1
|
Dutta K, Pal D, Li S, Shyam C, Shoghi KI. Corr-A-Net: Interpretable Attention-Based Correlated Feature Learning framework for predicting of HER2 Score in Breast Cancer from H&E Images. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.22.25326227. [PMID: 40313277 PMCID: PMC12045401 DOI: 10.1101/2025.04.22.25326227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is a critical biomarker for assessing breast cancer (BC) severity and guiding targeted anti-HER2 therapies. The standard method for measuring HER2 expression is manual assessment of IHC slides by pathologists, which is both time intensive and prone to inter- and intra-observer variability. To address these challenges, we developed an interpretable deep-learning pipeline with Correlational Attention Neural Network (Corr-A-Net) to predict HER2 score from H&E images. Each prediction was accompanied with a confidence score generated by the surrogate confidence score estimation network trained using incentivized mechanism. The shared correlated representations generated using the attention mechanism of Corr-A-Net achieved the best predictive accuracy of 0.93 and AUC-ROC of 0.98. Additionally, correlated representations demonstrated the highest mean effective confidence (MEC) score of 0.85 indicating robust confidence level estimation for prediction. The Corr-A-Net can have profound implications in facilitating prediction of HER2 status from H&E images.
Collapse
Affiliation(s)
- Kaushik Dutta
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Debojyoti Pal
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Suya Li
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Chandresh Shyam
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Kooresh I. Shoghi
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Lead contact
| |
Collapse
|
2
|
Medhus A, Schink KO, Longva AS, Engebraaten O, Berg K, Weyergang A. The mechanisms of HER2 targeted ADCs are dependent on Rab GTPases. Ther Adv Med Oncol 2025; 17:17588359251332473. [PMID: 40297622 PMCID: PMC12035104 DOI: 10.1177/17588359251332473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction In the era of personalized cancer therapy, antibody-drug conjugates (ADCs) have become one of the fastest-emerging groups of anticancer drugs. ADCs consist of an antibody coupled to a cytotoxic payload by a chemical linker, designed to be cleaved off intracellularly. Understanding the intracellular trafficking and processing of ADCs is crucial for elucidating their mechanism of action. Objective This study aimed to compare trastuzumab deruxtecan (T-DXd) to ado-trastuzumab emtansine (T-DM1) with emphasis on Rab GTPase-regulated intracellular trafficking and its impact on ADC efficacy. Methods The efficacy of T-DXd and T-DM1 was assessed in a panel of HER2-positive cell lines. Correlations between ADC efficacy and the expression of HER2 and Rab GTPases were evaluated. Functional studies, including knockdown (KD), overexpression, and microscopy, were performed to evaluate the impact of Rab GTPases on ADC cytotoxicity. Results In contrast to T-DM1, T-DXd efficacy was found not to correlate to HER2 expression in a panel of HER2-positive cell lines. However, a correlation to RAB5A expression was found for T-DXd efficacy, although not as strong as for T-DM1. Altering the expression of RAB5 in our model system confirmed RAB5 to have an impact on both T-DXd and T-DM1 cytotoxicity, but more on T-DM1. In addition, RAB4a was found to influence T-DXd sensitivity, but not T-DM1, indicating differences in intracellular processing between T-DXd and T-DM1. Conclusion The study demonstrates that ADC design significantly influences intracellular trafficking and processing. The linker design, in particular, plays a major role in determining the intracellular fate of an ADC.
Collapse
Affiliation(s)
- Astrid Medhus
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kay Oliver Schink
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute of Basic Medical Science, University of Oslo, Oslo, Norway
| | - Ane Sager Longva
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Olav Engebraaten
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kristian Berg
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Anette Weyergang
- Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Ullernchausseen 70, 0379 Oslo, Norway
| |
Collapse
|
3
|
Ruan Y, Liu X, Jin Y, Zhao M, Zhang X, Cheng X, Wang Y, Cao S, Yan M, Cai J, Li M, Gao B. Personalized predictions of neoadjuvant chemotherapy response in breast cancer using machine learning and full-field digital mammography radiomics. Front Med (Lausanne) 2025; 12:1582560. [PMID: 40313551 PMCID: PMC12043669 DOI: 10.3389/fmed.2025.1582560] [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: 02/24/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Objective This study aimed to develop a comprehensive nomogram model by integrating clinical pathological and full-field digital mammography (FFDM) radiomic features to predict the efficacy of neoadjuvant chemotherapy (NAC) in breast cancer patients, thereby providing personalized treatment recommendations. Methods A retrospective analysis was conducted on the clinical and imaging data of 227 breast cancer patients from 2016 to 2024 at the Second Affiliated Hospital of Harbin Medical University. The patients were divided into a training set (n = 159) and a test set (n = 68) with a 7:3 ratio. The region of interest (ROI) was manually segmented on FFDM images, and features were extracted and gradually selected. The rad-score was calculated for each patient. Five machine learning classifiers were used to build radiomics models, and the optimal model was selected. Univariate and multivariate regression analyses were performed to identify independent risk factors for predicting the efficacy of NAC in breast cancer patients. A nomogram prediction model was further developed by combining the independent risk factors and rad-score, and probability-based stratification was applied. An independent cohort was collected from an external hospital to evaluate the performance of the model. Results The radiomics model based on support vector machine (SVM) demonstrated the best predictive performance. FFDM tumor density and HER-2 status were identified as independent risk factors for achieving pathologic complete response (PCR) after NAC (p < 0.05). The nomogram prediction model, developed by combining the independent risk factors and rad-score, outperformed other models, with areas under the curve (AUC) of 0.91 and 0.85 for the training and test sets, respectively. Based on the optimal cutoff points of 103.42 from the nomogram model, patients were classified into high-probability and low-probability groups. When the nomogram model was applied to an independent cohort of 47 patients, only four patients had incorrect diagnoses. The nomogram model demonstrated stable and accurate predictive performance. Conclusion The nomogram prediction model, developed by integrating clinical pathological and radiomic features, demonstrated significant performance in predicting the efficacy of NAC in breast cancer, providing valuable reference for clinical personalized prediction planning.
Collapse
Affiliation(s)
- Ye Ruan
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xingyuan Liu
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yantong Jin
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingming Zhao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xingda Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaoying Cheng
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Wang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Siwei Cao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Menglu Yan
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianing Cai
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengru Li
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Gao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Freyer G, Rigault C, Guéroult Accolas L, Barea A, Radu N, Ouamer A, Saghatchian M. HERmione: Understanding the Needs of Patients Living with Metastatic HER2-Positive Breast Cancer Through a Cross-Sectional Survey in Parallel with Patients and Oncologists. Cancers (Basel) 2025; 17:1349. [PMID: 40282524 PMCID: PMC12025477 DOI: 10.3390/cancers17081349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES HER2-positive metastatic breast cancer continues to have a significant impact on patients' lives. The HERmione project was conducted in France to identify patients' needs for support and information, understand services offered, and identify differences in the perception of burden between patients and oncologists. METHODS Between July and October 2022, 273 patients with HER2-positive metastatic breast cancer and 40 oncologists were surveyed. The mean age of patients was 52 years, with most receiving treatment at specialized cancer centers (38%) or other public hospitals (34%). RESULTS The survey revealed a substantial burden of the disease and treatment in patients, perceptions that differed from those of oncologists. Both the physical and mental well-being of the patients were below average. Despite the burden of the disease, patients lacked access to many types of support, particularly support with sexual well-being. Additionally, 60% of patients did not have access to nursing support. Patients had high expectations regarding access to information but often did not know where to access this information. Despite this, they still exhibited treatment preferences. CONCLUSIONS These findings suggest that enhanced communication is critical to ensure that patients receive adequate support. Nursing support could improve patient-oncologist communication and thereby enhance patient well-being. Finally, to meet patient expectations regarding information access, a broader array of support tools should be offered.
Collapse
Affiliation(s)
- Gilles Freyer
- CHU Lyon-Sud, Institut de Cancérologie et Université de Lyon, 69002 Lyon, France
| | | | | | | | | | | | - Mahasti Saghatchian
- Hôpital Américain de Paris, Neuilly sur Seine, 92200 Neuilly-sur-Seine, France
| |
Collapse
|
5
|
Vincken R, Armendáriz-Martínez U, Ruiz-Sáenz A. ADCC: the rock band led by therapeutic antibodies, tumor and immune cells. Front Immunol 2025; 16:1548292. [PMID: 40308580 PMCID: PMC12040827 DOI: 10.3389/fimmu.2025.1548292] [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: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is a critical mechanism by which therapeutic antibodies leverage the immune system to target and eliminate cancer cells. The key agents of ADCC are natural killer (NK) cells, specifically targeting antibody-covered cancer cells through the CD16 receptor. While other immune cells and Fc receptors can contribute and enhance ADCC, NK cells and the CD16 receptor are crucial for the efficacy of cancer therapies such as trastuzumab, cetuximab and rituximab. Co-culture assays are essential for understanding the mechanisms of these therapies, overcoming resistance and optimizing novel therapeutic antibodies. This review highlights the importance of measuring ADCC to assess the efficacy of therapeutic antibodies. Here we also present the various in vitro models and assay methodologies available for studying ADCC, comparing the strengths and limitations of approaches like using PBMCs to better reflect real-life conditions or NK cell lines for standardization. It also covers different readouts for ADCC, either focusing on effector cells activation, including reporter and degranulation assays or in the target cell killing, including different molecule release assays, flow cytometry and immunofluorescence techniques. Selecting the best model for studying ADCC is crucial for the translational significance of therapeutic antibody research.
Collapse
Affiliation(s)
- Roos Vincken
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, CN, Rotterdam, Netherlands
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Uxue Armendáriz-Martínez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Ana Ruiz-Sáenz
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
6
|
Boonkaew S, Linfield S, Ferapontova EE. High-capacitance air-brushed electrodes for capacitive label-free bioassays. Talanta 2025; 293:128118. [PMID: 40220375 DOI: 10.1016/j.talanta.2025.128118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Non-invasive assays for protein biomarkers of cancer allow both its early diagnosis and continuous treatment monitoring. Yet, accurate point-of-care (POC) diagnostic devices for cancer diagnosis and monitoring, needed in point-of-care (POC) sites and places with limited resources, are scarce, not the least, due to their high current cost or bulky equipment necessary for analysis. Here, we show that the capacitive cellulase-linked electrochemical enzyme-linked aptamer-sorbent assay (e-ELASA) on magnetic beads (MBs) performed with airbrushed graphite (Gr) electrodes accurately and economically detects HER-2/neu, the protein biomarker of some aggressive forms of cancers and target of anticancer therapy. The disposable Gr electrodes were produced by airbrushing inexpensive graphite-powder/chitosan water inks onto polyester transparency films, producing high-capacitance electrodes, whose apparent specific capacitance ranged between 3.61 and 8.88 mF cm-2 as a function of the number of sprayed layers and graphite content in inks. The five-layer electrodes produced from 1.7 g of graphite powder (per 5 mL)/0.55 % chitosan water inks outperformed manually polished spectroscopic Gr electrodes earlier used in this label-free capacitive e-ELASA, as a result of the higher capacitive changes of the former, providing the same 0.1 fM limit of detection of HER-2/neu, in both buffer and 10 % serum, yet with a three-fold higher sensitivity. The portable and low cost airbrushed electrodes/e-ELASA set-up can be used for quick and accurate regular POC monitoring of HER-2/neu, particularly, in low and middle income settings, and, in perspective, the high-capacitance airbrushed electrodes can be adapted for other type label-free capacitive bioassays.
Collapse
Affiliation(s)
- Suchanat Boonkaew
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - Steven Linfield
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - Elena E Ferapontova
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark.
| |
Collapse
|
7
|
Robbins CJ, Bates KM, Rimm DL. HER2 testing: evolution and update for a companion diagnostic assay. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01016-y. [PMID: 40195456 DOI: 10.1038/s41571-025-01016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2025] [Indexed: 04/09/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2; encoded by ERBB2) testing has been a cornerstone of patient selection for HER2-targeted therapies, principally in breast cancer but also in several other solid tumours. Since the introduction of HercepTest as the original companion diagnostic for trastuzumab, HER2 assessment methods have evolved substantially, incorporating various testing modalities, from western blots, immunohistochemistry and fluorescence in situ hybridization, to early chromogenic quantitative methods and, probably in the future, fully quantitative methods. The advent of highly effective HER2-targeted antibody-drug conjugates with clinical activity at low levels of HER2 expression, such as trastuzumab deruxtecan, has necessitated the re-evaluation of HER2 testing, particularly for HER2-low tumours. In this Review, we provide an in-depth overview of the evolution of HER2 testing, the current clinical guidelines for HER2 testing across various solid tumours, challenges associated with current testing methodologies and the emerging potential of quantitative techniques. We discuss the importance of accurately defining HER2-low expression for therapeutic decision-making and how newer diagnostic approaches, such as quantitative immunofluorescence and RNA-based assays, might address the limitations of traditional immunohistochemistry-based methods. As the use of HER2-targeted therapies continues to expand to a wider range of tumour types, ensuring the precision and accuracy of HER2 testing will be crucial for guiding treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Charles J Robbins
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Katherine M Bates
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Mohamed G, Hamdy O, Alkallas A, Tahoun Y, Gomaa MM, Moaz I, Orabi A, Elzohery YH, Zakaria AS, Eltohamy MI. Role of artificial intelligence -based machine learning model in predicting HER2/neu gene status in breast cancer. Pathol Res Pract 2025; 270:155927. [PMID: 40233530 DOI: 10.1016/j.prp.2025.155927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 03/09/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025]
Abstract
Our study investigated the predictive efficacy of AI-based Machine Learning (ML) model for determining HER2 status in a population of 3424 breast cancer patients. Multivariate logistic regression analysis identified several independent variables that were predictive of HER2 positivity, namely age ≤ 40 years, tumor multicentricity, high tumor grade, high-grade DCIS, N3 stage disease, and negative ER status (p < 0.05). These findings suggest that patients presenting with these factors may benefit from more aggressive and targeted therapies. Furthermore, XGBoost ML model was trained using the dataset of 3324 patients, which was divided into an 80 % training set and a 20 % test set. The model achieved an impressive accuracy of 95 % on both training and test sets, as evidenced by the area under the curve (AUC) values of 0.95. The model ranked the presence of DCIS, DCIS component (major versus minor), DCIS grade, multiplicity of the tumor, and ER status as the top four variables for predicting HER2/neu status. To validate the performance of the proposed model, blind HER2 status data from an external validation cohort of 100 cases were utilized. Notably, the model demonstrated a sensitivity of 90.5 %, indicating its ability to accurately identify HER2-positive cases, and a specificity of 84.4 %, suggesting its capability to correctly classify HER2-negative cases. These results highlight the promising predictive efficacy of AI-based ML in determining HER2 status in breast cancer patients. The model's ability to accurately identify HER2-positive cases can assist in guiding treatment decisions, ensuring that patients receive appropriate and targeted therapies. However, further research with larger datasets is necessary to validate and generalize these findings.
