, Guizhou province, China
| | | | - Li Mei Guo
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| | - Jin Liang Niu
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| | - Xiao-Li Song
- Department of Radiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi province, China
| |
Collapse
|
22
|
Verma A, Patel K, Kumar A. Targeting drug resistance in breast cancer: the potential of miRNA and nanotechnology-driven delivery systems. NANOSCALE ADVANCES 2024:d4na00660g. [PMID: 39569336 PMCID: PMC11575621 DOI: 10.1039/d4na00660g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Breast cancer is the second leading cause of cancer-related deaths in females worldwide. Despite significant advancements in treatment, drug resistance remains a major challenge, limiting the effectiveness of therapies and leading to dismal outcomes. Approximately 50% of HER2+ breast cancer patients develop resistance to trastuzumab, and patients with triple-negative breast cancer often experience resistance to first-line therapies. The drug resistance mechanisms involve altered drug uptake, enhanced DNA repair, and dysregulated apoptosis pathways. MicroRNAs are essential in regulating cellular processes involved in both homeostasis and disease. Recent data suggest that microRNAs can overcome drug resistance by regulating the pathways that confer drug resistance. Combining different conventional anticancer agents with microRNA therapies holds promise for enhancing treatment effectiveness against drug resistant breast cancer. Advancements in nano-drug delivery systems have facilitated the effective delivery of microRNAs by improving their stability, targeting specific cells, and enhancing cellular uptake. This review elucidates the recent advancements in microRNA-based therapies, their effects on gene expression, and their clinical efficacy in overcoming drug resistance in breast cancer.
Collapse
Affiliation(s)
- Aditi Verma
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| | - Krunal Patel
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University Central Campus, Navrangpura Ahmedabad 380009 Gujarat India
| |
Collapse
|
23
|
Mou E, Ji J, Liu S, Shu L, Zou L, Li Z. Differences in axillary response and treatment implications in HER2 positive node positive breast cancer during neoadjuvant HER2 targeted dual therapy. Sci Rep 2024; 14:26364. [PMID: 39487187 PMCID: PMC11530619 DOI: 10.1038/s41598-024-78176-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
Explore whether the axillary outcomes differ among HER2 positive subgroups receiving standard dual-targeted therapy, aiming to identify subgroups exhibiting enhanced sensitivity to NAT among HER2-positive/node-positive breast cancer patients. HER2 positive female patients with biopsy-proven node-positive disease from April 2020 to May 2023 were included. All patients underwent standard Neoadjuvant HER2-targeted dual therapy and axillary lymph node dissection (ALND) at Breast Surgery Center of Sichuan Cancer Hospital. Univariate and multivariate analyses were used to identify factors associate with axillary pathological complete response (ApCR). Statistical analysis and graphing were performed using SPSS 24.0 and GraphPad Prism 9.0 software. This study enrolled 215 HER2 positive patients with a total ApCR rate of 76.7%, which included 49 HER2 2+/FISH + and 166 HER2 3 + cases with approximate ApCR rates of 63.3% and 80.7% (P = 0.011). Univariate and multivariate analysis indicated that HER2 3 + disease (OR = 2.43, 95% CI 1.21-4.88, P = 0.012), Ki-67 ≥ 20% disease (OR = 3.00, 95% CI 1.26-7.13, P = 0.013) and NAC regimen of TCb (OR = 2.71, 95% CI 1.39-5.38, P = 0.004) were more likely to achieve ApCR. Further subgroup analysis revealed that HER2 3 + patients receiving TCb regimen showed the highest ApCR rate of 88% compared to other subgroups. HER2 3 + breast cancer had a higher ApCR rate than HER2 2+/FISH + breast cancer during Neoadjuvant HER2-targeted dual therapy. HER2 positive patients could benefit from NAC regimen of TCb in axillary response.
Collapse
Affiliation(s)
- Exian Mou
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Juan Ji
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shiwei Liu
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Shu
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Liqun Zou
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhuoxuan Li
- Department of Breast, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
24
|
Yan L, Wen Z, Yang Y, Liu A, Li F, Zhang Y, Yang C, Li Y, Zhang Y. Dissecting the roles of prosaposin as an emerging therapeutic target for tumors and its underlying mechanisms. Biomed Pharmacother 2024; 180:117551. [PMID: 39405903 DOI: 10.1016/j.biopha.2024.117551] [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: 07/27/2024] [Revised: 09/22/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
As a dual-function protein, prosaposin (PSAP) is a lysosome-associated protein that participates in a variety of cellular processes. In the lysosome, PSAP is processed to activate enzymes that degrade lipids. In addition, PSAP proteins located extracellularly are involved in cancer progression, such as proliferation and tumor death suppression signaling. Moreover, under different situations, PSAP exhibits distinct metastasis potentials in tumors. However, comprehensive insight into PSAP in cancer progression has been lacking. Here, we provide a framework of the role of PSAP in cancer and its clinical application in cancer patients, providing a novel perspective on the clinical translation of PSAP.
Collapse
Affiliation(s)
- Lirong Yan
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Zhenpeng Wen
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Yi Yang
- Department of Laboratory Animal Science, China Medical University, Shenyang, China
| | - Aoran Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Fang Li
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Chunjiao Yang
- Department of Oncology, The Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning, Nanning, Guangxi, China
| | - Yanke Li
- Department of Anorectal Surgery, the First Hospital of China Medical University, Shenyang, China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, China.
| |
Collapse
|
25
|
Ouyang W, Liu Y, Cao J, Chen J. Trastuzumab Deruxtecan in HER2-Positive Breast Cancer with HER2 Loss After Dual-Target Adjuvant Therapy: A Case Report. Am J Ther 2024:00045391-990000000-00223. [PMID: 39451010 DOI: 10.1097/mjt.0000000000001795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Affiliation(s)
- Wei Ouyang
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
- Department of Medical Oncology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yixin Liu
- Department of Oncology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Jun Cao
- Department of Nephrology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Jinghua Chen
- Department of Medical Oncology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
26
|
Dai L, Gao T, Guo R, Chen Y, Wang J, Zhou S, Tang Y, Chen D, Huang S. Efficacy and safety of pyrotinib-based regimens in HER2 positive metastatic breast cancer: A retrospective real-world data study. Neoplasia 2024; 56:101029. [PMID: 39024777 PMCID: PMC11305273 DOI: 10.1016/j.neo.2024.101029] [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: 02/27/2024] [Revised: 04/18/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVE Pyrotinib is a novel irreversible tyrosine kinase inhibitor that has shown efficacy for human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC). This study explored the efficacy and safety of pyrotinib in the treatment of HER2-positive MBC patients in the real world. METHODS From September 2018 to February 2022, 137 female patients with HER2-positive MBC treated in this center were enrolled in this study. The follow-up period ended on January 12, 2023. The primary endpoint of this study was progression-free survival (PFS). Overall survival (OS), objective response rate (ORR), disease control rate (DCR), clinical benefit rate (CBR), central nervous system (CNS)-PFS, CNS-ORR, CNS-CBR, CNS-DCR, and adverse event (AE) were the secondary endpoints. RESULTS The ORR, DCR and CBR were 41.98 % (55/131), 87.79 % (115/131) and 44.27 % (58/131) in this cohort, respectively. The median PFS for this cohort was 10.37 months [95 % confidence interval (CI): 9.205-11.535] and the median OS was 37.53 months (not reached). Univariate and multivariate analyses showed that trastuzumab sensitivity was an independent predictor of improved PFS [hazard ratio (HR): 0.579 (0.371-0.904, p=0.016)] and improved OS [0.410 (0.213-0.790, p=0.008)]. Patients treated with a pyrotinib-based regimen as second-line and third-or-post-line therapy had poorer PFS [second-line: 3.315 (1.832-6.000, p<0.001); third-or-post-line: 3.304 (1.749-6.243, p<0.001)] and OS [second-line: 4.631 (1.033-20.771, p=0.045); third-or-post-line: 5.738 (1.212-27.174, p=0.028)]. There were 38 brain metastases (BM) patients in this study, the CNS-mPFS [14.37 months (7.815-20.925) vs. 7.83 months (7.047-8.613), p=0.375] and mOS [not reached vs. 36.40 months (18.551-54.249), p=0.034] were better in brain radiotherapy (BRT) group than NBRT group. 18.98 % (26/137) of patients experienced grade 3 or higher diarrhea. No AE-related death was reported. CONCLUSION This study confirms the promising antitumor activity and acceptable safety of real-world pyrotinib-based regimens for the treatment of HER2-positive MBC patients, particularly those who are trastuzumab-sensitive and who are receiving pyrotinib-based regimens as advanced first-line therapy. It has also been demonstrated that these regimens combined with BRT, provide better intracranial responses and long-term survival benefits for these patients with BM.