Collapse
Affiliation(s)
- Ghada Mohamed
- Department of Pathology, National Cancer Institute, Cairo University, Egypt.
| | - Omar Hamdy
- Faculty of Engineering, Computer department, Cairo University, Egypt
| | - Anwar Alkallas
- Data analyst, Baheya Foundation for Early Detection And Management Of Breast Cancer, Egypt
| | | | - Mohammed Mohammed Gomaa
- Radiodiagnosis Department, National Cancer Institute, Cairo University, Egypt; Radiodiagnosis Department, Baheya Foundation for Early Detection and Management of Breast Cancer, Egypt
| | - Inas Moaz
- Epidemiology and preventive medicine department, National Liver Institute, Menoufia university, Egypt
| | - Ahmed Orabi
- Surgical oncology Department, National Cancer Institute, Cairo University, Egypt
| | | | - Al-Shimaa Zakaria
- Department of Pathology, National Cancer Institute, Cairo University, Egypt
| | | |
Collapse
|
9
|
Zhou S, Qin X, Xing W, Xu Z, Wei C, Ren Y, Gong Z. Differences in treatment response and survival between HER2(2+)/FISH-positive and HER2(3+) breast cancer patients after dual-target neoadjuvant therapy: a matched case-control study. Front Oncol 2025; 15:1530793. [PMID: 40255431 PMCID: PMC12006182 DOI: 10.3389/fonc.2025.1530793] [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: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 04/22/2025] Open
Abstract
Background The efficacy of neoadjuvant therapy (NAT) comprising dual-target drugs has been confirmed among patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC). Therefore, we explored the differences in responses to NAT and prognosis between patients with HER2(3+) and HER2(2+)/fluorescence in-situ hybridization (FISH)-positive BC after TCbHP-based dual-target NAT. Methods Data from patients with HER2-positive invasive BC who underwent NAT and radical surgery between January 2019 and December 2022 at the Peking University First Hospital and Cancer Hospital of Chinese Academy of Medical Sciences were retrospectively summarized. Propensity score matching (PSM) was used to reduce confounding effects. Pathological complete response (pCR) and invasive disease-free survival (IDFS) were evaluated to respectively reflect therapeutic response and patients' survival status. Results We selected 132 BC patients (66 pairs) through PSM form a cohort of 308 patients. The pCR rate of patients in the HER2(3+) group was significantly higher than that in the HER2(2+)/FISH-positive group after NAT (P<0.001). Univariate and multivariate logistic regression analyses determined that pCR was significantly affected by tumor grade, hormone receptor (HR) status, HER2 status (P<0.05). The 3-year IDFS rate of HER2(3+) BC patients was better than that of HER2(2+)/FISH-positive BC patient (P=0.083), although the difference was not statistically significant. Furthermore, multivariable Cox regression analysis exhibited that positive lymph node, HER2(3+), and pCR were independent prognostic factors for IDFS. Conclusion HER2(2+)/FISH-positive BC patients exhibited worse treatment response and prognosis than HER2(3+) BC patients after dual-target NAT, indicating that HER2 expression level is a crucial factor influencing the therapeutic efficacy and prognosis of BC patients after TCbHP-based dual-target NAT.
Collapse
Affiliation(s)
- Sicheng Zhou
- Department of Thyroid and Breast Surgery, Peking University First Hospital, Beijing, China
| | - Xuhui Qin
- Department of General Surgery, Zanhuang County Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Wei Xing
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhao Xu
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chunlv Wei
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yining Ren
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zixing Gong
- Department of General Surgery, Hebei Provincial Hospital of Chinese Medicine/The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
10
|
Utkan Korun ZE, Kırıcı P, Elibol E, Kaplan S, Karacor T. The role of toll-like receptor 4 in the development of endometriosis and the benefits of trastuzumab in the treatment of endometriosis: a rat model. Biotech Histochem 2025; 100:129-136. [PMID: 40260736 DOI: 10.1080/10520295.2025.2486451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
We aimed to investigate TLR-4 receptor activity in the development of endometriosis and the effect of trastuzumab in experimentally induced endometriotic tissue via TLR-4 in this study. Twenty-eight female Wistar-Albino rats were divided into four groups: Group 1 (Control Group), Group 2 (Endometriosis Group), Group 3 (Endometriosis + Trastuzumab Group), and Group 4 (Trastuzumab Group). All animal tissue samples were collected. Histopathological, immunohistochemical, and biochemical analyses were performed. In histopathological analysis, there was a significant difference between Group 2 and other groups in terms of connective tissue edema, inflammation, hemorrhage, epithelial damage, and mast cell density. In immunohistochemical analysis with TLR-4, Group 2 exhibited strong staining. In biochemical analysis, it was found that there was a highly significant difference between Group 2 and Group 1 considering the Malondialdehyde (MDA) levels in plasma samples. There was no significant difference in terms of the MDA levels among other groups. Considering the glutathione levels in the plasma samples, it was found that there was a highly significant difference between Group 2 and Groups 3 and 4. Trastuzumab may play a role in the treatment of histopathological damage and fibrosis in experimentally induced endometriotic implants by showing anti-inflammatory and antiproliferative activity.
Collapse
Affiliation(s)
- Zeynep Ece Utkan Korun
- Department of Obstetrics and Gynecology, Yeditepe University Kozyatagi Hospital, Istanbul, Turkey
| | - Pınar Kırıcı
- School of Medicine, Department of Obstetrics and Gynecology, Malatya Turgut Ozal University, Malatya, Turkey
| | - Ebru Elibol
- School of Medicine, Department of Histology and Embryology, Adiyaman University, Adiyaman, Turkey
| | - Selcuk Kaplan
- School of Medicine, Department of Obstetrics and Gynecology, Adiyaman University, Adiyaman, Turkey
| | - Talip Karacor
- Obstetrics and Gynecology Clinic, Sakarya Training and Research Hospital, Sakarya, Turkey
| |
Collapse
|
11
|
Krupa K, Liszcz-Tymoszuk A, Czerw N, Czerw A, Sygit K, Kozłowski R, Deptała A, Badowska-Kozakiewicz A. CDK4/6 as a Therapeutic Target in HR+/HER2- Breast Cancer Cells-Current Treatment Status. Cancers (Basel) 2025; 17:1039. [PMID: 40149372 PMCID: PMC11940879 DOI: 10.3390/cancers17061039] [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: 02/05/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Breast cancer is the most frequently diagnosed neoplasm in the world. It can be classified into four main subtypes, each of them showing differences in the expression of hormone receptor (HR), human epidermal growth factor receptor 2 (HER2), and in cell metabolism. Since 2015, when The U.S. Food and Drug Administration (FDA) approved the first cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor that regulates the cell cycle, treatment of HR+/HER2- BC has become much more effective. Currently, palbociclib, ribociclib, and abemaciclib are more often used both in combination with endocrine therapy as well as in monotherapy. Their application has been extensively verified in many clinical trials such as PALOMA-1,2,3, MONALEESA-1,2,3,7, and MONARCH-1,2,3, which allowed the verification of differences in their effectiveness, dosage, and adverse effects. Subsequent studies, MonarchE and NATALEE, examined the role of these inhibitors as adjuvant therapy, as well as at verifying their safety. Moreover, dalpiciclib is being investigated in HR+/HER2- BC treatment. This article will summarize clinical efficacy, recommendations, and differences in toxicity profile between palbociclib, ribociclib, and abemaciclib and will also discuss the possibility of using dalpiciclib in the treatment of breast cancer.
Collapse
Affiliation(s)
- Kamila Krupa
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.L.-T.); (N.C.)
| | - Anna Liszcz-Tymoszuk
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.L.-T.); (N.C.)
| | - Natalia Czerw
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.L.-T.); (N.C.)
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 01-445 Warsaw, Poland;
- Department of Economic and System Analyses, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland;
| | - Remigiusz Kozłowski
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland;
| | - Andrzej Deptała
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.D.); (A.B.-K.)
| | - Anna Badowska-Kozakiewicz
- Department of Oncology Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (A.D.); (A.B.-K.)
| |
Collapse
|
12
|
Toms L, FitzPatrick L, Auckland P. Super-resolution microscopy as a drug discovery tool. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100209. [PMID: 39824440 DOI: 10.1016/j.slasd.2025.100209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
At the turn of the century a fundamental resolution barrier in fluorescence microscopy known as the diffraction limit was broken, giving rise to the field of super-resolution microscopy. Subsequent nanoscopic investigation with visible light revolutionised our understanding of how previously unknown molecular features give rise to the emergent behaviour of cells. It transpires that the devil is in these fine molecular details, and essential nanoscale processes were found everywhere researchers chose to look. Now, after nearly two decades, super-resolution microscopy has begun to address previously unmet challenges in the study of human disease and is poised to become a pivotal tool in drug discovery.
Collapse
Affiliation(s)
- Lauren Toms
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Macclesfield, Cheshire SK10 4ZF, United Kingdom.
| | - Lorna FitzPatrick
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Macclesfield, Cheshire SK10 4ZF, United Kingdom
| | - Philip Auckland
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Macclesfield, Cheshire SK10 4ZF, United Kingdom.
| |
Collapse
|
13
|
Pruss M, Cieslik JP, Török J, Dobrowolski J, Neubacher M, Helbig M, Friebe V, Häberle L, Krawczyk N, Borgmeier F, Fehm T, Dietzel F, Mohrmann S. Hormone and HER2-receptor status in breast cancer: determination using sonographically guided core needle biopsy and correlation with excision specimen-a German single institution diagnostic accuracy study. Arch Gynecol Obstet 2025; 311:881-891. [PMID: 39912929 PMCID: PMC11919962 DOI: 10.1007/s00404-024-07920-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND Sonographically guided core needle biopsy (CNB) is a well-established tool for diagnosing breast lesions. Preoperative estrogen receptor (ER), progesterone receptor (PR), and HER2-receptor status are essential for a personalized treatment approach. OBJECTIVES We evaluated the concordance of the hormone- and HER2-receptor status between the CNB and the surgical specimen to determine the accuracy of the CNB as a diagnostic method. DESIGN This is a non-interventional retrospective study analyzing breast cancer patients treated at the breast care center of the University Medical Center Duesseldorf between January 2002 and December 2005. METHODS Patients with paired CNB and surgical specimens and a diagnosis of invasive breast cancer were included. ER, PR, and HER2 status were determined by immunohistochemistry (IHC). Patients with IHC 2+ results were further examined by fluorescence in situ hybridization (FISH). Concordance of receptor status was calculated using specificity, sensitivity, and negative and positive predictive values. RESULTS We found a very good agreement between CNB and surgical specimens regarding receptor status. A total of 248 patients were analyzed. Concordance rates in cases of primary surgery for ER, PR, and HER2 were 92.9%, 92.9%, and 93%, respectively. In cases of neoadjuvant chemotherapy, the concordance rates for ER, PR, and HER2 were 100%, 87.5%, and 96%, respectively. CONCLUSION CNB demonstrated high diagnostic accuracy compared with surgical specimens regarding ER, PR, and HER2-receptor status. Our findings support the recommendation to use sonographically guided CNB as the initial diagnostic method for guiding tailored treatment plans.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/surgery
- Breast Neoplasms/diagnosis
- Breast Neoplasms/diagnostic imaging
- Female
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/analysis
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/analysis
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/analysis
- Middle Aged
- Retrospective Studies
- Adult
- Biopsy, Large-Core Needle
- Aged
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Aged, 80 and over
- Sensitivity and Specificity
- Germany
- Image-Guided Biopsy
- Ultrasonography, Interventional
Collapse
Affiliation(s)
- Maximilian Pruss
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| | - Jan-Philipp Cieslik
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Janet Török
- Med 360°, Breast Imaging Center of Radiology, Luegallee 52, 40545, Düsseldorf, Germany
| | - Jerome Dobrowolski
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Melissa Neubacher
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Martina Helbig
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Verena Friebe
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Lena Häberle
- Institute of Pathology, Medical Faculty, University Hospital Duesseldorf, Heinrich Heine University, 40204, Düsseldorf, Germany
| | - Natalia Krawczyk
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Felix Borgmeier
- MVZ Amedes for Prenatal-Medicine und Genetic GmbH, 40210, Düsseldorf, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Frederic Dietzel
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Duesseldorf, 40225, Duesseldorf, Germany
| | - Svjetlana Mohrmann
- Department of Obstetrics and Gynecology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| |
Collapse
|
14
|
Zhou L, Zhang Y, Zhang J, Wang H, Zhao B, Cai Y, Qu Y, Li X, Zhang D. Clinical characteristics and therapeutic direction of HER2 low-expression breast cancer. Front Oncol 2025; 15:1484103. [PMID: 40083869 PMCID: PMC11903420 DOI: 10.3389/fonc.2025.1484103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is one of the oncogenic drivers of breast cancer and is often used as a definitive therapeutic marker for breast cancer. This has led to significant improvements in both targeted therapy and prognosis for HER2-targeted breast cancer. Due to the differences in HER2 gene and protein expression levels, they are clinically classified into HER2 zero-expression breast cancer, low-expression breast cancer and high-expression breast cancer. Among them, HER2 low-expression is considered a special expression state, which is insensitive to conventional anti-HER2 therapy and has a poorer prognosis and thus has received attention from researchers. Some studies demonstrate that patients with HER2 low-expression can benefit from antibody-drug conjugates (ADC). Several studies are currently exploring the efficacy of various ADC drugs in breast cancer with HER2 low-expression, opening up new treatment avenues for patients with HER2 low-expression breast cancer. This review aims to summarize the clinical features of HER2 low-expression breast cancer and the recent advances in its therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dongwei Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
15
|
Du R, Cao C, Fan D, Li G, Pu S, Xu X, Liu M, Shi G, Wu Y, Hao Q, Gao Y, Zhang J, Zhao H, Zhang C. NK cell immunopotentiators-loaded nanoliposomes enhance ADCC effect for targeted therapy against HER2-positive breast cancer. Cell Commun Signal 2025; 23:106. [PMID: 39987140 PMCID: PMC11846243 DOI: 10.1186/s12964-024-02023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/30/2024] [Indexed: 02/24/2025] Open
Abstract
Trastuzumab serves as a cornerstone of first-line therapy for HER2-positive (HER2+) breast cancer; however, a significant challenge arises due to the emergence of resistance within approximately one year of commencement of treatment, particularly in advanced cases with metastatic disease where its efficacy is limited. Our investigation into the tumor tissue from HER2+ breast cancer patients, employing single-cell sequencing and bioinformatics analysis, has elucidated a crucial mechanism underlying the reduced responsiveness of tumors to trastuzumab: the diminished infiltration and activity of natural killer (NK) cells within the tumor microenvironment (TME). To counteract this impediment, we meticulously selected two potent immune-modulating peptides TKD and IP-10p, which are known to recruit and enhance the activity of NK cells. Through in vitro experiments, we substantiated that bolstering the tumor infiltration and activity of NK cells can lead to an enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) effect, thereby amplifying the anti-tumor activity of trastuzumab. Building upon this foundational discovery, we further designed HER2-targeted pH-sensitive nanoliposomes to encapsulate TKD and IP-10p peptides. The novel designed nanoliposomes were strategically employed in conjunction with NK cell supplement therapy within a HER2+ breast cancer model undergoing trastuzumab treatment, yielding a striking anti-tumor response and indicating that the combination strategy effectively reinvigorated the anti-tumor immune response. In essence, this study not only underscores a critical link between the diminished ADCC effect mediated by trastuzumab and the development of resistance in HER2+ breast cancer but also demonstrates leveraging HER2-targeted nanoliposomes to deliver NK cell immunopotentiators can significantly enhance the functional activity of NK cells and their infiltration within the TME, culminating in improved antitumor efficacy of trastuzumab through the augmentation of the ADCC effect.
Collapse
Affiliation(s)
- Ruoxin Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, P. R. China
| | - Changqing Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, P. R. China
- Department of General Surgery, The Second Affiliated Hospital of The Fourth Military Medical University, 710038, Xi'an, P. R. China
| | - Dong Fan
- Department of General Surgery, The Second Affiliated Hospital of The Fourth Military Medical University, 710038, Xi'an, P. R. China
| | - Guodong Li
- College of Life Science, Northwest University, Xi'an, 710069, P. R. China
| | - Shuangpeng Pu
- College of Life Science, Northwest University, Xi'an, 710069, P. R. China
| | - Xinyao Xu
- College of Life Science, Northwest University, Xi'an, 710069, P. R. China
| | - Mengmeng Liu
- College of Life Science, Northwest University, Xi'an, 710069, P. R. China
| | - Gege Shi
- College of Life Science, Northwest University, Xi'an, 710069, P. R. China
| | - Yuxin Wu
- College of Life Science, Northwest University, Xi'an, 710069, P. R. China
| | - Qiang Hao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, P. R. China
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, P. R. China
| | - Juliang Zhang
- Department of Vascular and Endocrine Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710000, China.
| | - Huadong Zhao
- Department of General Surgery, The Second Affiliated Hospital of The Fourth Military Medical University, 710038, Xi'an, P. R. China.
| | - Cun Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, P. R. China.
| |
Collapse
|
16
|
Khanmammadov N, Doğan I, Khishigsuren B, Azizy A, Saip P, Aydiner A. Rechallenge of trastuzumab-based therapy in HER2-positive breast cancer patients who progressed after lapatinib plus capecitabine. Medicine (Baltimore) 2025; 104:e41468. [PMID: 39928785 PMCID: PMC11813031 DOI: 10.1097/md.0000000000041468] [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: 07/01/2024] [Revised: 01/11/2025] [Accepted: 01/17/2025] [Indexed: 02/12/2025] Open
Abstract
Data regarding the use of rechallenge trastuzumab (RTmab)-based therapies in the management of heavily pretreated patients with HER2-positive breast cancer (BC) in the literature are limited. This study aimed to evaluate the efficacy of trastuzumab-based therapy in patients who experienced disease progression after receiving lapatinib plus capecitabine (LC). In this retrospective study, the data of thirty three HER2 positive metastatic BC patients who progressed after LC treatment and subsequently received trastuzumab-based treatment were evaluated. Trastuzumab was administered at an initial loading dose of 8 mg/kg followed by a maintenance dose of 6 mg/kg every 21 days. The average age of patients is 47 years (range 25-72 years). The predominant histopathological subtype was invasive ductal carcinoma, which was observed in 23 (70%) patients. Estrogen receptor (ER) positivity was also noted in 16 (48%) patients. All patients had received palliative trastuzumab plus chemotherapy (Cht) before the lapatinib. In conjunction with trastuzumab-based therapy, vinorelbine was administered to 14 (42%) patients, paclitaxel to 12 (36%), and other chemotherapeutic agents to 4 (12%). For all patients, the objective response and disease control rates were 27% and 69%, respectively. Furthermore, the median progression-free survival (PFS) was 8.8 months (95% confidence interval [CI]: 6.6-11), and the median overall survival was 20 months (95% CI: 15.1-25.8). There were no statistically significant differences in PFS rates based on several factors, including age, ER status, denovo metastasis, brain metastasis, perioperative Cht, pre-Rtmab hormone therapy, and which Cht was used along with Rtmab (P > .05). Mild to moderate adverse events were observed in 17 (52%) patients, whereas only 4 (12%) patients had Grade 3 to 4 toxicity. This study demonstrated that RTmab-based therapy is effective in patients who progressed after LC. These findings contribute to the literature by suggesting that RTmab is a viable treatment option for patients with HER2-positive metastatic BC.
Collapse
Affiliation(s)
- Nijat Khanmammadov
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Izzet Doğan
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Bayarmaa Khishigsuren
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Abdulmunir Azizy
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Pinar Saip
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Adnan Aydiner
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| |
Collapse
|
17
|
Boonkaew S, Teodori L, Vendelbo MH, Kjems J, Ferapontova EE. Nanobodies' duo facilitates ultrasensitive serum HER-2/neu immunoassays via enhanced avidity interactions. Anal Chim Acta 2025; 1335:343472. [PMID: 39643321 DOI: 10.1016/j.aca.2024.343472] [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: 09/19/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Existing liquid biopsy assays for protein biomarkers of cancer are mostly based on antibodies (Ab) contributing unfavorably to their high cost. Easy to express and modify in vitro, nanobodies may be a cost-effective alternative to Ab. RESULTS We show that serum HER-2/neu, a biomarker and target of aggressive HER-2/neu(+) cancers, can be accurately detected in a 1.2 h electrochemical cellulase-linked sandwich nanobody/aptamer assay on magnetic beads. Using a single nanobody receptor, 2Rs15d or 2Rb17c, reduces immunoassay's sensitivity by 35%-26 %. A combination of two nanobodies as a duo-receptor recovers the sensitivity of the enzyme-linked nanobody/aptamer-sorbent assay (ELNASA) to 11.9 ± 2.8 μC fM-1, due to the avidity effects making the nanobodies-duo binding properties comparable to those of Ab. Down to 0.1 fM HER-2/neu was detected by ELNASA in serum samples, with no interference from other blood-circulating proteins. In a 30 healthy-volunteers trial, ELNASA more accurately than optical ELISA assayed serum HER-2/neu. SIGNIFICANCE ELNASA performance rivals that of ELISA, yet estimated to be at least 200 times cheaper, due to the lower cost of nanobodies production, and may be better suited for routine clinical analysis of HER-2/neu, particularly, in low- and middle-income settings with limited resources. The ELNASA approach is generic and may be adapted for specific and ultrasensitive analysis of other blood-circulating proteins.
Collapse
Affiliation(s)
- Suchanat Boonkaew
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Laura Teodori
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Mikkel H Vendelbo
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus C, Denmark
| | - Elena E Ferapontova
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark.
| |
Collapse
|
18
|
Qin G, Shao X, Liu X, Xu J, Wang X, Wang W, Gao L, Liang Y, Xie L, Su D, Yang H, Zhou W, Fang X. A signaling molecule from intratumor bacteria promotes trastuzumab resistance in breast cancer cells. Proc Natl Acad Sci U S A 2025; 122:e2421710122. [PMID: 39786928 PMCID: PMC11745319 DOI: 10.1073/pnas.2421710122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
Emerging evidence indicates that intratumor bacteria exist as an active and specific tumor component in many tumor types beyond digestive and respiratory tumors. However, the biological impact and responsible molecules of such local bacteria-tumor direct interaction on cancer therapeutic response remain poorly understood. Trastuzumab is among the most commonly used drugs targeting the receptor tyrosine-protein kinase erbB-2 (ErbB2) in breast cancer, but its resistance is inevitable, severely limiting its clinical effectiveness. Here, we demonstrate that the quorum-sensing signaling molecule N-(3-oxo-dodecanoyl) homoserine lactone (3oc), a chemical compound released by Pseudomonas aeruginosa (P. aeruginosa), one tumor-resident bacteria with a relative high abundance in breast cancer, promotes breast cancer cell resistance to trastuzumab. Mechanically, 3oc directly leads to spontaneous dimerization of the transforming growth factor β (TGF-β) type II serine/threonine kinase receptor on the cell membrane in a ligand-independent manner. The 3oc-induced TGF-β signaling subsequently triggers ErbB2 phosphorylation and its downstream target activation, overcoming the inhibition effect of trastuzumab on ErbB2. With specific real-time qPCR, fluorescence in situ hybridization imaging, and liquid chromatography ionization tandem mass spectrometry analyses of clinical samples, we confirmed that P. aeruginosa and its signaling molecule 3oc exist in breast cancer tissues and there is a clinical correlation between P. aeruginosa colonization and trastuzumab resistance. This work expands the biological functions of intratumor bacteria in cancer treatment responsiveness and provides a unique perspective for overcoming trastuzumab resistance.
Collapse
Affiliation(s)
- Gege Qin
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua-Peking Center for Life Sciences, Institute of Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing100084, China
| | - Xiying Shao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Xiaolong Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Jiachao Xu
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Wenxi Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
| | - Lu Gao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Yuxin Liang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Lina Xie
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Dan Su
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou310022, Zhejiang, China
| | - Hongwei Yang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
| | - Wei Zhou
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing100190, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, Zhejiang, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
19
|
Liou L, García-González J, Wu HM, Wang Z, Hoggart CJ, Kontorovich AR, Kovacic JC, O'Reilly PF. Clinical and Genomic Prediction of Coronary Artery Disease Subtypes. Arterioscler Thromb Vasc Biol 2025; 45:90-103. [PMID: 39633571 DOI: 10.1161/atvbaha.124.321846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Coronary artery disease (CAD) is a complex, heterogeneous disease with distinct etiological mechanisms. These different etiologies may give rise to multiple subtypes of CAD that could benefit from alternative preventions and treatments. However, so far, there have been no systematic efforts to predict CAD subtypes using clinical and genetic factors. METHODS Here, we trained and applied statistical models incorporating clinical and genetic factors to predict CAD subtypes in 26 036 patients with CAD in the UK Biobank. We performed external validation of the UK Biobank models in the US-based All of Us cohort (8598 patients with CAD). Subtypes were defined as high versus normal LDL (low-density lipoprotein) levels, high versus normal Lpa (lipoprotein A) levels, ST-segment-elevation myocardial infarction versus non-ST-segment-elevation myocardial infarction, occlusive versus nonocclusive CAD, and stable versus unstable CAD. Clinical predictors included levels of ApoA, ApoB, HDL (high-density lipoprotein), triglycerides, and CRP (C-reactive protein). Genetic predictors were genome-wide and pathway-based polygenic risk scores (PRSs). RESULTS Results showed that both clinical-only and genetic-only models can predict CAD subtypes, while combining clinical and genetic factors leads to greater predictive accuracy. Pathway-based PRSs had higher discriminatory power than genome-wide PRSs for the Lpa and LDL subtypes and provided insights into their etiologies. The 10-pathway PRS most predictive of the LDL subtype involved cholesterol metabolism. Pathway PRS models had poor generalizability to the All of Us cohort. CONCLUSIONS In summary, we present the first systematic demonstration that CAD subtypes can be distinguished by clinical and genomic risk factors, which could have important implications for stratified cardiovascular medicine.
Collapse
Affiliation(s)
- Lathan Liou
- Department of Genetics and Genomic Sciences (L.L., J.G.-G., H.M.W., C.J.H., P.F.O.), Icahn School of Medicine, New York, NY
| | - Judit García-González
- Charles Bronfman Institute for Personalized Medicine (Z.W.), Icahn School of Medicine, New York, NY
| | - Hei Man Wu
- Department of Genetics and Genomic Sciences (L.L., J.G.-G., H.M.W., C.J.H., P.F.O.), Icahn School of Medicine, New York, NY
| | - Zhe Wang
- Charles Bronfman Institute for Personalized Medicine (Z.W.), Icahn School of Medicine, New York, NY
| | - Clive J Hoggart
- Department of Genetics and Genomic Sciences (L.L., J.G.-G., H.M.W., C.J.H., P.F.O.), Icahn School of Medicine, New York, NY
| | - Amy R Kontorovich
- Zena and Michael A. Wiener Cardiovascular Institute (A.R.K., J.C.K.), Icahn School of Medicine, New York, NY
- Cardiovascular Research Institute (A.R.K.), Icahn School of Medicine, New York, NY
- Biomedical Engineering and Imaging Institute (A.R.K.), Icahn School of Medicine, New York, NY
- The Institute for Genomic Health (A.R.K.), Icahn School of Medicine, New York, NY
| | - Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute (A.R.K., J.C.K.), Icahn School of Medicine, New York, NY
- Department of Cardiology, St Vincent's Hospital, Sydney, NSW, Australia (J.C.K.)
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia (J.C.K.)
- Victor Chang Cardiac Research Institute, Sydney, Australia (J.C.K.)
| | - Paul F O'Reilly
- Department of Genetics and Genomic Sciences (L.L., J.G.-G., H.M.W., C.J.H., P.F.O.), Icahn School of Medicine, New York, NY
| |
Collapse
|
20
|
Malecka-Baturo K, Grabowska I. Efficiency of electrochemical immuno- vs. apta(geno)sensors for multiple cancer biomarkers detection. Talanta 2025; 281:126870. [PMID: 39298804 DOI: 10.1016/j.talanta.2024.126870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
The interest in biosensors technology has been constantly growing over the last few years. It is still the biggest challenge to design biosensors able to detect two or more analytes in a single measurement. Electrochemical methods are frequently used for this purpose, mainly due to the possibility of applying two or more different redox labels characterized by independent and distinguished electrochemical signals. In addition to antibodies, nucleic acids (aptamers) have been increasingly used as bioreceptors in the construction of such sensors. Within this review paper, we have collected the examples of electrochemical immuno- and geno(apta)sensors for simultaneous detection of multiple analytes. Based on many published literature examples, we have emphasized the recent application of multiplexed platforms for detection of cancer biomarkers. It has allowed us to compare the progress in design strategies, including novel nanomaterials and amplification of signals, to get as low as possible limits of detection. We have focused on multi-electrode and multi-label strategies based on redox-active labels, such as ferrocene, anthraquinone, methylene blue, thionine, hemin and quantum dots, or metal ions such as Ag+, Pb2+, Cd2+, Zn2+, Cu2+ and others. We have finally discussed the possible way of development, challenges and prospects in the area of multianalyte electrochemical immuno- and geno(apta)sensors.