Collapse
Affiliation(s)
- Lanyi Dai
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China
| | - Ting Gao
- The Department of Thyroid and Breast Surgery, Dali Bai Autonomous Prefecture People's Hospital, China
| | - Rong Guo
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China
| | - Yuyuan Chen
- The Department of Thyroid and breast surgery, The affiliated Hospital of Ningbo University Medical College, China
| | - Jiankui Wang
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China
| | - Shaoqiang Zhou
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China
| | - Yiyin Tang
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China
| | - Dedian Chen
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China.
| | - Sheng Huang
- The 2nd Department of Breast Surgery, BC Centre, the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Building 3, No. 519 Kunzhou Road, Kunming 650118, China.
| |
Collapse
|
27
|
Gobbi H, Carvalho FM, Brot MD, Logullo AF, Silva CAM, Soares FA, Landeiro L, Rahal R, Barrios CH. Challenges in the evaluation of HER2 and HER2-low in breast cancer in Brazil and recommendations of a multidisciplinary working group. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240313. [PMID: 39356956 PMCID: PMC11444227 DOI: 10.1590/1806-9282.20240313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Helenice Gobbi
- Universidade Federal do Triângulo Mineiro, Discipline of Special Pathology – Uberaba (MG), Brazil
| | | | - Marina De Brot
- A.C.Camargo Cancer Center, Department of Pathological Anatomy – São Paulo (SP), Brazil
| | | | | | - Fernando Augusto Soares
- Universidade de São Paulo, Institute of Pathological Anatomy, School of Dentistry, D´Or São Luiz Network – São Paulo (SP), Brazil
| | | | - Rosemar Rahal
- Universidade Federal de Goiás, Department of Gynecology – Goiânia (GO), Brazil
| | - Carlos Henrique Barrios
- Latin American Cooperative Oncology Group and Oncoclínicas Group – Porto Alegre (RS), Brazil
| |
Collapse
|
28
|
Cheng D, Liu Z, Sun R, Jiang Y, Zeng Z, Zhao R, Mo Z. Overexpression of mir-489-3p inhibits proliferation and migration of non-small cell lung cancer cells by suppressing the HER2/PI3K/AKT/Snail signaling pathway. Heliyon 2024; 10:e35832. [PMID: 39224367 PMCID: PMC11367056 DOI: 10.1016/j.heliyon.2024.e35832] [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: 12/19/2023] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Background Lung cancer is a highly prevalent malignancy with significant morbidity and mortality rates. MiR-489-3p, a microRNA, has been identified as a regulator of tumor cell proliferation and invasion. Its expression is downregulated in non-small cell lung cancer (NSCLC). Elucidating the molecular mechanisms underlying miR-489-3p's role in NSCLC pathogenesis is crucial for identifying potential diagnostic and therapeutic targets. Methods To investigate the molecular mechanism of miR-489-3p in NSCLC, this study utilized A549, a commonly used NSCLC cell line. MiR-489-3p mimics and inhibitors were transfected into A549 cells. Additionally, co-transfection experiments using wortmannin, an inhibitor of the PI3K/AKT pathway, were performed. Expression of miR-489-3p and related proteins was analyzed by Western blotting and quantitative real-time PCR (qRT-PCR). Cell migration and proliferation were assessed by wound healing and colony formation assays, respectively. Results Overexpression of miR-489-3p significantly inhibited the proliferation and migration of A549 cells. This inhibitory effect was further enhanced upon co-transfected with wortmannin. Analysis of human lung specimens showed increased expression of HER2, PI3K, and AKT in lung adenocarcinoma tissues compared to adjacent non-cancerous tissues. Conclusions These findings suggest that miR-489-3p overexpression may inhibit NSCLC cell proliferation and migration by suppressing the HER2/PI3K/AKT/Snail signaling pathway. This study elucidates miR-489-3p's molecular mechanisms in NSCLC and provides experimental basis for identifying early diagnostic markers and novel therapeutic targets.
Collapse
Affiliation(s)
- Di Cheng
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Zhong Liu
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
- Joint Laboratory of Chronic Disease Prevention and Research in Guilin Medical University & Hunan Mingshun, Shaodong, 422800, China
| | - Renren Sun
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Yun Jiang
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Zhaoming Zeng
- Joint Laboratory of Chronic Disease Prevention and Research in Guilin Medical University & Hunan Mingshun, Shaodong, 422800, China
| | - Rui Zhao
- The First Affiliated Hospital of Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, Guangxi, China
- Joint Laboratory of Chronic Disease Prevention and Research in Guilin Medical University & Hunan Mingshun, Shaodong, 422800, China
| |
Collapse
|
29
|
Wang Y, Zeng R, Tian S, Chen S, Bi Z, Tang D, Knopp D. Bimetallic Single-Atom Nanozyme-Based Electrochemical-Photothermal Dual-Function Portable Immunoassay with Smartphone Imaging. Anal Chem 2024; 96:13663-13671. [PMID: 39126679 PMCID: PMC11339724 DOI: 10.1021/acs.analchem.4c02606] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Rapid and accurate detection of human epidermal growth factor receptor 2 (HER2) is crucial for the early diagnosis and prognosis of breast cancer. In this study, we reported an iron-manganese ion N-doped carbon single-atom catalyst (FeMn-NCetch/SAC) bimetallic peroxidase mimetic enzyme with abundant active sites etched by H2O2 and further demonstrated unique advantages of single-atom bimetallic nanozymes in generating hydroxyl radicals by density functional theory (DFT) calculations. As a proof of concept, a portable device-dependent electrochemical-photothermal bifunctional immunoassay detection platform was designed to achieve reliable detection of HER2. In the enzyme-linked reaction, H2O2 was generated by substrate catalysis via secondary antibody-labeled glucose oxidase (GOx), while FeMn-NCetch/SAC nanozymes catalyzed the decomposition of H2O2 to form OH*, which catalyzed the conversion of 3,3',5,5'-tetramethylbenzidine (TMB) to ox-TMB. The ox-TMB generation was converted from the colorimetric signals to electrical and photothermal signals by applied potential and laser irradiation, which could be employed for the quantitative detection of HER2. With the help of this bifunctional detection technology, HER2 was accurately detected in two ways: photothermally, with a linear scope of 0.01 to 2.0 ng mL-1 and a limit of detection (LOD) of 7.5 pg mL-1, and electrochemically, with a linear scope of 0.01 to 10 ng mL-1 at an LOD of 3.9 pg mL-1. By successfully avoiding environmental impacts, the bifunctional-based immunosensing strategy offers strong support for accurate clinical detection.