Collapse
Affiliation(s)
- Kamila Malecka-Baturo
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Str. 10, 10-748, Olsztyn, Poland
| | - Iwona Grabowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Str. 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
21
|
Yoon SM, Wu S, Schwer A, Glaser S, DeWees T, Bazan JG. Are All Prognostic Stage IB Breast Cancers Equivalent? Cancers (Basel) 2024; 16:3830. [PMID: 39594785 PMCID: PMC11592610 DOI: 10.3390/cancers16223830] [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: 10/17/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: The 8th edition of the American Joint Committee on Cancer integrates histology and biomarker status with anatomic extent in breast cancer (BC) pathologic prognostic staging (PPS). However, PPS IB includes anatomic locally advanced hormone-receptor-positive/HER2-negative (LA-HR+/HER2-) and early-stage triple-negative BC (ES-TNBC). Previous research shows that increased nodal involvement is a critical predictor of worse prognosis, raising questions about whether biological subtype or anatomic stage has a greater influence on outcomes in these discordant cases. We hypothesized that overall survival (OS) remains worse for LA-HR+/HER2- BC compared to ES-TNBC, despite both being classified as PPS IB. Methods: Using the National Cancer Database, we identified patients with LA-HR+/HER2- BC (pT3N1 or pT1-3N2, grade 1-2) and ES-TNBC (T1N0, grade 2-3) treated between 2004 and 2017. Patients without complete primary tumor stage, biomarker status, grade, TNM staging, or treated with neoadjuvant therapy were excluded. The primary endpoint was OS. Multivariable Cox regression evaluated OS between LA-HR+/HER2- BC and ES-TNBC. Results: Among 45,818 patients (17,359 LA-HR+/HER2- BC and 28,459 ES-TNBC), LA-HR+/HER2- BC had significantly worse 6-year OS (86.1% vs. 90.4%; HR = 1.63; p < 0.0001). Among patients receiving appropriate therapies, patients with LA-HR+/HER2- BC had 35% relatively higher risk of death (HR = 1.35; 1.24-1.48; p < 0.0001). These results highlight that LA-HR+/HER2- breast cancer has worse survival compared to ES-TNBC, despite both being classified as PPS IB and receiving all appropriate treatments. Conclusions: Anatomic disease extent remains an important factor in patients with discordant AS and PPS. Future iterations of PPS should re-classify LA-HR+/HER2- breast cancer from PPS IB to ensure more accurate prognostic and survival information.
Collapse
Affiliation(s)
- Stephanie M. Yoon
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Shengyang Wu
- City of Hope Arcadia Radiation Oncology, Arcadia, CA 91007, USA
| | - Amanda Schwer
- Department of Radiation Oncology, City of Hope Orange County Lennar Foundation Cancer Center, Irvine, CA 92618, USA
| | - Scott Glaser
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Todd DeWees
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jose G. Bazan
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
22
|
Lepage V, Clarenne J, Fulbert B, Slimano F, Jouannaud C, Parent D. A 14-year retrospective of HER2 + metastatic breast cancer treatment at one comprehensive cancer center: Impact of the trastuzumab/pertuzumab and trastuzumab-emtansine sequence versus trastuzumab on overall survival. J Oncol Pharm Pract 2024:10781552241289923. [PMID: 39397571 DOI: 10.1177/10781552241289923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Introduction: Monoclonal antibodies represent a significant improvement in the treatment of the HER2 + metastatic cancer, which is associated with a worse prognosis. The objective of this study was to compare overall survival (OS) data in patients treated with trastuzumab + pertuzumab followed by trastuzumab-emtansine (T-DM1) in the second line of metastatic treatment (Arm A) versus patients treated with trastuzumab alone (Arm B). Progression-free survival (PFS) in first-line metastatic patients was also compared in both arms. Methods: This single-center retrospective study included patients from February 2008 to August 2022. OS and PFS of both arms were described and estimated using the Kaplan-Meier method. Data were extracted from electronic medical records and CHIMIO prescribing software. Results: The total duration of metastatic treatment of the 82 patients was significantly longer in the arm A (43.2 ± 28 months vs 33.6 ± 28.9 months), as was the median time to death (59 vs 52 months). The OS data showed a significant reduction in the risk of death in the arm A (Hazard Ratio = 0.59; 95% Confidence Interval [0.37-0.94]; p = 0.02). No difference was shown for PFS. Conclusion: The trastuzumab/pertuzumab/T-DM1 combination showed a significant improvement in OS. Our results are in line with the CLEOPATRA and EMILIA studies, but long-term responders in the arm A may have impacted our results. The absence of difference in term of PFS in first metastatic line may be explained by a selection bias, as patients in the arm A potentially have more aggressive forms.
Collapse
Affiliation(s)
- V Lepage
- CHU Reims, Department of Pharmacy, Reims, France
| | - J Clarenne
- CHU Reims, Department of Pharmacy, Reims, France
| | - B Fulbert
- CHU Reims, Department of Pharmacy, Reims, France
| | - F Slimano
- CHU Reims, Department of Pharmacy, Reims, France
- Université de Reims Champagne-Ardenne, UR7506 BioSpect, CHU Reims, Department of Pharmacy, Reims, France
| | - C Jouannaud
- Pharmacy Department, Godinot Institute, Reims, France
| | - D Parent
- Pharmacy Department, Godinot Institute, Reims, France
| |
Collapse
|
23
|
Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH, Lee KB. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300744. [PMID: 37058079 PMCID: PMC10576016 DOI: 10.1002/smll.202300744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Nanotechnology has emerged as a promising approach for the targeted delivery of therapeutic agents while improving their efficacy and safety. As a result, nanomaterial development for the selective targeting of cancers, with the possibility of treating off-target, detrimental sequelae caused by chemotherapy, is an important area of research. Breast and ovarian cancer are among the most common cancer types in women, and chemotherapy is an essential treatment modality for these diseases. However, chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy are common side effects that can affect breast and ovarian cancer survivors quality of life. Therefore, there is an urgent need to develop effective prevention and treatment strategies for these adverse effects. Nanoparticles (NPs) have extreme potential for enhancing therapeutic efficacy but require continued research to elucidate beneficial interventions for women cancer survivors. In short, nanotechnology-based approaches have emerged as promising strategies for preventing and treating chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy. NP-based drug delivery systems and therapeutics have shown potential for reducing the side effects of chemotherapeutics while improving drug efficacy. In this article, the latest nanotechnology approaches and their potential for the prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors are discussed.
Collapse
Affiliation(s)
- Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Callan D. McLoughlin
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Joshua B. Stein
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mohammad Abdur Rashid
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| |
Collapse
|
24
|
Liu HN, Zhu Y, Chi Y, Zhang Y, Li X, Wen W, Shan LS, Wang YT, Dai B. Synthetic routes and clinical application of Small-Molecule HER2 inhibitors for cancer therapy. Bioorg Chem 2024; 151:107653. [PMID: 39024803 DOI: 10.1016/j.bioorg.2024.107653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
This comprehensive review undertakes a meticulous scrutiny of the synthesis and clinical applications pertaining to small-molecule tyrosine kinase inhibitors (TKIs) directed towards the human epidermal growth factor receptor 2 (HER2), a pivotal protagonist in the pathogenesis of cancer. Focused on compounds like lapatinib, neratinib, and tucatinib, the review delves into the intricate synthesis strategies, emphasizing the challenges associated with their structural complexity. The clinical utilization of HER2 TKIs underscores noteworthy strides in the therapeutic landscape for HER2-positive breast and gastric malignancies. Lapatinib, a dual HER2/ epidermal growth factor receptor (EGFR) inhibitor, has demonstrated efficacy in combination therapies, addressing the need for overcoming resistance mechanisms. Neratinib, an irreversible HER2 inhibitor, presents a promising avenue for patients with refractory tumors. Tucatinib, strategically engineered to traverse the blood-brain barrier, epitomizes a groundbreaking advancement in the management of metastatic HER2-positive breast cancer manifesting cerebral involvement. Despite their success, challenges such as resistance mechanisms and off-target effects persist, urging continuous research for the development of next-generation HER2 TKIs. This comprehensive review serves as a valuable resource for pharmaceutical scientists, offering insights into the synthetic intricacies and clinical impact of small-molecule TKIs targeting HER2.
Collapse
Affiliation(s)
- He-Nan Liu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yao Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xun Li
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wen Wen
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Li-Shen Shan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Ya-Tao Wang
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000 Leuven, Belgium.
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
25
|
Cho D, Lord SJ, Ward R, IJzerman M, Mitchell A, Thomas DM, Cheyne S, Martin A, Morton RL, Simes J, Lee CK. Criteria for assessing evidence for biomarker-targeted therapies in rare cancers-an extrapolation framework. Ther Adv Med Oncol 2024; 16:17588359241273062. [PMID: 39229469 PMCID: PMC11369883 DOI: 10.1177/17588359241273062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/09/2024] [Indexed: 09/05/2024] Open
Abstract
Background Advances in targeted therapy development and tumor sequencing technology are reclassifying cancers into smaller biomarker-defined diseases. Randomized controlled trials (RCTs) are often impractical in rare diseases, leading to calls for single-arm studies to be sufficient to inform clinical practice based on a strong biological rationale. However, without RCTs, favorable outcomes are often attributed to therapy but may be due to a more indolent disease course or other biases. When the clinical benefit of targeted therapy in a common cancer is established in RCTs, this benefit may extend to rarer cancers sharing the same biomarker. However, careful consideration of the appropriateness of extending the existing trial evidence beyond specific cancer types is required. A framework for extrapolating evidence for biomarker-targeted therapies to rare cancers is needed to support transparent decision-making. Objectives To construct a framework outlining the breadth of criteria essential for extrapolating evidence for a biomarker-targeted therapy generated from RCTs in common cancers to different rare cancers sharing the same biomarker. Design A series of questions articulating essential criteria for extrapolation. Methods The framework was developed from the core topics for extrapolation identified from a previous scoping review of methodological guidance. Principles for extrapolation outlined in guidance documents from the European Medicines Agency, the US Food and Drug Administration, and Australia's Medical Services Advisory Committee were incorporated. Results We propose a framework for assessing key assumptions of similarity of the disease and treatment outcomes between the common and rare cancer for five essential components: prognosis of the biomarker-defined cancer, biomarker test analytical validity, biomarker actionability, treatment efficacy, and safety. Knowledge gaps identified can be used to prioritize future studies. Conclusion This framework will allow systematic assessment, standardize regulatory, reimbursement and clinical decision-making, and facilitate transparent discussions between key stakeholders in drug assessment for rare biomarker-defined cancers.
Collapse
Affiliation(s)
- Doah Cho
- National Health and Medical Research Council Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Australia
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Locked Bag 77, Camperdown, NSW 1450, Australia
| | - Sarah J. Lord
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Robyn Ward
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Maarten IJzerman
- Faculty of Medicine, Dentistry and Health Sciences, Centre for Health Policy, University of Melbourne Centre for Cancer Research, Parkville, VIC, Australia
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Andrew Mitchell
- Department of Health Economics Wellbeing and Society, The Australian National University, Canberra, ACT, Australia
| | - David M. Thomas
- Centre for Molecular Oncology, University of New South Wales, Sydney, NSW, Australia
| | - Saskia Cheyne
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Andrew Martin
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- Centre for Clinical Research, University of Queensland, St Lucia, QLD, Australia
| | - Rachael L. Morton
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - John Simes
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Chee Khoon Lee
- Faculty of Medicine and Health, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
26
|
Zahednezhad F, Allahyari S, Sarfraz M, Zakeri-Milani P, Feyzizadeh M, Valizadeh H. Liposomal drug delivery systems for organ-specific cancer targeting: early promises, subsequent problems, and recent breakthroughs. Expert Opin Drug Deliv 2024; 21:1363-1384. [PMID: 39282895 DOI: 10.1080/17425247.2024.2394611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/16/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Targeted liposomal systems for cancer intention have been recognized as a specific and robust approach compared to conventional liposomal delivery systems. Cancer cells have a unique microenvironment with special over-expressed receptors on their surface, providing opportunities for discovering novel and effective drug delivery systems using active targeting. AREAS COVERED Smartly targeted liposomes, responsive to internal or external stimulations, enhance the delivery efficiency by increasing accumulation of the encapsulated anti-cancer agent in the tumor site. The application of antibodies and aptamers against the prevalent cell surface receptors is a potent and ever-growing field. Moreover, immuno-liposomes and cancer vaccines as adjuvant chemotherapy are also amenable to favorable immune modulation. Combinational and multi-functional systems are also attractive in this regard. However, potentially active targeted liposomal drug delivery systems have a long path to clinical acceptance, chiefly due to cross-interference and biocompatibility affairs of the functionalized moieties. EXPERT OPINION Engineered liposomal formulations have to be designed based on tissue properties, including surface chemistry, charge, and microvasculature. In this paper, we aimed to investigate the updated targeted liposomal systems for common cancer therapy worldwide.