Collapse
Affiliation(s)
- Yunsen Wang
- Key
Laboratory of Analytical Science for Food Safety and Biology (MOE
& Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People’s Republic
of China
| | - Ruijin Zeng
- Key
Laboratory of Analytical Science for Food Safety and Biology (MOE
& Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People’s Republic
of China
| | - Shuo Tian
- Key
Laboratory of Analytical Science for Food Safety and Biology (MOE
& Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People’s Republic
of China
| | - Shuyun Chen
- Key
Laboratory of Analytical Science for Food Safety and Biology (MOE
& Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People’s Republic
of China
| | - Zhilan Bi
- Key
Laboratory of Analytical Science for Food Safety and Biology (MOE
& Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People’s Republic
of China
| | - Dianping Tang
- Key
Laboratory of Analytical Science for Food Safety and Biology (MOE
& Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou 350108, People’s Republic
of China
| | - Dietmar Knopp
- TUM
School of Natural Sciences, Department of Chemistry, Chair of Analytical
Chemistry and Water Chemistry, Technical
University Munich, Lichtenbergstrasse
4, Garching 85748, Germany
| |
Collapse
|
30
|
Forester E, Belsare A, Kim DW, Whitaker K, Obeid E, Goldstein LJ, Bleicher RJ, Daly MB, Williams AD. What Proportion of BRCA-Associated Breast Cancer Is Human Epidermal Growth Factor 2-Low and Eligible for Additional Targeted Therapy? J Surg Res 2024; 299:217-223. [PMID: 38776577 DOI: 10.1016/j.jss.2024.04.032] [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: 12/05/2023] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION DESTINY B04 provided clinical meaning to a new classification of human epidermal growth factor 2 (HER2) expression in breast cancer: HER2-low. Patients with germline breast cancer type 1 gene pathogenic variants (gBRCA1) often develop triple negative breast cancer (TNBC), but the proportion who could be classified as HER2-low and qualify for an additional targeted therapy option is unknown. This study aims to characterize the proportion of gBRCA1 or germline breast cancer type 2 gene pathogenic variants patients for whom these novel targeted therapies may be an option. METHODS We performed a retrospective chart review of patients with gBRCA1/2 treated at our institution for invasive breast cancer from 2000 to 2021. Synchronous or metachronous contralateral breast cancers were recorded separately. HER2 status was determined by immunohistochemistry and fluorescence in situ hybridization. We excluded patients without complete HER2 data. RESULTS Among the 95 breast cancers identified in our cohort of 85 gBRCA1/2 patients, 41 (43%) were TNBC, 38 (40%) were hormone receptor positive (HR+)/HER2-negative, and 16 (17%) were HER2-positive based on standard conventions. We found that 82% of the HR+/HER2-cancers and 66% of TNBCs would be reclassified as HER2-low. After stratifying by BRCA gene status, 64% of cancers in patients with gBRCA1 and 58% of cancers in patients with germline breast cancer type 2 gene pathogenic variants were HER2-low. CONCLUSIONS A significant portion of gBRCA1/2 patients who were previously diagnosed with TNBC or HR+/HER2- breast cancer would now be classified as HER2-low and could be considered for the use of trastuzumab deruxtecan in the metastatic setting. Outcome differences from therapy changes in this cohort should now be assessed.
Collapse
Affiliation(s)
- Emily Forester
- Division of Breast Surgical Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Rowan-Virtua School of Osteopathic Medicine, Stratford, New Jersey
| | - Aakash Belsare
- Division of Breast Surgical Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Dong Won Kim
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kristen Whitaker
- Department of Medical Oncology, MedStar Health, Washington, District of Columbia
| | - Elias Obeid
- Hackensack Meridian Health Network, Edison, New Jersey
| | - Lori J Goldstein
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Richard J Bleicher
- Division of Breast Surgical Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Austin D Williams
- Division of Breast Surgical Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
31
|
Schaffrin-Nabe D, Josten-Nabe A, Tannapfel A, Uhl W, Garmer M, Kurzrock R, Crook T, Limaye S, Schuster S, Patil D, Schaffrin M, Mokbel K, Voigtmann R. Dynamic changes in tumor profiling reveal intra- and inter-tumoral heterogeneity focused on an uncharacterized HER2 mutation: a case report of a young breast cancer patient. Front Oncol 2024; 14:1395618. [PMID: 38764581 PMCID: PMC11099277 DOI: 10.3389/fonc.2024.1395618] [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: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024] Open
Abstract
Despite multiple recent advances in systemic therapy for metastatic breast cancer, cases which display suboptimal response to guideline-driven treatment are frequently seen in the clinic. Effective options for such patients are limited, particularly in later line of therapy, and selection of optimal treatment options is essentially empirical and based largely on considerations of previous regimens received. Comprehensive cancer profiling includes detection of genetic alterations in tissue and circulating tumor DNA (ctDNA), immunohistochemistry (IHC) from re-biopsied metastatic disease, circulating tumor cells (CTCs), gene expression analysis and pharmacogenomics. The advent of this methodology and application to metastatic breast cancer, facilitates a more scientifically informed approach to identification of optimal systemic therapy approaches independent of the restrictions implied by clinical guidelines. Here we describe a case of metastatic breast cancer where consecutive comprehensive tumor profiling reveals ongoing tumor evolution, guiding the identification of novel effective therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St. Josef-Hospital, Bochum, Germany
| | | | - Razelle Kurzrock
- Medical College of Winconsin (MCW) Cancer Center, Froedtert Hospital & Medical College of Wisconsin, Milwaukee, WI, United States
| | - Timothy Crook
- Oncology Department, Cromwell Hospital, London, United Kingdom
| | - Sewanti Limaye
- Medical Oncology, Sir H.N. Reliance Foundation Hospital, Mumbai, India
| | | | | | | | - Kefah Mokbel
- London Breast Institute, Princess Grace Hospital, HCA Healthcare, London, United Kingdom
| | | |
Collapse
|
32
|
Katrini J, Boldrini L, Santoro C, Valenza C, Trapani D, Curigliano G. Biomarkers for Antibody-Drug Conjugates in Solid Tumors. Mol Cancer Ther 2024; 23:436-446. [PMID: 38363729 DOI: 10.1158/1535-7163.mct-23-0482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024]
Abstract
The clinical development and then the progressive entry in clinical practice of antibody-drug conjugates (ADC) have marked a transformative advancement in the overall cancer treatment. ADCs have been extensively tested for a large number of tumors, reporting heterogeneous clinical efficacy and safety results. In some diseases, the advent of ADCs has yielded significant changes in the prognostic trajectory, portending an improvement of the survival and/or quality of life. ADCs are targeted agents, capable of delivering highly cytotoxic payloads selectively to antigen-expressing cancer cells. As such, they have been intended as perfect "bullets" to enable the promise of precision medicine, toward high-efficacy and limited-toxicity treatment options. However, only some approved ADCs are intended for the use in biomarker-selected patient populations, restricting potentially the opportunity to be more precise. Yet, key characteristics of modern ADCs might allow the activity of ADCs in tumors with heterogeneous or low expression of cancer antigens, resulting in a clinical activity that could sublimate the classic paradigm of a drug-to-target perfect match. In our review, we portrayed the current landscape of approved ADCs, reporting data of activity as related to the expression of the cancer antigens, and elucidating possible determinants of the safety and efficacy, including when used in a therapeutic sequence.
Collapse
Affiliation(s)
- Jalissa Katrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Laura Boldrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Celeste Santoro
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
33
|
Baez-Navarro X, van Bockstal MR, Jager A, van Deurzen CHM. HER2-low breast cancer and response to neoadjuvant chemotherapy: a population-based cohort study. Pathology 2024; 56:334-342. [PMID: 38341307 DOI: 10.1016/j.pathol.2023.10.022] [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: 06/20/2023] [Revised: 10/09/2023] [Accepted: 10/29/2023] [Indexed: 02/12/2024]
Abstract
About half of breast cancers (BC) without amplification of the human epidermal growth factor receptor 2 (HER2) have a low HER2 protein expression level (HER2-low). The clinical impact of HER2-low and the response to neoadjuvant chemotherapy (NAC) is unclear. This study aimed to assess the association between HER2-low BC and pathological response to NAC. Data from the Dutch Pathology Registry were collected for 11,988 BC patients treated with NAC between 2014 and 2022. HER2-low BC was defined as an immunohistochemical score of 1+ or 2+ and a negative molecular reflex test. We compared clinicopathological features of HER2-0 versus HER2-low BC and assessed the correlation between HER2 status and the pathological complete response (pCR) rate after NAC, including overall survival. Among hormone receptor (HR)-positive tumours, 67% (n=4,619) were HER2-low, compared to 47% (n=1,167) in the HR-negative group. Around 32% (n=207) of patients had a discordant HER2 status between the pre-NAC biopsy and the corresponding post-NAC resection, within which 87% (n=165) changed from HER2-0 to HER2-low or vice versa. The pCR rate was significantly lower in HER2-low BC compared to HER2-0 BC within the HR-positive group (4% versus 5%; p=0.022). However, the absolute difference was limited, so the clinical relevance is questionable. In HR-negative cases, the difference in pCR was not significant (32% versus 34%; p=0.266). No significant difference in overall survival was observed between HER2-low and HER2-0 tumours, regardless of hormone receptor status. The antibody-drug conjugate trastuzumab deruxtecan (T-DXd) has improved survival outcomes of patients with HER2-low metastatic BC. The finding that one-third of the patients in this study had a discordant HER2 status between the pre-NAC biopsy and the post-NAC resection specimen could impact clinical decision-making should T-DXd be used in early BC treatment.