Collapse
Affiliation(s)
- Fahimeh Zahednezhad
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Saeideh Allahyari
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Feyzizadeh
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
27
|
Nami B, Wang Z. A Non-Canonical p75HER2 Signaling Pathway Underlying Trastuzumab Action and Resistance in Breast Cancer. Cells 2024; 13:1452. [PMID: 39273024 PMCID: PMC11394428 DOI: 10.3390/cells13171452] [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: 07/14/2024] [Revised: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Overexpression of HER2 occurs in 25% of breast cancer. Targeting HER2 has proven to be an effective therapeutic strategy for HER2-positive breast cancer. While trastuzumab is the most commonly used HER2 targeting agent, which has significantly improved outcomes, the overall response rate is low. To develop novel therapies to boost trastuzumab efficacy, it is critical to identify the mechanisms underlying trastuzumab action and resistance. We recently showed that the inhibition of breast cancer cell growth by trastuzumab is not through the inhibition of HER2 canonical signaling. Here we report the identification of a novel non-canonical HER2 signaling pathway and its interference by trastuzumab. We showed that HER2 signaled through a non-canonical pathway, regulated intramembrane proteolysis (RIP). In this pathway, HER2 is first cleaved by metalloprotease ADAM10 to produce an extracellular domain (ECD) that is released and the p95HER2 that contains the transmembrane domain (TM) and intracellular domain (ICD). p95HER2, if further cleaved by an intramembrane protease, γ-secretase, produced a soluble ICD p75HER2 with nuclear localization signal (NLS). p75HER2 is phosphorylated and translocated to the nucleus. Nuclear p75HER2 promotes cell proliferation. Trastuzumab targets this non-canonical HER2 pathway via inhibition of the proteolytic cleavage of HER2 by both ADAM10 and γ-secretase. However, p75HER2 pathway also confers resistance to trastuzumab once aberrantly activated. Combination of trastuzumab with ADAM10 and γ-secretase inhibitors completely blocks p75HER2 production in both BT474 and SKBR3 cells. We concluded that HER2 signals through the RIP signaling pathway that promotes cell proliferation and is targeted by trastuzumab. The aberrant HER2 RIP signaling confers resistance to trastuzumab that could be overcome by the application of inhibitors to ADAM10 and γ-secretase.
Collapse
Affiliation(s)
| | - Zhixiang Wang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
28
|
Li Z, Song Z, Hong W, Yang N, Wang Y, Jian H, Liang Z, Hu S, Peng M, Yu Y, Wang Y, Jiao Z, Zhao K, Song K, Li Y, Shi W, Lu S. SHR-A1811 (antibody-drug conjugate) in advanced HER2-mutant non-small cell lung cancer: a multicenter, open-label, phase 1/2 study. Signal Transduct Target Ther 2024; 9:182. [PMID: 39004647 PMCID: PMC11247081 DOI: 10.1038/s41392-024-01897-y] [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: 02/07/2024] [Revised: 05/27/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
A dose-escalation and expansion, phase 1/2 study (ClinicalTrials.gov, NCT04818333) was conducted to assess the novel antibody-drug conjugate SHR-A1811 in pretreated HER2-altered advanced non-small cell lung cancer (NSCLC). Here, we report results from the phase 1 portion. Patients who had previously failed or were intolerant to platinum-based chemotherapy were enrolled and received SHR-A1811 intravenously at doses of 3.2 to 8.0 mg/kg every 3 weeks. Dose escalation followed a Bayesian logistic regression model that included overdose control, with subsequent selection of tolerable levels for dose expansion. Overall, 63 patients were enrolled, including 43 receiving a recommended dose for expansion of 4.8 mg/kg. All patients had HER2-mutant disease. Dose-limiting toxicity occurred in one patient in the 8.0 mg/kg dose cohort. Grade ≥ 3 treatment-related adverse events occurred in 29 (46.0%) patients. One patient in the 6.4 mg/kg cohort died due to interstitial lung disease. As of April 11, 2023, the 4.8 mg/kg cohort showed an objective response rate of 41.9% (95% CI 27.0-57.9), and a disease control rate of 95.3% (95% CI 84.2-99.4). The median duration of response was 13.7 months, with 13 of 18 responses ongoing. The median progression-free survival was 8.4 months (95% CI 7.1-15.0). SHR-A1811 demonstrated favourable safety and clinically meaningful efficacy in pretreated advanced HER2-mutant NSCLC.
Collapse
Affiliation(s)
- Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Zhengbo Song
- Phase I Clinical Trial Ward, Zhejiang Cancer Hospital, Hangzhou, 310000, China
| | - Wei Hong
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China
| | - Nong Yang
- Department of Lung & Gastrointestinal Oncology, Hunan Cancer Hospital, Changsha, 410031, China
| | - Yongsheng Wang
- Thoracic Oncology Ward/Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hong Jian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Zibin Liang
- Department of Thoracic Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Wuhan, 430000, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430200, China
| | - Yan Yu
- Department of Thoracic Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yan Wang
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zicong Jiao
- Geneplus-Beijing, Co., Ltd., Beijing, 102206, China
| | - Kaijing Zhao
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - Ke Song
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - You Li
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - Wei Shi
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
29
|
Ray S, Gupta S, Panda G, Chatterjee P, Das A, Patawri P, Hosseinzadeh P, Ray A, Banerjee S. Identification of pseudobaptigenin as a novel polyphenol-based multi-target antagonist of different hormone receptors for breast cancer therapeutics. J Biomol Struct Dyn 2024; 42:5502-5514. [PMID: 37409735 DOI: 10.1080/07391102.2023.2226750] [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: 02/15/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023]
Abstract
Breast cancer (BC) is one of the most prevalent cancers in the world and is one of the major reasons for the death of women worldwide. BC is majorly categorized based on the presence or absence of three cell receptors ER, PR and HER2. The latest treatment for BC involves interfering with the production and action of hormones such as estrogen and progesterone. These hormones bind with receptors such as ER and PR and enhance the growth and proliferation of the BC cells. Although the available are effective, the increasing resistance and side effects related to hormonal imbalance are significant and hence there is a need for designing. On the other hand, plant-derivative products have gained a lot of popularity for their promising anti-cancerous activities. Polyphenols are one such group of plant derivatives that have proven to be useful against cancer. In the present study, an in-silico approach was used to search for a polyphenol that can inhibit ER. In this work, a total of 750 polyphenols were taken into consideration. This number was narrowed down to 55, based on their ADMET properties. These 55 polyphenols were then docked to the receptors, ER, PR and HER2. The molecular docking was followed by Molecular Dynamics (MD) simulations. Based on molecular docking and MD simulation results it was concluded that Pseudobaptigenin has the potential to be an inhibitor of ER, PR and HER2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sarbajit Ray
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Suchetana Gupta
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Gayatri Panda
- Department of Computational Biology, Indraprastha Institute of Information Technoloy, New Delhi, India
| | - Prarthana Chatterjee
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Anoushka Das
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Purvi Patawri
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Parisa Hosseinzadeh
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Arjun Ray
- Department of Computational Biology, Indraprastha Institute of Information Technoloy, New Delhi, India
| | - Satarupa Banerjee
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
30
|
Khoirunnisa SM, Suryanegara FDA, de Jong LA, Setiawan D, Postma MJ. A Systematic Review of Health-Related Quality of Life in Women with HER2-Positive Metastatic Breast Cancer Treated with Trastuzumab. PHARMACOECONOMICS - OPEN 2024; 8:511-524. [PMID: 38489021 PMCID: PMC11252097 DOI: 10.1007/s41669-024-00478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Many trials of human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer treatment with trastuzumab have provided evidence of improved clinical outcomes. This systematic review examined how a regimen that includes trastuzumab affects patients' health-related quality of life (HRQoL) during and after treatment. METHODS A systematic search for articles published up to February 2023 without restrictions of language or publication year was performed using the Pubmed, Embase, and Scopus databases. We included studies of women aged > 18 years with metastatic HER2-positive breast cancer treated with a trastuzumab-containing regimen. We assessed the quality of the studies using the Cochrane Risk of Bias (RoB) tool (2.0) for randomized controlled trials (RCTs) and the Risk Of Bias In Non-randomised Studies-of Interventions (ROBINS-I) tool for cross-sectional studies. We used Microsoft Excel to extract and synthesize data, and documented the review procedure following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS In total, eight studies compared 1104 trastuzumab-treated patients and 1003 non-trastuzumab-treated patients. Most studies were RCTs (n = 7) and one was a prospective observational study. All the included studies used the EORTC-QLQ-C30, EORTC-QLQ-BR23, FACT-B, or FACT-G questionnaires. During treatment, patients taking regimens that included trastuzumab showed clinical improvement in HRQoL, social functioning, and role functioning. After the treatment, patients' HRQoL profiles in the trastuzumab and chemotherapy arms were similar. However, trastuzumab (versus chemotherapy) treatment led to clinically improved functional status, role and physical functioning, and fatigue. The quality assessment revealed some concerns in most RCTs, with the risk of bias being high in two studies, low in one study, and moderate in the cross-sectional study. CONCLUSIONS Trastuzumab-containing regimens administered to HER2-positive breast cancer patients at the metastatic stage evidenced beneficial effects on total HRQoL during and after treatment. Upon therapy cessation, patients' HRQoL scores for both interventions improved. Nevertheless, HRQoL profiles of patients treated with trastuzumab were more favorable, particularly for functional status, role functioning, physical well-being, and fatigue. CLINICAL TRIALS REGISTRATION This review was registered in PROSPERO (registration number: CRD42021259826).
Collapse
Affiliation(s)
- Sudewi Mukaromah Khoirunnisa
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands.
- Science in Healthy Aging and healthcaRE (SHARE) Institute, University of Groningen, 9713 GZ, Groningen, The Netherlands.
- Department of Pharmacy, Institut Teknologi Sumatera, Lampung Selatan, 35365, Indonesia.
| | - Fithria Dyah Ayu Suryanegara
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Science in Healthy Aging and healthcaRE (SHARE) Institute, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Department of Pharmacy, Universitas Islam Indonesia, Yogyakarta, 55584, Indonesia
| | - Lisa Aniek de Jong
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Science in Healthy Aging and healthcaRE (SHARE) Institute, University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Didik Setiawan
- Faculty of Pharmacy, Universitas Muhammadiyah Purwokerto, Banyumas, 53182, Indonesia
- Centre for Health Economic Studies, Universitas Muhammadiyah Purwokerto, Banyumas, 53182, Indonesia
| | - Maarten Jacobus Postma
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Science in Healthy Aging and healthcaRE (SHARE) Institute, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Department of Economics, Econometrics and Finance, Faculty of Economics and Business, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Airlangga, Surabaya, 60115, Indonesia
- Centre of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, 45363, Indonesia
| |
Collapse
|
31
|
Nishimura R, Fujiki Y, Taira T, Miyaki T, Kanemitsu S, Yotsumoto D, Teraoka M, Kawano J, Gondo N, Mitsueda R, Baba S, Ohi Y, Rai Y, Sagara Y, Sagara Y. The Clinicopathological and Prognostic Significance of HER2-Low Breast Cancer: A Comparative Analysis Between HER2-Low and HER2-Zero Subtypes. Clin Breast Cancer 2024; 24:431-438. [PMID: 38472058 DOI: 10.1016/j.clbc.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/19/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND HER2-low breast cancer (BC) is a newly defined subset of HER2-negative BC. However, it is still uncertain whether HER2-low BC can be categorized as a distinct biological/clinical subgroup with any prognostic significance. METHODS Invasive BC cases (n = 10,215) with Stage I-III were retrospectively analyzed to determine the HER2 status. The HER2 status was then divided into 3 groups: HER2-0, HER2-low, and HER2-positive. RESULTS The HER2 status was classified as HER2-0 in 1,227 cases (12.0%), HER2-low in 7,209 cases (70.6%), and HER2-positive in 1779 cases (17.4%). HER2-low cases had more positive nodes and were significantly associated with positive ER/PgR, lower nuclear grade, and lower Ki-67 index. HER2-0 had the lowest OS rate in the primary cases and after recurrence. HER2-0 in the node positive group had the lowest OS and was significantly different from HER2-low in the same group. The pathological complete response (pCR) rate for NAC was lowest in the HER2-low group. The DFS after NAC was significantly better in all the pCR cases, regardless of the HER2 status. However, the DFS was significantly lower in the HER2-low non-pCR cases. CONCLUSION HER2-low accounted for 70% of the cases and correlated with favorable biological markers. The HER2-low group had a significantly better OS than the HER2-0 group. However, the response to NAC was low in the HER2-low group, and this group had the poorest prognosis among all the non-pCR cases. These findings indicate that HER2-low may have a different biology and prognosis and therefore should be classified as a new entity.
Collapse
Affiliation(s)
- Reiki Nishimura
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan.
| | - Yoshitaka Fujiki
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Tetsuhiko Taira
- Clinical Oncology and Pathology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Toshiko Miyaki
- Clinical Oncology and Pathology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Shuichi Kanemitsu
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Daisuke Yotsumoto
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Megumi Teraoka
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Junko Kawano
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Naomi Gondo
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Reiko Mitsueda
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Shinichi Baba
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Yasuyo Ohi
- Pathology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Yoshiaki Rai
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Yoshiaki Sagara
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| | - Yasuaki Sagara
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima City, Japan
| |
Collapse
|
32
|
Lan Y, Zhao J, Zhao F, Li J, Li X. Partial response to trastuzumab deruxtecan (DS8201) following progression in HER2-amplified breast cancer with pulmonary metastases managed with disitamab vedotin (RC48): a comprehensive case report and literature review. Front Oncol 2024; 14:1338661. [PMID: 38952555 PMCID: PMC11215061 DOI: 10.3389/fonc.2024.1338661] [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: 02/16/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
Breast cancer remains one of the predominant malignancies worldwide. In the context of inoperable advanced or metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer, systemic management primarily relies on HER2-targeting monoclonal antibodies. With the successful development of anti-HER2 antibody-drug conjugates (ADCs), these agents have been increasingly integrated into therapeutic regimens for metastatic breast cancer. Here, we present the case of a 42-year-old female patient with HER2-positive pulmonary metastatic breast cancer who underwent an extensive treatment protocol. This protocol included chemotherapy, radiation therapy, hormonal therapy, surgical intervention on the breast, and anti-HER2 therapies. The anti-HER2 therapies involved both singular and dual targeting strategies using trastuzumab and the ADC disitamab vedotin (RC48) over an 8-year period. After experiencing disease progression following HER2-targeted therapy with RC48, the patient achieved noticeable partial remission through a therapeutic regimen that combined trastuzumab deruxtecan (DS8201) and tislelizumab. The data suggest a promising role for DS8201 in managing advanced stages of HER2-amplified metastatic breast cancer, especially in cases that demonstrate progression after initial HER2-directed therapies using ADCs. Furthermore, its combination with anti-PD-1 agents enhances therapeutic efficacy by augmenting the anti-tumoral immune response.