Collapse
Affiliation(s)
- Ximena Baez-Navarro
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | - Agnes Jager
- Department of Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
34
|
Syed RU, Afsar S, Aboshouk NAM, Salem Alanzi S, Abdalla RAH, Khalifa AAS, Enrera JA, Elafandy NM, Abdalla RAH, Ali OHH, Satheesh Kumar G, Alshammari MD. LncRNAs in necroptosis: Deciphering their role in cancer pathogenesis and therapy. Pathol Res Pract 2024; 256:155252. [PMID: 38479121 DOI: 10.1016/j.prp.2024.155252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
Necroptosis, a controlled type of cell death that is different from apoptosis, has become a key figure in the aetiology of cancer and offers a possible target for treatment. A growing number of biological activities, including necroptosis, have been linked to long noncoding RNAs (lncRNAs), a varied family of RNA molecules with limited capacity to code for proteins. The complex interactions between LncRNAs and important molecular effectors of necroptosis, including mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting protein kinase 3 (RIPK3), will be investigated. We will explore the many methods that LncRNAs use to affect necroptosis, including protein-protein interactions, transcriptional control, and post-transcriptional modification. Additionally, the deregulation of certain LncRNAs in different forms of cancer will be discussed, highlighting their dual function in influencing necroptotic processes as tumour suppressors and oncogenes. The goal of this study is to thoroughly examine the complex role that LncRNAs play in controlling necroptotic pathways and how that regulation affects the onset and spread of cancer. In the necroptosis for cancer treatment, this review will also provide insight into the possible therapeutic uses of targeting LncRNAs. Techniques utilising LncRNA-based medicines show promise in controlling necroptotic pathways to prevent cancer from spreading and improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India.
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | | | - Amna Abakar Suleiman Khalifa
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nancy Mohammad Elafandy
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Randa Abdeen Husien Abdalla
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Omar Hafiz Haj Ali
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - G Satheesh Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| |
Collapse
|
35
|
Curigliano G, Dent R, Earle H, Modi S, Tarantino P, Viale G, Tolaney SM. Open questions, current challenges, and future perspectives in targeting human epidermal growth factor receptor 2-low breast cancer. ESMO Open 2024; 9:102989. [PMID: 38613914 PMCID: PMC11024577 DOI: 10.1016/j.esmoop.2024.102989] [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: 12/07/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 04/15/2024] Open
Abstract
Approximately 60% of traditionally defined human epidermal growth factor receptor 2 (HER2)-negative breast cancers express low levels of HER2 [HER2-low; defined as immunohistochemistry (IHC) 1+ or IHC 2+/in situ hybridization (ISH)-]. HER2-low breast cancers encompass a large percentage of both hormone receptor-positive (up to 85%) and triple-negative (up to 63%) breast cancers. The DESTINY-Breast04 trial established that HER2-low tumors are targetable, leading to the approval of trastuzumab deruxtecan (T-DXd) as the first HER2-directed therapy for the treatment of HER2-low breast cancer in the United States and Europe. This change in the clinical landscape results in a number of questions and challenges-including those related to HER2 assessment and patient identification-and highlights the need for careful assessment of HER2 expression to identify patients eligible for T-DXd. This review provides context for understanding how to identify patients with HER2-low breast cancer with respect to sample types, scoring and reporting HER2 status, and testing methods and assays. It also discusses management of important T-DXd-related adverse events. Available evidence supports the efficacy of T-DXd in patients with any history of IHC 1+ or IHC 2+/ISH- scores; however, future research may further refine the population who could benefit from T-DXd or other HER2-directed therapies and identify novel methods for patient identification. Because HER2 expression can change with disease progression or treatment, and variability exists in scoring and interpretation of HER2 status, careful re-evaluation in certain scenarios may help to identify more patients who may benefit from T-DXd.
Collapse
Affiliation(s)
- G Curigliano
- European Institute of Oncology, IRCCS, Milan; Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy.
| | - R Dent
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - H Earle
- Blogger at hannahincancerland.com, New Hampshire, USA; Patient at Dana-Farber Cancer Institute, Harvard Medical School, Boston
| | - S Modi
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - P Tarantino
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - G Viale
- European Institute of Oncology, IRCCS, Milan
| | - S M Tolaney
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Nobbe K, Erices-Leclercq M, Foerster F, Förster R, Baldus SE, Rudlowski C, Schröder L, Lubig S. HER2 Low Expression in Primary Male Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:141-148. [PMID: 38562651 PMCID: PMC10984208 DOI: 10.2147/bctt.s450682] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Purpose The introduction of HER2-targeting antibody drug conjugates (ADCs) offers new treatment options for female breast cancer patients (FBC) expressing low levels of HER2 (HER2 low). No evidence was found that HER2 low describes a new FBC subtype. There is a lack of studies determining the impact of HER2 low in male breast cancer (MBC). In this study, we evaluate the prevalence of HER2 low in primary MBC and correlate the results with patient characteristics. Patients and Methods In this study, histological specimens were obtained from 120 male patients diagnosed and treated for primary invasive breast cancer from 1995 to 2022 at Breast Cancer Units in Bergisch Gladbach, Chemnitz, and Zwickau, Germany. HER2 immunostaining and in situ hybridization were performed by central pathology and evaluated based on the ASCO/CAP guidelines. The correlation of expression of HER2 low with tumor biological characteristics and patient outcomes was investigated. Results Out of all cases, four patients (3.3%) showed HER2 positivity (3+), 39 (32.5%) patients were classified as HER2 low, 7 (5.8%) were HER2 2+ (no amplification), 32 (26.7%) were HER2 1+, and 77 (64.2%) were classified as HER2 zero. Out of 77 HER2 zero cases, 47 tumors (61.0%) showed incomplete staining, with <10% of tumor cells classified as HER2 ultralow. No statistical correlation between HER2 low and tumor biological characteristics and patients' survival was found. Conclusion Our findings show a notable, albeit lower, prevalence of HER2 low expression in primary MBC. However, tumors expressing HER2 low do not show specific tumor biological features to define a new breast cancer subtype in MBC. Our results suggest that a significant number of MBC patients could benefit from ADCs, as shown in FBC. Further studies are required to better understand HER2 low breast cancer, both generally and in MBC.
Collapse
Affiliation(s)
- Katleen Nobbe
- Breast Unit, Lutheran Hospital Bergisch Gladbach, Bergisch Gladbach, Germany
| | | | - Frank Foerster
- Department of Economical Sciences, University of Applied Sciences, Zwickau, Germany
- Outpatient Department of Gynaecological Oncology and Palliative Care, Chemnitz, Germany
| | - Robert Förster
- Institute for Radiation Oncology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Stephan E Baldus
- Institute for Pathology, Cytology and Molecular Pathology, Bergisch Gladbach, Germany
| | - Christian Rudlowski
- Breast Unit, Lutheran Hospital Bergisch Gladbach, Bergisch Gladbach, Germany
- Department of Gynecology, University Hospital Bonn, Bonn, Germany
| | - Lars Schröder
- Department of Gynaecology and Obstetrics, Ketteler Krankenhaus, Offenbach, Germany
| | - Sabine Lubig
- Breast Unit, Lutheran Hospital Bergisch Gladbach, Bergisch Gladbach, Germany
| |
Collapse
|
37
|
Baez-Navarro X, van den Ende NS, Nguyen AH, Sinke R, Westenend P, van Brakel JB, Stobbe C, Westerga J, van Deurzen CHM. HER2-low and tumor infiltrating lymphocytes in triple-negative breast cancer: Are they connected? Breast Cancer Res 2024; 26:41. [PMID: 38468323 PMCID: PMC10926638 DOI: 10.1186/s13058-024-01783-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/11/2024] [Indexed: 03/13/2024] Open
Abstract
Most patients with triple-negative breast cancer (TNBC) are not candidates for targeted therapy, leaving chemotherapy as the primary treatment option. Recently, immunotherapy has demonstrated promising results in TNBC, due to its immunogenicity. In addition, a novel antibody-drug conjugate, namely, trastuzumab-deruxtecan, has shown effectiveness in TNBC patients with low-HER2 expression (HER2-low). These novel treatment options raise the question about the potential association between the density of stromal tumor-infiltrating lymphocytes (sTILs) and the level of HER2 expression. We aimed to evaluate the association between the level of HER2 expression (HER2-low versus HER2-0) and density of sTILs in TNBC patients, and how they impact the response to neoadjuvant chemotherapy (NAC). This was a retrospective multicenter study including all TNBC patients diagnosed between 2018 and 2022. Central pathology review included sTILs percentages and level of HER2 expression. Tumors were reclassified as either HER2-0 (HER2 IHC 0) or HER2-low (IHC 1 + or 2 + with negative reflex test). Various clinicopathologic characteristics, including sTILs density, and response to NAC were compared between HER2-0 and HER2-low cases. In total, 753 TNBC patients were included in this study, of which 292 patients received NAC. Interobserver agreement between the original pathology report and central review was moderate (77% had the same IHC status after reclassification in either HER2-0 or HER2-low; k = 0.45). HER2-low TNBC represented about one third (36%) of the tumors. No significant difference in sTILs density or complete pathologic response rate was found between HER2-0 and HER2-low cases (p = 0.476 and p = 0.339, respectively). The density of sTILs (≥ 10% sTILs vs. < 10%) was independently associated with achieving a pCR (p = 0.011). In conclusion, no significant association was found between HER2-low status and density of sTILs nor response to NAC. Nonetheless, sTILs could be an independent biomarker for predicting NAC response in TNBC patients.
Collapse
Affiliation(s)
- Ximena Baez-Navarro
- Department of Pathology, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands.
| | - Nadine S van den Ende
- Department of Pathology, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Anh H Nguyen
- Department of Pathology, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
- Department of Pathology, HMC, The Hague, The Netherlands
| | - Renata Sinke
- Department of Pathology, Pathan B.V., Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Pieter Westenend
- Laboratory of Pathology, PAL Dordrecht, Dordrecht, The Netherlands
| | | | - Claudia Stobbe
- Department of Pathology, Pathan B.V., Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Johan Westerga
- Department of Pathology, Pathan B.V., Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | | |
Collapse
|
38
|
Zhao L, Xing Y, Liu C, Ma S, Huang W, Cheng Z, Zhao J. Detection of HER2 expression using 99mTc-NM-02 nanobody in patients with breast cancer: a non-randomized, non-blinded clinical trial. Breast Cancer Res 2024; 26:40. [PMID: 38459598 PMCID: PMC10924314 DOI: 10.1186/s13058-024-01803-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: 01/27/2023] [Accepted: 03/03/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND 99mTc radiolabeled nanobody NM-02 (99mTc-NM-02) is a novel single photon emission computed tomography (SPECT) probe with a high affinity and specificity for human epidermal growth factor receptor 2 (HER2). In this study, a clinical imaging trial was conducted to investigate the relationship between 99mTc-NM-02 uptake and HER2 expression in patients with breast cancer. METHODS Thirty patients with pathologically confirmed breast cancer were recruited and imaged with both 99mTc-NM-02 SPECT/computed tomography (CT) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/CT. According to the treatment conditions before recruitment, patients were divided into two groups, the newly diagnosed group (n = 24) and the treated group (n = 6). The maximal standard uptake value (SUVmax) of 18F-FDG and SUVmax and mean SUV (SUVmean) of 99mTc-NM-02 in the lesions were determined to analyze the relationship with HER2 expression. RESULTS No meaningful relationship was observed between 18F-FDG uptake and HER2 expression in 30 patients with breast cancer. 99mTc-NM-02 uptake was positively correlated with HER2 expression in the newly diagnosed group, but no correlation was observed in the treated group. 99mTc-NM-02 uptake in HER2-positive lesions was lower in those with effective HER2-targeted therapy compared with the newly diagnosed group. 99mTc-NM-02 SPECT/CT detected brain and bone metastases of breast cancer with a different imaging pattern from 18F-FDG PET/CT. 99mTc-NM-02 showed no non-specific uptake in inflamed tissues and revealed intra- and intertumoral HER2 heterogeneity by SPECT/CT imaging in 9 of the 30 patients with breast cancer. CONCLUSIONS 99mTc-NM-02 SPECT/CT has the potential for visualizing whole-body HER2 overexpression in untreated patients, making it a promising method for HER2 assessment in patients with breast cancer. TRIAL REGISTRATION NCT04674722, Date of registration: December 19, 2020.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Changcun Liu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Shaofei Ma
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Hongkou District, Shanghai, 200080, China
| | - Wenhua Huang
- Nanomab Technology Limited, No. 333, North Chengdu Road, Jingan District, Shanghai, 200041, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555, Zuchongzhi Road, Pudong New District, Shanghai, 201203, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, No. 198, Binhai East Road, High-Tech District, Yantai, 264000, Shandong, China.
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Road, Hongkou District, Shanghai, 200080, China.
| |
Collapse
|
39
|
Tanei T, Seno S, Sota Y, Hatano T, Kitahara Y, Abe K, Masunaga N, Tsukabe M, Yoshinami T, Miyake T, Shimoda M, Matsuda H, Shimazu K. High HER2 Intratumoral Heterogeneity Is a Predictive Factor for Poor Prognosis in Early-Stage and Locally Advanced HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:1062. [PMID: 38473420 DOI: 10.3390/cancers16051062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
PURPOSE Breast cancer tumors frequently have intratumoral heterogeneity (ITH). Tumors with high ITH cause therapeutic resistance and have human epidermal growth factor receptor 2 (HER2) heterogeneity in response to HER2-targeted therapies. This study aimed to investigate whether high HER2 heterogeneity levels were clinically related to a poor prognosis for HER2-targeted adjuvant therapy resistance in primary breast cancers. METHODS This study included patients with primary breast cancer (n = 251) treated with adjuvant HER2-targeted therapies. HER2 heterogeneity was manifested by the shape of HER2 fluorescence in situ hybridization amplification (FISH) distributed histograms with the HER2 gene copy number within a tumor sample. Each tumor was classified into a biphasic grade graph (high heterogeneity [HH]) group or a monophasic grade graph (low heterogeneity [LH]) group based on heterogeneity. Both groups were evaluated for disease-free survival (DFS) and overall survival (OS) for a median of ten years of annual follow-up. RESULTS Of 251 patients with HER2-positive breast cancer, 46 (18.3%) and 205 (81.7%) were classified into the HH and LH groups, respectively. The HH group had more distant metastases and a poorer prognosis than the LH group (DFS: p < 0.001 (HH:63% vs. LH:91% at 10 years) and for the OS: p = 0.012 (HH:78% vs. LH:95% at 10 years). CONCLUSIONS High HER2 heterogeneity is a poor prognostic factor in patients with HER2-positive breast cancer. A novel approach to heterogeneity, which is manifested by the shape of HER2 FISH distributions, might be clinically useful in the prognosis prediction of patients after HER2 adjuvant therapy.