Collapse
Affiliation(s)
- Yanfang Lan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jiahui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fangrui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
33
|
Metin NO, Balcı S, Metin Y, Taşçı F, Gözükara MG. Correlation Between Quantitative Parameters Obtained by Dual Energy Spectral CT and Prognostic Histopathological Factors and Biomarkers in Breast Cancer. Clin Breast Cancer 2024; 24:e279-e288. [PMID: 38423947 DOI: 10.1016/j.clbc.2024.01.022] [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: 10/21/2023] [Revised: 01/25/2024] [Accepted: 01/28/2024] [Indexed: 03/02/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate the correlation between quantitative parameters obtained by dual energy spectral computed tomography (DESCT) and various histopathological factors and biomarkers associated with the prognosis of breast cancer. MATERIALS AND METHODS Quantitative parameters such as iodine content (IC), normalized IC (nIC), iodine enhancement (IE) and normalized IE (nIE) were measured on virtual monochromatic images and iodine mapping images obtained from DESCT in 116 female breast cancer patients. The relationship between these parameters and prognostic biomarkers such as estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) status and Ki67 levels, as well as the correlation with histological grade (HG), lymphovascular invasion (LVI), and metastatic axillary lymphadenopathy (LAP) were evaluated. RESULTS ER-negative tumors had significantly higher values of IC, nIC, IE, and nIE compared to ER-positive tumors. PR-negative tumors had significantly higher values of IE and nIEc compared to PR-positive tumors. HER2 overexpressed and Ki-67 high proliferation tumors showed significantly higher values of all quantitative parameters compared to HER2 negative and Ki-67 low proliferation tumors. All quantitative parameters were significantly higher in HG 3 tumors, tumors with detected LVI, and tumors with metastatic axillary LAP compared to low-grade tumors, LVI-negative tumors and tumors without metastatic axillary lymph nodes, respectively. CONCLUSION Quantitative parameters of IC and IE obtained from DESCT have shown potential for predicting prognosis in breast cancer patients. Higher values of these parameters have been found to correlate with poor prognostic biomarkers and histopathological features. These results suggest that quantitative DESCT imaging may offer an additional benefit in the noninvasive prediction of breast cancer prognosis.
Collapse
Affiliation(s)
| | - Sinan Balcı
- Department of Radiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yavuz Metin
- Department of Radiology, Ankara University Faculty of Medicine, Ankara, Turkey.
| | - Filiz Taşçı
- Department of Radiology, Recep Tayyip Erdogan University Faculty of Medicine, Rize, Turkey
| | | |
Collapse
|
34
|
Ujlaky-Nagy L, Szöllősi J, Vereb G. Disrupting EGFR-HER2 Transactivation by Pertuzumab in HER2-Positive Cancer: Quantitative Analysis Reveals EGFR Signal Input as Potential Predictor of Therapeutic Outcome. Int J Mol Sci 2024; 25:5978. [PMID: 38892166 PMCID: PMC11173106 DOI: 10.3390/ijms25115978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Pertuzumab (Perjeta®), a humanized antibody binding to the dimerization arm of HER2 (Human epidermal growth factor receptor-2), has failed as a monotherapy agent in HER2 overexpressing malignancies. Since the molecular interaction of HER2 with ligand-bound EGFR (epidermal growth factor receptor) has been implied in mitogenic signaling and malignant proliferation, we hypothesized that this interaction, rather than HER2 expression and oligomerization alone, could be a potential molecular target and predictor of the efficacy of pertuzumab treatment. Therefore, we investigated static and dynamic interactions between HER2 and EGFR molecules upon EGF stimulus in the presence and absence of pertuzumab in HER2+ EGFR+ SK-BR-3 breast tumor cells using Förster resonance energy transfer (FRET) microscopy and fluorescence correlation and cross-correlation spectroscopy (FCS/FCCS). The consequential activation of signaling and changes in cell proliferation were measured by Western blotting and MTT assay. The autocorrelation functions of HER2 diffusion were best fitted by a three-component model corrected for triplet formation, and among these components the slowly diffusing membrane component revealed aggregation induced by EGFR ligand binding, as evidenced by photon-counting histograms and co-diffusing fractions. This aggregation has efficiently been prevented by pertuzumab treatment, which also inhibited the post-stimulus interaction of EGFR and HER2, as monitored by changes in FRET efficiency. Overall, the data demonstrated that pertuzumab, by hindering post-stimulus interaction between EGFR and HER2, inhibits EGFR-evoked HER2 aggregation and phosphorylation and leads to a dose-dependent decrease in cell proliferation, particularly when higher amounts of EGF are present. Consequently, we propose that EGFR expression on HER2-positive tumors could be taken into consideration as a potential biomarker when predicting the outcome of pertuzumab treatment.
Collapse
Affiliation(s)
- László Ujlaky-Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| |
Collapse
|
35
|
Kim BK, Kim B, You SH, Jang MS, Im GH, Kim KH. Early therapy evaluation of intra-arterial trastuzumab injection in a human breast cancer xenograft model using multiparametric MR imaging. PLoS One 2024; 19:e0300171. [PMID: 38701062 PMCID: PMC11068173 DOI: 10.1371/journal.pone.0300171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/22/2024] [Indexed: 05/05/2024] Open
Abstract
PURPOSE To investigate the treatment efficacy of intra-arterial (IA) trastuzumab treatment using multiparametric magnetic resonance imaging (MRI) in a human breast cancer xenograft model. MATERIALS AND METHODS Human breast cancer cells (BT474) were stereotaxically injected into the brains of nude mice to obtain a xenograft model. The mice were divided into four groups and subjected to different treatments (IA treatment [IA-T], intravenous treatment [IV-T], IA saline injection [IA-S], and the sham control group). MRI was performed before and at 7 and 14 d after treatment to assess the efficacy of the treatment. The tumor volume, apparent diffusion coefficient (ADC), and dynamic contrast-enhanced (DCE) MRI parameters (Ktrans, Kep, Ve, and Vp) were measured. RESULTS Tumor volumes in the IA-T group at 14 d after treatment were significantly lower than those in the IV-T group (13.1 mm3 [interquartile range 8.48-16.05] vs. 25.69 mm3 [IQR 20.39-30.29], p = 0.005), control group (IA-S, 33.83 mm3 [IQR 32.00-36.30], p<0.01), and sham control (39.71 mm3 [IQR 26.60-48.26], p <0.001). The ADC value in the IA-T group was higher than that in the control groups (IA-T, 7.62 [IQR 7.23-8.20] vs. IA-S, 6.77 [IQR 6.48-6.87], p = 0.044 and vs. sham control, 6.89 [IQR 4.93-7.48], p = 0.004). Ktrans was significantly decreased following the treatment compared to that in the control groups (p = 0.002 and p<0.001 for vs. IA-S and sham control, respectively). Tumor growth was decreased in the IV-T group compared to that in the sham control group (25.69 mm3 [IQR 20.39-30.29] vs. 39.71 mm3 [IQR 26.60-48.26], p = 0.27); there was no significant change in the MRI parameters. CONCLUSION IA treatment with trastuzumab potentially affects the early response to treatment, including decreased tumor growth and decrease of Ktrans, in a preclinical brain tumor model.
Collapse
Affiliation(s)
- Bo Kyu Kim
- Department of Radiology, Korea University Anam Hospital, Seoul, Korea
| | - Byungjun Kim
- Department of Radiology, Korea University Anam Hospital, Seoul, Korea
| | - Sung-Hye You
- Department of Radiology, Korea University Anam Hospital, Seoul, Korea
| | - Moon-Sun Jang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul, Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research, SungKyunkwan University, Suwon, Korea
| | - Keon-Ha Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Shramova EI, Deyev SM, Proshkina GM. A Vector Nanoplatform for the Bioimaging of Deep-Seated Tumors. Acta Naturae 2024; 16:72-81. [PMID: 39188260 PMCID: PMC11345090 DOI: 10.32607/actanaturae.27425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 05/16/2024] [Indexed: 08/28/2024] Open
Abstract
Today, in preclinical studies, optical bioimaging based on luminescence and fluorescence is indispensable in studying the development of neoplastic transformations, the proliferative activity of the tumor, its metastatic potential, as well as the therapeutic effect of antitumor agents. In order to expand the capabilities of optical imaging, sensors based on the bioluminescence resonance energy transfer (BRET) mechanism and, therefore, independent of an external light source are being developed. A targeted nanoplatform based on HER2-specific liposomes whose internal environment contains a genetically encoded BRET sensor was developed in this study to visualize deep-seated tumors characterized by overexpression of human epidermal growth factor receptor type 2 (HER2). The BRET sensor is a hybrid protein consisting of the highly catalytic luciferase NanoLuc (an energy donor) and a LSSmKate1 red fluorescent protein with a large Stokes shift (an energy acceptor). During the bioimaging of disseminated intraperitoneal tumors formed by HER2-positive SKOV3.ip1cells of serous ovarian cystadenocarcinoma, it was shown that the developed system is applicable in detecting deep-seated tumors of a certain molecular profile. The developed system can become an efficient platform for optimizing preclinical studies of novel targeted drugs.
Collapse
Affiliation(s)
- E. I. Shramova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of science, Moscow, 117997 Russian Federation
| | - S. M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of science, Moscow, 117997 Russian Federation
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991 Russian Federation
- National Research Centre “Kurchatov Institute”, Moscow, 123098 Russian Federation
| | - G. M. Proshkina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of science, Moscow, 117997 Russian Federation
| |
Collapse
|
37
|
Nomani A, Yousefi S, Sargsyan D, Hatefi A. A quantitative MRI-based approach to estimate the permeation and retention of nanomedicines in tumors. J Control Release 2024; 368:728-739. [PMID: 38493951 PMCID: PMC11412736 DOI: 10.1016/j.jconrel.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Despite the potential of the enhanced permeability and retention (EPR) effect in tumor passive targeting, many nanotherapeutics have failed to produce meaningful clinical outcomes due to the variable and challenging nature of the tumor microenvironment (TME) and EPR effect. This EPR variability across tumors and inconsistent translation of nanomedicines from preclinical to clinical settings necessitates a reliable method to assess its presence in individual tumors. This study aimed to develop a reliable and non-invasive approach to estimate the EPR effect in tumors using a clinically compatible quantitative magnetic resonance imaging (qMRI) technique combined with a nano-sized MRI contrast agent. A quantitative MR imaging was developed using a dynamic contrast-enhanced (DCE) MRI protocol. Then, the permeability and retention of the nano-sized MRI contrast agent were evaluated in three different ovarian xenograft tumor models. Results showed significant differences in EPR effects among the tumor models, with tumor growth influencing the calculated parameters of permeability (Ktrans) and retention (Ve) based on Tofts pharmacokinetic (PK) modeling. Our data indicate that the developed quantitative DCE-MRI method, combined with the Tofts PK modeling, provides a robust and non-invasive approach to screen tumors for their responsiveness to nanotherapeutics. These results imply that the developed qMRI method can be beneficial for personalized cancer treatments by ensuring that nanotherapeutics are administered only to patients with tumors showing sufficient EPR levels.
Collapse
Affiliation(s)
- Alireza Nomani
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA.
| | - Siavash Yousefi
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Davit Sargsyan
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA
| | - Arash Hatefi
- Department of Pharmaceutics, Rutgers University, Piscataway, NJ 08854, USA; Cancer Pharmacology Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA.
| |
Collapse
|
38
|
Ma R, Shi Y, Yan R, Yin S, Bu H, Huang J. Efficacy and safety of trastuzumab deruxtecan in treating human epidermal growth factor receptor 2-low/positive advanced breast cancer:A meta-analysis of randomized controlled trials. Crit Rev Oncol Hematol 2024; 196:104305. [PMID: 38442809 DOI: 10.1016/j.critrevonc.2024.104305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND A novel antibody-drug conjugate, trastuzumab deruxtecan is a combination of a human epidermal growth factor receptor 2 (HER2)-targeting antibody and DNA topoisomerase I inhibitor used to treat HER2-low/positive advanced breast cancer. To determine its safety and efficacy in treating HER2-low/positive advanced breast cancer, we performed a meta-analysis of several randomized clinical trials (RCTs) including DESTINY-Breast02 (NCT03523585), DESTINY-Breast03 (NCT03529110), and DESTINY-Breast04 (NCT03734029). METHODS We searched PubMed, Embase, and the Cochrane Library for RCTs on the efficacy and safety of trastuzumab deruxtecan that were published before May 2023. The efficacy endpoints included median progressive-free survival (PFS), overall survival (OS), duration of response (DOR), overall response rate (ORR), and clinical benefit rate (CBR). The safety endpoints included treatment-related adverse events. Statistical analyses were performed using RevMan 5.4 software. To ensure transparency, this study was registered on the International Prospective Register of Systematic Reviews website (CRD42023414170). RESULTS Three RCTs involving 1689 patients were included. Compared with physician-recommended and conventional treatments, trastuzumab deruxtecan exhibited statistically significant improvements in PFS, ORR, and CBR. The median OS and DOR failed to be combined; however, the analyzed studies showed that they were longer. The incidence of adverse events was generally higher with trastuzumab deruxtecan than with physician-recommended or conventional treatments. CONCLUSION The results of this study suggest that trastuzumab deruxtecan is more effective in treating HER2-low/positive advanced breast cancer than physician-recommended or conventional treatments. However, trastuzumab deruxtecan-related adverse drug reactions should be closely monitored because of its higher incidence of adverse events.