Collapse
Affiliation(s)
- Tomonori Tanei
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Takaaki Hatano
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Yuri Kitahara
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Kaori Abe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masami Tsukabe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
40
|
Valenza C, Guidi L, Battaiotto E, Trapani D, Sartore Bianchi A, Siena S, Curigliano G. Targeting HER2 heterogeneity in breast and gastrointestinal cancers. Trends Cancer 2024; 10:113-123. [PMID: 38008666 DOI: 10.1016/j.trecan.2023.11.001] [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: 09/18/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023]
Abstract
About 20% of breast and gastric cancers and 3% of colorectal carcinomas overexpress the human epidermal growth factor receptor 2 (HER2) and are sensitive to HER2-directed agents. The expression of HER2 may differ within the same tumoral lesion (spatial intralesional heterogeneity), from different tumor locations (spatial interlesional heterogeneity), and throughout treatments (temporal heterogeneity). Spatial and temporal heterogeneity may impact on response and resistance to HER2-targeting agents and its prevalence and predictive role changes across HER2-overexpressing solid tumors. Therefore, the definition and the characterization of HER2 heterogeneity pose many challenges and its implementation as a reproducible predictive biomarker would help in guiding treatment modulation.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Battaiotto
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Andrea Sartore Bianchi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
41
|
Gondry O, Caveliers V, Xavier C, Raes L, Vanhoeij M, Verfaillie G, Fontaine C, Glorieus K, De Grève J, Joris S, Luyten I, Zwaenepoel K, Vandenbroucke F, Waelput W, Thyparambil S, Vaneycken I, Cousaert J, Bourgeois S, Devoogdt N, Goethals L, Everaert H, De Geeter F, Lahoutte T, Keyaerts M. Phase II Trial Assessing the Repeatability and Tumor Uptake of [ 68Ga]Ga-HER2 Single-Domain Antibody PET/CT in Patients with Breast Carcinoma. J Nucl Med 2024; 65:178-184. [PMID: 38302159 PMCID: PMC10858381 DOI: 10.2967/jnumed.123.266254] [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: 07/05/2023] [Revised: 11/03/2023] [Indexed: 02/03/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) status is used for decision-making in breast carcinoma treatment. The status is obtained through immunohistochemistry or in situ hybridization. These two methods have the disadvantage of necessitating tissue sampling, which is prone to error due to tumor heterogeneity or interobserver variability. Whole-body imaging might be a solution to map HER2 expression throughout the body. Methods: Twenty patients with locally advanced or metastatic breast carcinoma (5 HER2-positive and 15 HER2-negative patients) were included in this phase II trial to assess the repeatability of uptake quantification and the extended safety of the [68Ga]Ga-NOTA-anti-HER2 single-domain antibody (sdAb). The tracer was injected, followed by a PET/CT scan at 90 min. Within 8 d, the procedure was repeated. Blood samples were taken for antidrug antibody (ADA) assessment and liquid biopsies. On available tissues, immunohistochemistry, in situ hybridization, and mass spectrometry were performed to determine the correlation of HER2 status with uptake values measured on PET. If relevant preexisting [18F]FDG PET/CT images were available (performed as standard of care), a comparison was made. Results: With a repeatability coefficient of 21.8%, this imaging technique was repeatable. No clear correlation between PET/CT uptake values and pathology could be established, as even patients with low levels of HER2 expression showed moderate to high uptake. Comparison with [18F]FDG PET/CT in 16 patients demonstrated that in 7 patients, [68Ga]Ga-NOTA-anti-HER2 shows interlesional heterogeneity within the same patient, and [18F]FDG uptake did not show the same heterogeneous uptake in all patients. In some patients, the extent of disease was clearer with the [68Ga]Ga-NOTA-anti-HER2-sdAb. Sixteen adverse events were reported but all without a clear relationship to the tracer. Three patients with preexisting ADAs did not show adverse reactions. No new ADAs developed. Conclusion: [68Ga]Ga-NOTA-anti-HER2-sdAb PET/CT imaging shows similar repeatability to [18F]FDG. It is safe for clinical use. There is tracer uptake in cancer lesions, even in patients previously determined to be HER2-low or -negative. The tracer shows potential in the assessment of interlesional heterogeneity of HER2 expression. In a subset of patients, [68Ga]Ga-NOTA-anti-HER2-sdAb uptake was seen in lesions with no or low [18F]FDG uptake. These findings support further clinical development of [68Ga]Ga-NOTA-anti-HER2-sdAb as a PET/CT tracer in breast cancer patients.
Collapse
Affiliation(s)
- Odrade Gondry
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Vicky Caveliers
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Catarina Xavier
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurens Raes
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marian Vanhoeij
- Department of Surgical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Guy Verfaillie
- Department of Surgical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christel Fontaine
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Katrien Glorieus
- Department of Surgical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jacques De Grève
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sofie Joris
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ine Luyten
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Karen Zwaenepoel
- Centre for Oncological Research, University of Antwerp, Wilrijk, Belgium
| | | | - Wim Waelput
- Department of Pathology, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Experimental Pathology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Ilse Vaneycken
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Julie Cousaert
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sophie Bourgeois
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lode Goethals
- Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Hendrik Everaert
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Frank De Geeter
- Department of Nuclear Medicine, Algemeen Ziekenhuis Sint-Jan Brugge Oostende, Bruges, Belgium
| | - Tony Lahoutte
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium;
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
42
|
Wang Y, Chen S, Tian S, Wei Q, Tang D. Edge-generated N-doped carbon-supported dual-metal active sites for enhancing electrochemical immunoassay. Anal Chim Acta 2023; 1284:342006. [PMID: 37996161 DOI: 10.1016/j.aca.2023.342006] [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: 09/21/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023]
Abstract
Development of high-precision human epidermal growth factor receptor 2 (HER2) assay is essential for the early diagnostic and prevention of breast cancer. In this work, an innovative Fe/Mn bimetallic nanozyme at the edge of N-doped carbon defects (FeMn-NCedge) with abundant active sites was prepared through the hydrothermal synthetic method. FeMn-NCedge nanozyme displayed excellent peroxidase-like activity relative to the H2O2-catalyzed 3,3',5,5'-tetramethylbenzidine (TMB) system for generation of the oxidized TMB (oxTMB). As a proof-of-concept application, we constructed an electrochemical immunoassay for the detection of HER2 based on the unique merits of FeMn-NCedge. Initially, a sandwiched immunoreaction was carried out in the microtiter plate coated with monoclonal anti-HER2 capture antibodies using glucose oxidase (GOx)-labeled anti-HER2 as detection antibody. The carried GOx could catalyze glucose to produce H2O2, thus resulting in the formation of oxTMB with the assistance of TMB and FeMn-NCedge nanozyme. The produced oxTMB could be determined on the electrode by the chronoamperometry at an applied potential of +10 mV. Experimental results revealed that the steady-state current increased with the increasing HER2 concentration in the sample, and gave a good linear relationship within the dynamic range of 0.01-10 ng/mL at a limit of detection of 5.4 pg/mL HER2. In addition, good reproducibility, high specificity and acceptable accuracy were acquired for the measurement of human serum samples. Importantly, this method can be extended for quantitative monitoring other disease-related proteins by changing the corresponding antibodies.
Collapse
Affiliation(s)
- Yunsen Wang
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Shuyun Chen
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Shuo Tian
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Qiaohua Wei
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China.
| | - Dianping Tang
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China.