Collapse
Affiliation(s)
- Rui Ma
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Tuanbo New City West, Jinghai, Tianjin, China
| | - Yixun Shi
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Tuanbo New City West, Jinghai, Tianjin, China
| | - Ruijuan Yan
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Tuanbo New City West, Jinghai, Tianjin, China
| | - Shiqing Yin
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Tuanbo New City West, Jinghai, Tianjin, China
| | - Huanen Bu
- School of Public Health, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Tuanbo New City West, Jinghai, Tianjin, China.
| | - Jie Huang
- Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440 Jiyan Road, Jinan, Shandong, China.
| |
Collapse
|
39
|
El-Tanani M, Nsairat H, Aljabali AA, Matalka II, Alkilany AM, Tambuwala MM. Dual-loaded liposomal carriers to combat chemotherapeutic resistance in breast cancer. Expert Opin Drug Deliv 2024; 21:309-324. [PMID: 38284386 DOI: 10.1080/17425247.2024.2311812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
INTRODUCTION The resistance to chemotherapy is a significant hurdle in breast cancer treatment, prompting the exploration of innovative strategies. This review discusses the potential of dual-loaded liposomal carriers to combat chemoresistance and improve outcomes for breast cancer patients. AREAS COVERED This review discusses breast cancer chemotherapy resistance and dual-loaded liposomal carriers. Drug efflux pumps, DNA repair pathways, and signaling alterations are discussed as chemoresistance mechanisms. Liposomes can encapsulate several medicines and cargo kinds, according to the review. It examines how these carriers improve medication delivery, cancer cell targeting, and tumor microenvironment regulation. Also examined are dual-loaded liposomal carrier improvement challenges and techniques. EXPERT OPINION The use of dual-loaded liposomal carriers represents a promising and innovative strategy in the battle against chemotherapy resistance in breast cancer. This article has explored the various mechanisms of chemoresistance in breast cancer, emphasizing the potential of dual-loaded liposomal carriers to overcome these challenges. These carriers offer versatility, enabling the encapsulation and precise targeting of multiple drugs with different modes of action, a crucial advantage when dealing with the complexity of breast cancer treatment.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Pharmacy, Yarmouk University, Irbid, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Department of Pathology and Microbiology, Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | | |
Collapse
|
40
|
Ye J, Liao X, Qiu Y, Wei Q, Bao Y. A systematic review and meta-analysis for human epidermal growth factor receptor 2 on upper tract urothelial carcinoma patients. TUMORI JOURNAL 2024; 110:25-33. [PMID: 37555322 DOI: 10.1177/03008916231186178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
OBJECTIVE The expression and significance of human epidermal growth factor receptor 2 (Her2) in upper tract urothelial carcinoma (UTUC) remains controversial. Thus, we aimed to systemically review the Her2 expression in UTUC patients and its relationship with pathological characters and clinical outcomes with meta-analysis. MATERIALS AND METHODS A systematically computerized search in PubMed, Scopus, Embase and Cochrane was conducted. From a total of 454 related articles, 35 articles were finally reviewed and 16 papers were chosen for further analysis. Pathological characters included tumor stage, grade, lymph node metastasis (LNM) and lymphovascular invasion (LVI). The clinical outcomes included overall survival (OS), recurrence-free survival (RFS), cancer specific survival (CSS), metastatic-free survival (MFS) and progression-free survival (PFS). RevMan software was used for meta-analyses. RESULTS In total 16 studies from 1994 to 2020 were chosen, 14 studies used immunohistochemistry to assess the expression of Her2 and 5 studies used in situ hybridization, with a positive rate of 0 to 74.0% and 7.2 to 18.1%, respectively. Her2-positive was significantly associated with stage (pooled HR 1.86; 95 % CI 1.43-2.42), grade (pooled HR 2.81; 95 % CI 1.01-7.85) and LNM (pooled HR 1.93; 95 % CI 1.18-3.15). However, there was no statistically relationship between Her2-positive with LVI (pooled HR 1.48; 95 % CI 0.64-3.46) and RFS (pooled HR 1.41; 95 % CI 0.98-1.83). CONCLUSIONS This review indicated that UTUC patients with Her2-positive tended to develop higher stage and grade tumors and LNM. The Her2 expression in UTUC patients deserves further investigation in the future.
Collapse
Affiliation(s)
- Jianjun Ye
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xinyang Liao
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Qiu
- Department of Thyroid and Breast Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Qiang Wei
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yige Bao
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Sharma R, Mukherjee A, Kumar A, Sarma HD. Evaluation of 177Lu-Labeled Pertuzumab F(ab') 2 Fragments for HER2-Positive Cancer Targeting: A Comparative In Vitro and In Vivo Study. Cancer Biother Radiopharm 2024; 39:64-74. [PMID: 38363819 DOI: 10.1089/cbr.2023.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024] Open
Abstract
Background: Radiolabeled antibody fragments present a promising opportunity as theranostic agents, offering distinct advantages over whole antibodies. In this study, the authors investigate the potential of [177Lu]Lu-DTPA-F(ab')2-pertuzumab as a theranostic agent for precise targeting of human epidermal growth factor receptor 2 (HER2)-positive cancers. Additionally, the authors aim to quantitatively assess the binding synergism in the presence of cold trastuzumab. Materials and Methods: F(ab')2-pertuzumab was prepared by pepsin digestion and conjugated with a bifunctional chelator. The immunoconjugate was radiolabeled with 177Lu and characterized by chromatography techniques. Binding parameters (affinity, specificity, and immunoreactivity) and cellular binding enhancement studies were evaluated in HER2-overexpressing and triple-negative cell lines. The in vivo enhancement in tumor uptake of the radiolabeled immunoformulation was assessed in severe combined immunodeficient (SCID) mice bearing tumors, both in the presence and absence of unlabeled trastuzumab. Results: The formulation of [177Lu]Lu-DTPA-F(ab')2-pertuzumab could be prepared in high yields and with consistent radiochemical purity, ensuring reproducibility. Comprehensive in vitro and in vivo evaluation studies confirmed high specificity and immunoreactivity of the formulation toward HER2 receptors. Binding synergism of radiolabeled pertuzumab fragments in the presence of trastuzumab to HER2 receptors was observed. Conclusions: The radioformulation of [177Lu]Lu-DTPA-F(ab')2-pertuzumab holds great promise as a targeted approach for addressing HER2-positive cancers. A potentially effective strategy to amplify therapeutic efficacy involves dual epitope targeting by combining radiolabeled pertuzumab with cold trastuzumab.
Collapse
Affiliation(s)
- Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai, India
| | - Anuj Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Haladhar Dev Sarma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| |
Collapse
|
42
|
Baek SK, Jeong JH, Jung K, Ahn HK, Kim MH, Sohn J, Park IH, Ahn JS, Lee DW, Im SA, Sim SH, Lee KS, Hyun Kim J, Shim HJ, Chae Y, Koh SJ, Lee H, Lee J, Byun JH, Seol Y, Lee EM, Jee HJ, An H, Park EB, Suh YJ, Lee KE, Park YH. A nationwide real-world study for evaluation of effectiveness and safety of T-DM1 in patients with HER2-positive metastatic breast cancer in Korea (KCSG BR19-15). Ther Adv Med Oncol 2024; 16:17588359231225029. [PMID: 38288157 PMCID: PMC10823858 DOI: 10.1177/17588359231225029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/19/2023] [Indexed: 01/31/2024] Open
Abstract
Purpose This study aimed to investigate clinical practices and factors related to the outcomes of T-DM1 use in patients with HER2-positive metastatic breast cancer (mBC). Methods We included patients with HER2-positive mBC who received T-DM1 as a palliative therapy between August 2017 and December 2018. The safety and outcomes of T-DM1, including overall response rate (ORR), progression-free survival (PFS), and overall survival (OS), were evaluated. A Cox proportional hazards model was used to estimate the hazard ratio and 95% confidence interval (CI) for mortality or progression to HER2-positive mBC. Results In total, 824 patients were enrolled during the study period. The mean age of patients was 58 years, and 516 (62.6%) patients relapsed after curative treatment. Excluding a history of endocrine therapy, 341 (41.4%) patients previously received none or first-line chemotherapy, 179 (21.7%) received second-line therapy, and 303 (36.9%) received third-or later-line chemotherapy before T-DM1 therapy. During a median follow-up of 16.8 months, the ORR was 35%, the median PFS was 6.6 months, and the median OS was not reached. The clinical factors associated with the hazard of progression were age (<65 years), poor performance status (⩾2), advanced line of palliative chemotherapy (⩾2), prior pertuzumab use, and treatment duration of palliative trastuzumab (<10 months). Common grade 3-4 adverse events were thrombocytopenia (n = 107, 13.2%), neutropenia (n = 23, 2.8%), anemia (n = 21, 2.6%), and elevated liver enzyme (n = 20, 2.5%). Hypokalemia (⩽3.0 mmol/L) and any-grade bleeding events occurred in 25 (3.1%) and 94 (22.6%) patients, respectively. Conclusion This is the first nationwide real-world study of T-DM1 use in patients with HER2-positive mBC in Korea. The effectiveness and toxicity profiles of T-DM1 in real-world practice were comparable to those in randomized trials. Moreover, patient factors and previous anti-HER2 therapy could predict the outcomes of T-DM1 therapy.
Collapse
Affiliation(s)
- Sun Kyung Baek
- Department of Internal Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Jae-ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - KyungHae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Kyung Ahn
- Division of Medical Oncology and Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Min Hwan Kim
- Division of Medical Oncology and Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joohyuk Sohn
- Division of Medical Oncology and Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - In Hae Park
- Division of Oncology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Hoon Sim
- Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Keun Seok Lee
- Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Hyun-Jeong Shim
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Gwangju, Republic of Korea
| | - Yeesoo Chae
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Su-Jin Koh
- Department of Hematology and Oncology, Ulsan University Hospital, Ulsan, Republic of Korea
| | - Hyorak Lee
- Division of Hematology/Oncology, Korea Cancer Center Hospital, Seoul, Republic of Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae-Ho Byun
- Division of Medical Oncology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Youngmi Seol
- Division of Hematology and Oncology, Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Eun Mi Lee
- Department of Internal Medicine, Kosin University Gaspel Hospital, Busan, Republic of Korea
| | - Hee-Jung Jee
- Department of Biostatistics, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hyonggin An
- Department of Biostatistics, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Eun Byeol Park
- Department of Biostatistics, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Young Ju Suh
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Kyoung Eun Lee
- Department of Hematology and Oncology, Ewha Womans University Hospital, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 07985, Republic of Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | | |
Collapse
|
43
|
Tanaka T, Suzuki H, Ohishi T, Kaneko MK, Kato Y. Antitumor activities against breast cancers by an afucosylated anti-HER2 monoclonal antibody H 2 Mab-77-mG 2a -f. Cancer Sci 2024; 115:298-309. [PMID: 37942574 PMCID: PMC10823288 DOI: 10.1111/cas.16008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/10/2023] Open
Abstract
Breast cancer patients with high levels of human epidermal growth factor receptor 2 (HER2) expression have worse clinical outcomes. Anti-HER2 monoclonal antibody (mAb) is the most important therapeutic modality for HER2-positive breast cancer. We previously immunized mice with the ectodomain of HER2 to create the anti-HER2 mAb, H2 Mab-77 (mouse IgG1 , kappa). This was then altered to produce H2 Mab-77-mG2a -f, an afucosylated mouse IgG2a . In the present work, we examined the reactivity of H2 Mab-77-mG2a -f and antitumor effects against breast cancers in vitro and in vivo. BT-474, an endogenously HER2-expressing breast cancer cell line, was identified by H2 Mab-77-mG2a -f with a strong binding affinity (a dissociation constant [KD ]: 5.0 × 10-9 M). H2 Mab-77-mG2a -f could stain HER2 of breast cancer tissues in immunohistochemistry and detect HER2 protein in Western blot analysis. Furthermore, H2 Mab-77-mG2a -f demonstrated strong antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) for BT-474 cells. MDA-MB-468, a HER2-negative breast cancer cell line, was unaffected by H2 Mab-77-mG2a -f. Additionally, in the BT-474-bearing tumor xenograft model, H2 Mab-77-mG2a -f substantially suppressed tumor development when compared with the control mouse IgG2a mAb. In contrast, the HER2-negative MDA-MB-468-bearing tumor xenograft model showed no response to H2 Mab-77-mG2a -f. These findings point to the possibility of H2 Mab-77-mG2a -f as a treatment regimen by showing that it has antitumor effects on HER2-positive breast tumors.
Collapse
Affiliation(s)
- Tomohiro Tanaka
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
| | - Hiroyuki Suzuki
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
- Department of Antibody Drug DevelopmentTohoku University Graduate School of MedicineSendaiMiyagiJapan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), NumazuMicrobial Chemistry Research FoundationShizuokaJapan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of OncologyMicrobial Chemistry Research FoundationTokyoJapan
| | - Mika K. Kaneko
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
- Department of Antibody Drug DevelopmentTohoku University Graduate School of MedicineSendaiMiyagiJapan
| | - Yukinari Kato
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
- Department of Antibody Drug DevelopmentTohoku University Graduate School of MedicineSendaiMiyagiJapan
| |
Collapse
|
44
|
Irmak-Yazicioglu MB, Arslan A. Navigating the Intersection of Technology and Depression Precision Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1456:401-426. [PMID: 39261440 DOI: 10.1007/978-981-97-4402-2_20] [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: 09/13/2024]
Abstract
This chapter primarily focuses on the progress in depression precision medicine with specific emphasis on the integrative approaches that include artificial intelligence and other data, tools, and technologies. After the description of the concept of precision medicine and a comparative introduction to depression precision medicine with cancer and epilepsy, new avenues of depression precision medicine derived from integrated artificial intelligence and other sources will be presented. Additionally, less advanced areas, such as comorbidity between depression and cancer, will be examined.
Collapse
Affiliation(s)
| | - Ayla Arslan
- Department of Molecular Biology and Genetics, Üsküdar University, İstanbul, Türkiye.
| |
Collapse
|
45
|
Zheng X, Yin J. Efficacy of texture analysis in determining the gene amplification status of HER2 2+ for invasive ductal carcinoma cases. Minerva Med 2023; 114:832-838. [PMID: 32239879 DOI: 10.23736/s0026-4806.20.06536-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
BACKGROUND Gene amplification of human epidermal growth factor receptor2 (HER2) 2+ is essential to be determined for treatment planning. A search of the PubMed database indicates that the correlation between texture features from dynamic contrast enhanced (DCE)-MRI and HER2 2+ status has not been investigated extensively in invasive ductal carcinoma cases. METHODS Seventy-one DCE-MRI cases of HER2 2+ status verified using fluorescence in-situ hybridization (FISH) were selected, including 36 positive and 35 negative cases. Overall, 279 texture features were derived from lesion regions of interest manually drawn onto the subtraction images between pre- and post-contrast agent. Fisher coefficient, mutual information, minimization of both classification error probability and average correlation coefficients as well as a combination of all three methods (MPF) were independently used to reduce the dimensionality of texture parameters. A popular machine learning algorithm, the Support Vector Machine, was further applied to determine HER2 2+ status. Receiver operating characteristic (ROC) analysis was conducted to evaluate the classification performance. RESULTS Diagnostic accuracy was optimal when the most significant discriminatory features were selected using MPF. The area under ROC curve reached 0.863 with corresponding accuracy, sensitivity and specificity rates of 81.80%, 85.71% and 77.78%, respectively. CONCLUSIONS Texture analysis based on breast MRI delivered consistently high performance with FISH detection and may serve as a useful supplementary tool for determining the gene amplification status of HER2 2+ for cases with invasive ductal carcinoma.
Collapse
Affiliation(s)
- Xu Zheng
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China -
| |
Collapse
|
46
|
Sui L, Yan Y, Jiang T, Ou D, Chen C, Lai M, Ni C, Zhu X, Wang L, Yang C, Li W, Yao J, Xu D. Ultrasound and clinicopathological characteristics-based model for prediction of pathologic response to neoadjuvant chemotherapy in HER2-positive breast cancer: a case-control study. Breast Cancer Res Treat 2023; 202:45-55. [PMID: 37639063 PMCID: PMC10504141 DOI: 10.1007/s10549-023-07057-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND The objective of this study was to develop a model combining ultrasound (US) and clinicopathological characteristics to predict the pathologic response to neoadjuvant chemotherapy (NACT) in human epidermal growth factor receptor 2 (HER2)-positive breast cancer. MATERIALS AND METHODS This is a retrospective study that included 248 patients with HER2-positive breast cancer who underwent NACT from March 2018 to March 2022. US and clinicopathological characteristics were collected from all patients in this study, and characteristics obtained using univariate analysis at p < 0.1 were subjected to multivariate analysis and then the conventional US and clinicopathological characteristics independently associated with pathologic complete response (pCR) from the analysis were used to develop US models, clinicopathological models, and their combined models by the area under the receiver operating characteristic (ROC) curve (AUC), accuracy, sensitivity, and specificity to assess their predictive efficacy. RESULTS The combined model had an AUC of 0.808, a sensitivity of 88.72%, a specificity of 60.87%, and an accuracy of 75.81% in predicting pCR of HER2-positive breast cancer after NACT, which was significantly better than the clinicopathological model (AUC = 0.656) and the US model (AUC = 0.769). In addition, six characteristics were screened as independent predictors, namely the Clinical T stage, Clinical N stage, PR status, posterior acoustic, margin, and calcification. CONCLUSION The conventional US combined with clinicopathological characteristics to construct a combined model has a good diagnostic effect in predicting pCR in HER2-positive breast cancer and is expected to be a useful tool to assist clinicians in effectively determining the efficacy of NACT in HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Lin Sui
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| | - Yuqi Yan
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| | - Tian Jiang
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Di Ou
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Chen Chen
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
- Graduate School, Wannan Medical College, Wuhu, China
| | - Min Lai
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Ni
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Xi Zhu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| | - Liping Wang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Chen Yang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Wei Li
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Jincao Yao
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, China
| | - Dong Xu
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial IntelligenceTaizhou Branch of Zhejiang Cancer Hospital(Taizhou Cancer Hospital), Taizhou, China
| |
Collapse
|
47
|
Ma F, Yan M, Li W, Ouyang Q, Tong Z, Teng Y, Wang Y, Wang S, Geng C, Luo T, Zhong J, Zhang Q, Liu Q, Zeng X, Sun T, Mo Q, Liu H, Cheng Y, Cheng J, Wang X, Nie J, Yang J, Wu X, Wang X, Li H, Ye C, Dong F, Wu S, Zhu X, Xu B. Pyrotinib versus placebo in combination with trastuzumab and docetaxel as first line treatment in patients with HER2 positive metastatic breast cancer (PHILA): randomised, double blind, multicentre, phase 3 trial. BMJ 2023; 383:e076065. [PMID: 37907210 PMCID: PMC10616786 DOI: 10.1136/bmj-2023-076065] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 11/02/2023]
Abstract
OBJECTIVE To assess the efficacy and safety of pyrotinib (an irreversible pan-HER (human epidermal growth factor receptor) inhibitor), trastuzumab, and docetaxel compared with placebo, trastuzumab, and docetaxel for untreated HER2 positive metastatic breast cancer. DESIGN Randomised, double blind, placebo controlled, multicentre, phase 3 trial. SETTING 40 centres in China between 6 May 2019 and 17 January 2022. PARTICIPANTS 590 female patients (median age 52 (interquartile range 46-58) years) with untreated HER2 positive metastatic breast cancer. INTERVENTIONS Eligible patients were randomised 1:1 to receive either oral pyrotinib (400 mg once daily) or placebo, both combined with intravenous trastuzumab (8 mg/kg in cycle 1 and 6 mg/kg in subsequent cycles) and docetaxel (75 mg/m2) on day 1 of each 21 day cycle. Randomisation was stratified by treatment history of trastuzumab in the (neo)adjuvant setting and hormone receptor status. Patients, investigators, and the sponsor's study team were masked to treatment assignment. MAIN OUTCOME MEASURES The primary endpoint was progression-free survival as assessed by the investigator. RESULTS Of the 590 randomised patients, 297 received pyrotinib, trastuzumab, and docetaxel treatment (pyrotinib group), and 293 received placebo, trastuzumab, and docetaxel treatment (placebo group). At data cut-off on 25 May 2022, the median follow-up was 15.5 months. The median progression-free survival according to the investigator was significantly longer in the pyrotinib group than in the placebo group (24.3 (95% confidence interval 19.1 to 33.0) months versus 10.4 (9.3 to 12.3) months; hazard ratio 0.41 (95% confidence interval 0.32 to 0.53); one sided P<0.001). Treatment related adverse events of grade 3 or higher were reported in 267 (90%) of the 297 patients in the pyrotinib group and 224 (76%) of the 293 patients in the placebo group. No treatment related deaths occurred in the pyrotinib group, and one (<1%; diabetic hyperosmolar coma) treatment related death occurred in the placebo group. Survival and toxicities are still under assessment with longer follow-up. CONCLUSIONS Pyrotinib, trastuzumab, and docetaxel showed superiority by significantly improving progression-free survival compared with placebo, trastuzumab, and docetaxel in patients with untreated HER2 positive metastatic breast cancer. The toxicity was manageable. The findings support this dual anti-HER2 regimen as an alternative first line treatment option in this patient population. TRIAL REGISTRATION ClinicalTrials.gov NCT03863223.
Collapse
Affiliation(s)
- Fei Ma
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Yan
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Wei Li
- Department of Oncology, The First Hospital of Jilin University, Changchun, China
| | - Quchang Ouyang
- Breast Internal Medicine Department, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Yuee Teng
- Department of Breast Internal Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Yongsheng Wang
- Breast Surgery, Shandong Cancer Hospital and Institute, Jinan, China
| | - Shusen Wang
- Department of Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Cuizhi Geng
- Breast Center, The Fourth Hospital of Hebei Medical University and Hebei Tumor Hospital, Shijiazhuang, China
| | - Ting Luo
- Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jincai Zhong
- Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qingyuan Zhang
- Ward One of Mammary Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qiang Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohua Zeng
- Breast Cancer Center, Affiliated Cancer Hospital of Chongqing University, Chongqing, China
| | - Tao Sun
- Breast Internal Medicine Department, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Qinguo Mo
- Breast Surgery, Guangxi Medical University Affiliated Tumor Hospital, Nanning, China
| | - Hu Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC West District, Hefei, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| | - Jing Cheng
- Oncology Center Breast Department, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xiaojia Wang
- Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jianyun Nie
- Breast Surgery, Yunnan Cancer Hospital, Kunming, China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinhong Wu
- Department of Breast Oncology, Hubei Cancer Hospital, Wuhan, China
| | - Xinshuai Wang
- Department of Medical Oncology, Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Changsheng Ye
- Department of Breast, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Fangli Dong
- Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Shuchao Wu
- Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Xiaoyu Zhu
- Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Binghe Xu
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
48
|
Ehsasatvatan M, Kohnehrouz BB. The lyophilized chloroplasts store synthetic DARPin G3 as bioactive encapsulated organelles. J Biol Eng 2023; 17:63. [PMID: 37798746 PMCID: PMC10557345 DOI: 10.1186/s13036-023-00383-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND The high cost of fermentation, purification, cold storage and transportation, short shelf life, and sterile delivery methods of biopharmaceuticals, is a matter for producers and consumers as well. Since the FDA has now approved plant cells for large-scale, cost-effective biopharmaceutical production, the isolation and lyophilization of transplastomic chloroplasts can cover concerns about limitations. DARPins are engineered small single-domain proteins that have been selected to bind to HER2 with high affinity and specificity. HER2 is an oncogene involved in abnormal cell growth in some cancers and the target molecule for cancer immunotherapy. RESULTS In this study, we reported the prolonged stability and functionality of DARPin G3 in lyophilized transplastomic tobacco leaves and chloroplasts. Western blot analysis of lyophilized leaves and chloroplasts stored at room temperature for up to nine months showed that the DARPin G3 protein was stable and preserved proper folding. Lyophilization of leaves and isolated chloroplasts increased DARPin G3 protein concentrations by 16 and 32-fold, respectively. The HER2-binding assay demonstrated that the chloroplast-made DARPin G3 can maintain its stability and binding activity without any affinity drop in lyophilized leaf materials throughout this study for more than nine months at room temperature. CONCLUSION Lyophilization of chloroplasts expressing DARPin G3 would further reduce costs and simplify downstream processing, purification, and storage. Compressed packages of lyophilized chloroplasts were much more effective than lyophilized transplastomic leaves considering occupied space and downstream extraction and purification of DARPin G3 after nine months. These methods facilitate any relevant formulation practices for these compounds to meet any demand-oriented needs.
Collapse
Affiliation(s)
- Maryam Ehsasatvatan
- Department of Plant Breeding & Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, 51666, Iran
| | - Bahram Baghban Kohnehrouz
- Department of Plant Breeding & Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, 51666, Iran.
| |
Collapse
|
49
|
Campuzano IDG. A Research Journey: Over a Decade of Denaturing and Native-MS Analyses of Hydrophobic and Membrane Proteins in Amgen Therapeutic Discovery. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:2413-2431. [PMID: 37643331 DOI: 10.1021/jasms.3c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Membrane proteins and associated complexes currently comprise the majority of therapeutic targets and remain among the most challenging classes of proteins for analytical characterization. Through long-term strategic collaborations forged between industrial and academic research groups, there has been tremendous progress in advancing membrane protein mass spectrometry (MS) analytical methods and their concomitant application to Amgen therapeutic project progression. Herein, I will describe a detailed and personal account of how electrospray ionization (ESI) native mass spectrometry (nMS), ion mobility-MS (IM-MS), reversed phase liquid chromatographic mass spectrometry (RPLC-MS), high-throughput solid phase extraction mass spectrometry, and matrix-assisted laser desorption ionization mass spectrometry methods were developed, optimized, and validated within Amgen Research, and importantly, how these analytical methods were applied for membrane and hydrophobic protein analyses and ultimately therapeutic project support and progression. Additionally, I will discuss all the highly important and productive collaborative efforts, both internal Amgen and external academic, which were key in generating the samples, methods, and associated data described herein. I will also describe some early and previously unpublished nano-ESI (nESI) native-MS data from Amgen Research and the highly productive University of California Los Angeles (UCLA) collaboration. I will also present previously unpublished examples of real-life Amgen biotherapeutic membrane protein projects that were supported by all the MS (and IM) analytical techniques described herein. I will start by describing the initial nESI nMS experiments performed at Amgen in 2011 on empty nanodisc molecules, using a quadrupole time-of-flight MS, and how these experiments progressed on to the 15 Tesla Fourier transform ion cyclotron resonance MS at UCLA. Then described are monomeric and multimeric membrane protein data acquired in both nESI nMS and tandem-MS modes, using multiple methods of ion activation, resulting in dramatic spectral simplification. Also described is how we investigated the far less established and less published subject, that is denaturing RPLC-MS analysis of membrane proteins, and how we developed a highly robust and reproducible RPLC-MS method capable of effective separation of membrane proteins differing in only the presence or absence of an N-terminal post translational modification. Also described is the evolution of the aforementioned RPLC-MS method into a high-throughput solid phase extraction MS method. Finally, I will give my opinion on key developments and how the area of nMS of membrane proteins needs to evolve to a state where it can be applied within the biopharmaceutical research environment for routine therapeutic project support.
Collapse
Affiliation(s)
- Iain D G Campuzano
- Amgen Research, Center for Research Acceleration by Digital Innovation, Molecular Analytics, Thousand Oaks, California 91320, United States
| |
Collapse
|
50
|
Rasheed MW, Abiodun AE, Eziagu UB, Idowu NA, Kabiru A, Adegboye TA, Oluogun WA, Ayoade AA. Clinicopathological and immunohistochemical characterization of gastrointestinal stromal tumour at four tertiary health centers in Nigeria using CD117, DOG1, and human epidermal growth factor receptor-2 biomarkers. Ann Afr Med 2023; 22:501-507. [PMID: 38358152 PMCID: PMC10775934 DOI: 10.4103/aam.aam_180_22] [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: 12/22/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 02/16/2024] Open
Abstract
Aims Gastrointestinal stromal tumors (GISTs) are neoplastic lesions that primarily affect the digestive tract and develop from interstitial cells of Cajal. These lesions require histopathological and immunohistochemical characterization due to their malignant potential and personalized treatment. In this investigation, the sex, age, lesional sites of origin, histopathological types, the prevalence of human epidermal growth factor receptors (HER-2) expression, prognostic indices (based on tumor size and mitotic figures), expression of CD117 and DOG1, and characteristics of patients with GIST were all characterized. Materials and Methods This was a retrospective cross-sectional analysis of GIST cases seen at four tertiary health-care centers in Nigeria over a 10-year period (2008-2017) and investigated utilizing histopathological and immunohistochemical (CD117, DOG1, and HER-2) methods. Results In this investigation, there were twenty GIST cases. Notably, the majority (40%) of the cases had tumors with sizes between 7.0 and 8.0 cm; the stomach was the most frequent site (70%) and the spindle cell type of GIST was the most prevalent (80%) histopathological type. In addition, the stomach was significantly associated with GIST as an origin site (with a P = 0.001), and 100% and 50% of these tumors were immunoreactive with CD117 and DOG1, respectively. Conclusions In our study, GISTs most frequently develop in the stomach, and CD117 and DOG1 are essential for correctly diagnosing these tumors. However, HER-2 immunoreactivity is a predictive marker of survival for personalized care.
Collapse
Affiliation(s)
- Mumini Wemimo Rasheed
- Department of Anatomic Pathology, University of Ilorin Teaching Hospital, Ilorin, Kwara State, Nigeria
| | - Afolayan Enoch Abiodun
- Department of Anatomic Pathology, University of Ilorin Teaching Hospital, Ilorin, Kwara State, Nigeria
| | | | - Najeem Adedamola Idowu
- Department of Surgery, Ladoke Akintola University of Technology, Ogbomosho, Oyo, Nigeria
| | - Abdullahi Kabiru
- Department of Histopathology, Usmanu Danfodiyo University Teaching Hospital, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Taiwo Adeyemi Adegboye
- Department of Epidemiology and Community Health, University of Teaching Hospital, Ilorin, Kwara State, Nigeria
| | - Waheed Akanni Oluogun
- Department of Histopathology, Ladoke Akintola University of Technology, Osogho, Nigeria
| | - Adekunle Adebayo Ayoade
- Department of Morbid Anatomy and Histopathology, Ladoke Akintola University of Technology, Ogbomosho, Oyo, Nigeria
| |
Collapse
|