| |
Collapse
|
43
|
Nicolò E, Serafini MS, Munoz-Arcos L, Pontolillo L, Molteni E, Bayou N, Andreopoulou E, Curigliano G, Reduzzi C, Cristofanilli M. Real-time assessment of HER2 status in circulating tumor cells of breast cancer patients: Methods of detection and clinical implications. THE JOURNAL OF LIQUID BIOPSY 2023; 2:100117. [PMID: 40028485 PMCID: PMC11863949 DOI: 10.1016/j.jlb.2023.100117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/01/2023] [Indexed: 03/05/2025]
Abstract
The human epidermal growth factor receptor 2 (HER2) plays a central role in breast cancer (BC). Therefore, it is critical to develop a method that can capture its spatial and temporal heterogeneity. Nowadays, therapeutic decisions for BC patients relies on evaluation of HER2 status from tissue biopsies using immunohistochemistry and in situ hybridization. Nevertheless, considering the technical and logistical challenges associated with tissue biopsies, there is an unmet need for a non-invasive and accurate approach to obtain real-time assessment of HER2 status. In this context, circulating biomarkers, particularly circulating tumor cells (CTCs), emerged as promising candidates. HER2 assessment on CTCs can be performed at genomic, transcriptomic, and protein levels on both bulk CTCs and at the single-cell resolution. However, the main limitation of the literature to date is the lack of a consistent definition of HER2-positive CTCs, which poses a major challenge for both, future research and clinical applications. Several studies revealed discordance in HER2 status between the primary tumor and corresponding CTCs. For instance, HER2-positive CTCs have been detected among patients with HER2-negative BC and vice versa. As a result, researchers have evaluated the prognostic and predictive value of HER2 status in CTCs, both in the early and metastatic settings, to increase the possibility of using anti-HER2 therapy also for these patients and to dissect mechanisms of treatment resistance. This review aims to provide an overview of the methods to determine HER2 status in CTCs and to summarize the evidence and future perspective on how CTCs-HER2 assessment can be integrated into the clinical management of BC patients.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Mara Serena Serafini
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Laura Munoz-Arcos
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Letizia Pontolillo
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Medical Oncology Department, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Elisabetta Molteni
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, University of Udine, Via Chiusaforte, Udine, Italy
| | - Nadia Bayou
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, 2092, Tunisia
| | - Eleni Andreopoulou
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
| | - Carolina Reduzzi
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
44
|
Dai L, Huang Q, Guo R, Zhu K, Tang Y, Chen D, Huang S. Clinicopathologic Features and Prognosis of Female Early Breast Cancer With HER2 Low Expression: A Propensity Score Matched Analysis. Clin Med Insights Oncol 2023; 17:11795549231202463. [PMID: 38023289 PMCID: PMC10676074 DOI: 10.1177/11795549231202463] [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: 03/15/2023] [Accepted: 09/04/2023] [Indexed: 12/01/2023] Open
Abstract
Background Metastatic breast cancer (MBC) patients with low expression of human epidermal growth factor 2 (HER2) have been proven to benefit from HER2 targeted therapy. We aimed to determine how HER2-low status affected survival and metastatic risk as well as how it affected pathological complete response (pCR) in neoadjuvant chemotherapy (NAC) patients. Methods According to the results of immunohistochemistry (IHC) and in situ hybridization (ISH) testing, 321 female patients were sorted into HER2-low (IHC 1+/2+ with ISH negative) and HER2-zero (IHC 0) groups using propensity score matching (PSM). Overall survival (OS), disease-free survival (DFS), and distant disease-free survival (DDFS) were compared for both groups, while pCR was only analyzed for NAC patients. Results In total, 97 patients in each group after PSM were included. We discovered that pCR was not associated with HER2 expression status in 45 patients who underwent NAC. Five-year OS in the HER2-low group was significantly higher (98.99%) than in the HER2-zero group (95.87%, P = .044); however, this difference was not reflected in the 5-year DFS (90.61 vs 90.52%, P = .868) and 5-year DDFS (93.67 vs 91.53%, P = .757). Meanwhile, multivariate analysis revealed that HER2-low expression could indicate better OS (P = .047, hazard ratios [HRs] = 16.121, 95% confidence interval [CI] = 1.035-251.046), but it had no prognostic value for DFS or DDFS. Conclusion When compared with HER2-zero expression, HER2-low expression was not connected to pCR and could not modify metastasis risk in female patients with early-stage breast cancer (BC), but it may prolong patient survival.
Collapse
Affiliation(s)
| | | | | | - Keying Zhu
- The 2nd Department of Breast Surgery, Breast Cancer Centre, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunzhou, China
| | - Yiyin Tang
- The 2nd Department of Breast Surgery, Breast Cancer Centre, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunzhou, China
| | - Dedian Chen
- The 2nd Department of Breast Surgery, Breast Cancer Centre, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunzhou, China
| | - Sheng Huang
- The 2nd Department of Breast Surgery, Breast Cancer Centre, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunzhou, China
| |
Collapse
|
45
|
Yang Y, Sun Q, Deng Z, Shi W, Cheng H. Cbl induced ubiquitination of HER2 mediate immune escape from HER2-targeted CAR-T. J Biochem Mol Toxicol 2023; 37:e23446. [PMID: 37354072 DOI: 10.1002/jbt.23446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/12/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
Breast cancer (BC) with high HER2 expression has higher recurrence rate and worse prognosis, and its immunotherapy is promising. Based on the high expression of HER2, develop Chimeric Antigen Receptor T-cell (CAR-T) and PDL-1 immunotherapy, and study the molecular pathways of related immune cells and recurrence. HER2-CAR-T cells were constructed using retroviruses, and their specific recognition and immune effects on HER2+ BC cells were verified by in vivo and in vitro experiments. PDL-1 was used as adjuvant immunotherapy, knocking down PDL-1 in tumor cells or dendritic cells, or depleted macrophages to study immune pathways. The negative regulation of HER2 by cbl was determined by IP, ubiquitination experiments, and segmented plasmids, elucidating the molecular mechanism of HER2+ BC recurrence after immunotherapy. HER2-CAR-T specifically recognizes HER2-positive tumor cells and inhibits tumor growth in vivo and in vitro, and anti-PDL1 treatment enhances the therapeutic effect of HER2-CAR-T on tumors. HER2-CART therapy eradicated solid tumors after PDL1 knockdown in dendritic cells. Immunotherapy of relapsed tumors lost HER2 expression by upregulating cbl. HER2-CAR-T shows specific recognition of HER2+ cells and can mediate immune response therapy with the cooperation of PDL-1.
Collapse
Affiliation(s)
- Yanqiu Yang
- Department of Ultrasonography, The Fifth People's Hospital of Qinghai Province, Qinghai, China
| | - Qingqing Sun
- The Third Department of Convalescence, Lintong Rehabilitation and Recuperation Center, Xi'an, China
| | - Zhiping Deng
- Department of Breast Surgery, Shaanxi Cancer Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Wencong Shi
- Department of Breast Surgery, Shaanxi Cancer Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Hao Cheng
- Department of Ultrasonography, Shaanxi Cancer Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
46
|
Quidute P, Quidute R, Perez MM, Pereira EC, da Veiga GL, Alves BDCA, Fonseca FLA. Survivin expression as a prognostic marker for breast cancer. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230167. [PMID: 37729357 PMCID: PMC10508893 DOI: 10.1590/1806-9282.20230167] [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: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 09/22/2023]
Abstract
OBJECTIVE Due to the speed of development observed in breast cancer, several studies aimed at discovering new biomarkers have been carried out in order to arrive at an early diagnosis. As survivin plays a fundamental role in the evasion of apoptosis in tumor cells, the aim of this study was to verify the expression profile of the survivin gene in paraffin-embedded breast tumor samples and associate it with the clinical characteristics of the patients. METHODS This is a cross-sectional study, for which 100 tumor samples were obtained from cancer patients treated throughout the year 2019 at Instituto de Mama do Cariri (Juazeiro do Norte, in the state of Ceará). This study included women over 30 years old who had confirmed breast cancer through anatomopathological examination but excluded those with non-neoplastic breast comorbidities, other neoplasms, or chronic diseases. Survivin gene expression was assessed by quantitative polymerase chain reaction. RESULTS The expression of survivin is associated with the lack of expression of estrogen (p=0.027) and progesterone (p>0.0005) receptors. It means that survivin expression is higher in patients in which labeling was absent for estrogen receptor and progesterone receptor. CONCLUSION Our data reinforce that survivin expression is higher in estrogen receptor-patients, thus representing an additional prognostic tool.
Collapse
Affiliation(s)
- Patricia Quidute
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | - Ricardo Quidute
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | - Matheus Moreira Perez
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | | | - Glaucia Luciano da Veiga
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | | | - Fernando Luiz Affonso Fonseca
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
- Universidade Federal de São Paulo, Instituto de Ciências Farmacêuticas – Diadema (SP), Brazil
| |
Collapse
|
47
|
Shiino S, Tokura M, Nakayama J, Yoshida M, Suto A, Yamamoto Y. Investigation of Tumor Heterogeneity Using Integrated Single-Cell RNA Sequence Analysis to Focus on Genes Related to Breast Cancer-, EMT-, CSC-, and Metastasis-Related Markers in Patients with HER2-Positive Breast Cancer. Cells 2023; 12:2286. [PMID: 37759508 PMCID: PMC10527746 DOI: 10.3390/cells12182286] [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: 08/23/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) protein, which is characterized by the amplification of ERBB2, is a molecular target for HER2-overexpressing breast cancer. Many targeted HER2 strategies have been well developed thus far. Furthermore, intratumoral heterogeneity in HER2 cases has been observed with immunohistochemical staining and has been considered one of the reasons for drug resistance. Therefore, we conducted an integrated analysis of the breast cancer single-cell gene expression data for HER2-positive breast cancer cases from both scRNA-seq data from public datasets and data from our cohort and compared them with those for luminal breast cancer datasets. In our results, heterogeneous distribution of the expression of breast cancer-related genes (ESR1, PGR, ERBB2, and MKI67) was observed. Various gene expression levels differed at the single-cell level between the ERBB2-high group and ERBB2-low group. Moreover, molecular functions and ERBB2 expression levels differed between estrogen receptor (ER)-positive and ER-negative HER2 cases. Additionally, the gene expression levels of typical breast cancer-, CSC-, EMT-, and metastasis-related markers were also different across each patient. These results suggest that diversity in gene expression could occur not only in the presence of ERBB2 expression and ER status but also in the molecular characteristics of each patient.
Collapse
Affiliation(s)
- Sho Shiino
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Momoko Tokura
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; (M.T.); (J.N.)
| | - Jun Nakayama
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; (M.T.); (J.N.)
| | - Masayuki Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Akihiko Suto
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Yusuke Yamamoto
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo 104-0045, Japan; (M.T.); (J.N.)
| |
Collapse
|
48
|
Santana MDFM, Sawada MIBAC, Santos AS, Reis M, Xavier J, Côrrea-Giannella ML, Hirata AHDL, Gebrim LH, Soriano FG, Camacho CP, Passarelli M. Increased Expression of miR-223-3p and miR-375-3p and Anti-Inflammatory Activity in HDL of Newly Diagnosed Women in Advanced Stages of Breast Cancer. Int J Mol Sci 2023; 24:12762. [PMID: 37628945 PMCID: PMC10454463 DOI: 10.3390/ijms241612762] [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: 07/04/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The expression of inflammation-related miRs bound to high-density lipoproteins (HDLs), the anti-inflammatory activity of HDLs isolated from individuals with breast cancer, and controls were determined. Forty newly diagnosed women with breast cancer naïve of treatment and 10 control participants were included. Cholesterol-loaded bone-marrow-derived macrophages were incubated with HDL from both groups and challenged with lipopolysaccharide (LPS). Interleukin 6 (IL6) and tumor necrosis factor (TNF) in the medium were quantified. The miRs in HDLs were determined by RT-qPCR. Age, body mass index, menopausal status, plasma lipids, and HDL composition were similar between groups. The ability of HDL to inhibit IL6 and TNF production was higher in breast cancer compared to controls, especially in advanced stages of the disease. The miR-223-3p and 375-3p were higher in the HDLs of breast cancer independent of the histological type of the tumor and had a high discriminatory power between breast cancer and controls. The miR-375-3p was greater in the advanced stages of the disease and was inversely correlated with the secretion of inflammatory cytokines. Inflammation-related miRs and the anti-inflammatory role of HDLs may have a significant impact on breast cancer pathophysiology.
Collapse
Affiliation(s)
- Monique de Fatima Mello Santana
- Laboratório de Lípides (LIM 10), Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil;
| | - Maria Isabela Bloise Alves Caldas Sawada
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01525-000, Brazil; (M.I.B.A.C.S.); (M.R.); (J.X.); (C.P.C.)
- Hospital da Força Aérea de São Paulo, São Paulo 02012-021, Brazil
| | - Aritania Sousa Santos
- Laboratório de Carboidratos e Radioimunoensaio (LIM 18), Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil; (A.S.S.); (M.L.C.-G.); (A.H.d.L.H.)
| | - Mozania Reis
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01525-000, Brazil; (M.I.B.A.C.S.); (M.R.); (J.X.); (C.P.C.)
- Unidade Básica de Saúde Dra. Ilza Weltman Hutzler, São Paulo 02472-180, Brazil
| | - Jacira Xavier
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01525-000, Brazil; (M.I.B.A.C.S.); (M.R.); (J.X.); (C.P.C.)
- Unidade Básica de Saúde Dra. Ilza Weltman Hutzler, São Paulo 02472-180, Brazil
| | - Maria Lúcia Côrrea-Giannella
- Laboratório de Carboidratos e Radioimunoensaio (LIM 18), Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil; (A.S.S.); (M.L.C.-G.); (A.H.d.L.H.)
| | - Andrea Harumy de Lima Hirata
- Laboratório de Carboidratos e Radioimunoensaio (LIM 18), Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil; (A.S.S.); (M.L.C.-G.); (A.H.d.L.H.)
| | - Luiz Henrique Gebrim
- Centro de Referência da Saúde da Mulher–Hospital Pérola Byington, São Paulo 01215-000, Brazil;
| | - Francisco Garcia Soriano
- Laboratório de Emergências Clínicas (LIM 51), Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil;
| | - Cleber Pinto Camacho
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01525-000, Brazil; (M.I.B.A.C.S.); (M.R.); (J.X.); (C.P.C.)
| | - Marisa Passarelli
- Laboratório de Lípides (LIM 10), Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil;
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01525-000, Brazil; (M.I.B.A.C.S.); (M.R.); (J.X.); (C.P.C.)
| |
Collapse
|
49
|
Franchina M, Pizzimenti C, Fiorentino V, Martini M, Ricciardi GRR, Silvestris N, Ieni A, Tuccari G. Low and Ultra-Low HER2 in Human Breast Cancer: An Effort to Define New Neoplastic Subtypes. Int J Mol Sci 2023; 24:12795. [PMID: 37628975 PMCID: PMC10454084 DOI: 10.3390/ijms241612795] [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: 07/24/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
HER2-low and ultra-low breast cancer (BC) have been recently proposed as new subcategories of HER2 BC, supporting a re-consideration of immunohistochemical negative scores of 0, 1+ and the 2+/in situ hybridization (ISH) negative phenotype. In the present review, we outline the criteria needed to exactly distinguish HER2-low and ultra-low BC. Recent clinical trials have demonstrated significant clinical benefits of novel HER2 directing antibody-drug conjugates (ADCs) in treating these groups of tumors. In particular, trastuzumab-deruxtecan (T-Dxd), a HER2-directing ADC, has been recently approved by the US Food and Drug Administration as the first targeted therapy to treat HER2-low BC. Furthermore, ongoing trials, such as the DESTINY-Breast06 trial, are currently evaluating ADCs in patients with HER2-ultra low BC. Finally, we hope that new guidelines may help to codify HER2-low and ultra-low BC, increasing our knowledge of tumor biology and improving a targetable new therapeutical treatment.
Collapse
Affiliation(s)
- Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (M.F.); (V.F.); (M.M.); (N.S.); (A.I.)
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (M.F.); (V.F.); (M.M.); (N.S.); (A.I.)
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (M.F.); (V.F.); (M.M.); (N.S.); (A.I.)
| | | | - Nicola Silvestris
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (M.F.); (V.F.); (M.M.); (N.S.); (A.I.)
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (M.F.); (V.F.); (M.M.); (N.S.); (A.I.)
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, 98125 Messina, Italy; (M.F.); (V.F.); (M.M.); (N.S.); (A.I.)
| |
Collapse
|
50
|
Orsini A, Diquigiovanni C, Bonora E. Omics Technologies Improving Breast Cancer Research and Diagnostics. Int J Mol Sci 2023; 24:12690. [PMID: 37628869 PMCID: PMC10454385 DOI: 10.3390/ijms241612690] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC) has yielded approximately 2.26 million new cases and has caused nearly 685,000 deaths worldwide in the last two years, making it the most common diagnosed cancer type in the world. BC is an intricate ecosystem formed by both the tumor microenvironment and malignant cells, and its heterogeneity impacts the response to treatment. Biomedical research has entered the era of massive omics data thanks to the high-throughput sequencing revolution, quick progress and widespread adoption. These technologies-liquid biopsy, transcriptomics, epigenomics, proteomics, metabolomics, pharmaco-omics and artificial intelligence imaging-could help researchers and clinicians to better understand the formation and evolution of BC. This review focuses on the findings of recent multi-omics-based research that has been applied to BC research, with an introduction to every omics technique and their applications for the different BC phenotypes, biomarkers, target therapies, diagnosis, treatment and prognosis, to provide a comprehensive overview of the possibilities of BC research.
Collapse
Affiliation(s)
| | - Chiara Diquigiovanni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40131 Bologna, Italy; (A.O.); (E.B.)
| | | |
Collapse
|